Skip to main content

SYSTEMATIC REVIEW article

Front. Physiol., 07 October 2021
Sec. Integrative Physiology
This article is part of the Research Topic Functional Profile of the Lipocalin Protein Family View all 12 articles

The Lipocalin Apolipoprotein D Functional Portrait: A Systematic Review

  • Instituto de Biologia y Genetica Molecular, Unidad de Excelencia, Universidad de Valladolid-Consejo Superior de Investigaciones Cientificas, Valladolid, Spain

Apolipoprotein D is a chordate gene early originated in the Lipocalin protein family. Among other features, regulation of its expression in a wide variety of disease conditions in humans, as apparently unrelated as neurodegeneration or breast cancer, have called for attention on this gene. Also, its presence in different tissues, from blood to brain, and different subcellular locations, from HDL lipoparticles to the interior of lysosomes or the surface of extracellular vesicles, poses an interesting challenge in deciphering its physiological function: Is ApoD a moonlighting protein, serving different roles in different cellular compartments, tissues, or organisms? Or does it have a unique biochemical mechanism of action that accounts for such apparently diverse roles in different physiological situations? To answer these questions, we have performed a systematic review of all primary publications where ApoD properties have been investigated in chordates. We conclude that ApoD ligand binding in the Lipocalin pocket, combined with an antioxidant activity performed at the rim of the pocket are properties sufficient to explain ApoD association with different lipid-based structures, where its physiological function is better described as lipid-management than by long-range lipid-transport. Controlling the redox state of these lipid structures in particular subcellular locations or extracellular structures, ApoD is able to modulate an enormous array of apparently diverse processes in the organism, both in health and disease. The new picture emerging from these data should help to put the physiological role of ApoD in new contexts and to inspire well-focused future research.

Introduction

ApoD, identified and named almost 50 years ago, is a protein belonging to the Lipocalin family. Experimental research on ApoD has been accumulating, encouraged by numerous findings of ApoD relationship to many human diseases, from cancer to cardiovascular, metabolic or neurodegenerative conditions. This affluence of scientific reports has described many aspects of ApoD functional features, but a fundamental question remains to be responded: does ApoD moonlight, performing different biochemical functions in different biological contexts? or does it display a distinctive biochemical role that is being used in several physiological systems?

Along this half-a-century of ApoD research many reviews focused on this protein have been published. All of them are narrative in nature and many concentrate on specific details of ApoD such as its relationship to disease, often underlining partial and not critically assessed views on many aspects of ApoD biology.

Aiming at answering the central questions posed above, we have performed a systematic review of all primary research published until January 2021 where ApoD properties have been investigated in the chordate phylum. Reports were tagged and classified according to their contributions to “molecular properties,” “gene data,” “regulation of expression,” “disease-related,” “cellular trafficking,” “tissue and organ function,” and “protein physiology.” The level and quality of experimental evidence were critically evaluated to try to identify cause-effect relationships. The picture emerging from this approach should help to understand the physiological role of ApoD and to inspire well-focused future research.

Methods

To assess the current knowledge on the physiology of ApoD, we performed a literature review of primary publications in a systematic manner, searching the National Library of Medicine database with the PubMed engine (published until January 31, 2021). Using the search query “Apolipoprotein D” OR Apo-D OR ApoD, we recovered 851 entries from which 39 narrative reviews were excluded. Following a Title/Keywords screen, we selected 787 entries for further assessment. Following exclusion criteria we finally selected 417 articles reporting research on the Lipocalin ApoD in chordates as the final review sources, and stored them in a Zotero (v5.0.88) collection. The complete collection is available in the Supplementary Tables 18. According to the abstract information, articles were tagged with the following terms: Disease-related (DR, n = 216), Regulation of Expression (RE, n = 186), Gene Data (GD, n = 37), Molecular Properties (MP, n = 59), Cellular Trafficking (CT, n = 33), Tissue and Organ Function (TOF, n = 36), and Protein Physiology (PP, n = 35). Figure 1 summarizes the review process workflow and outcome.

FIGURE 1
www.frontiersin.org

Figure 1. ApoD literature search and inclusion criteria. (A) Yearly timeline of articles recovered by PubMed using the search query designed for our review. (B) PRISMA flow diagram for record inclusion in our review.

Following full article reading, we classified each tagged report with subheading terms to guide the organization of the review. We then performed an evaluation of the conclusions statements of each report based on technical and argumentative consistency, according to existing state-of-the-art standards and required experimental controls. In cases of uncertainty, experts in each field were contacted and asked for their objective judgement.

The following databases and in silico prediction platforms and tools were used in this work: ProtParam (https://web.expasy.org/protparam/); DeepLoc-1.0 (http://www.cbs.dtu.dk/services/DeepLoc/); Gene Ontology database (http://geneontology.org/); Human Protein Atlas (https://www.proteinatlas.org); Mouse gene expression (http://www.informatics.jax.org/expression.shtml); miRNA database (mirdb.org). The ApoD multiple sequence alignment was generated with ClustalX2 (http://www.clustal.org), and the 3D structure of ApoD was visualized with ViewerLite 4.2 (https://chemweb.ir/accelrys-viewerlite/). A model of HApoD with sugars attached was built with GlyProt (http://glycosciences.de/modeling/glyprot/php/main.php).

Results and Discussion

ApoD is an early-diverging member of the Lipocalin family, with its phylogenetic origins traced back to the origin of chordates (Ganfornina et al., 2000; Diez-Hermano et al., 2021). Furthermore, ApoD is the chordate Lipocalin most similar to those in other phyla. ApoD primary structure is well-conserved in chordates, as deduced from a multiple sequence alignment of 22 chordate species (Figure 2A; Table 1), with an average 67% identity (range: 55–90%) in mature protein sequence. An intriguing aspect of this alignment is a favored residue conservation of the region encompassing the first three β-strands of the protein primary structure (Figure 2A).

FIGURE 2
www.frontiersin.org

Figure 2. ApoD protein sequence and gene features. (A) Multiple sequence analysis (MSA) of the mature amino acid sequence of selected vertebrate species recovered from GenBank (Apla, Anas platyrhynchos_EOB05196.1; Caur, Cathartes aura_KFP53002.1; Ggal, Gallus gallus_NP001011692.1; Pcri, Pelecanus crispus_KFQ60274.1; Pcrs, Podiceps cristatus_KFZ69168.1; Prub, Phoenicopterus ruber_KFQ85568.1; Bmut, Bos mutus_ELR54927.1; Cela, Cervus elaphus_ABB77207.1; Chir, Capra hircus_XP005675150.1; Oari, Ovis aries_XP004003075.1; Sscr, Sus scrofa_XP001926098.2; Fcat, Felis catus_XP006936237.1; Umar, Ursus marinus_XP008706566.1; Oorc, Orcinus orca_XP004278821.1; Mbra, Myotis brandtii_EPQ12038.1; Pale, Pteropus Alecto_XP006906222.1; Ocun, Oryctolagus cuniculus_ NP001075727.1; Hsap, Hoo sapiens_ NP001638.1; Ptro, Pan troglodites_XP516965.1; Fdam, Fukomys damarensis_KFO33128.1; Mmus, Mus musculus_CAA57974.1; Rnor, Rattus norvegicus_NP036909.1). Asterisks represent identical residues in all sequences, and dots/double dots point to similar residues. α-helices and β-strands are shown on top of the MSA, based on the solved tertiary structure of human ApoD. Colored residues are: four conserved cysteines involved in intramolecular disulfide bonds (pink), the human-specific unpaired cysteine (purple), conserved tryptophan residue in the ligand binding pocket (green), two glycosylated Asn residues (yellow), the antioxidant Met residue (blue), and residues in the hydrophobic surface patches at the rim of the binding pocket (orange). (B) Schematic representation of the chromosomal location of ApoD gene in human, mouse and chicken genomes. (C) Schematic representation of a consensus gene architecture for chordate ApoD, with four coding sequence (CDS)-containing exons and several 5′-UTR exons.

TABLE 1
www.frontiersin.org

Table 1. ApoD protein parameters.

Molecular Properties

ApoD is a monodomain globular glycoprotein with two intramolecular disulfide bonds, which are molecular properties suitable for working in extracellular non-reducing milieus. ApoD shows an N-terminal signal peptide in all chordates that lets the nascent protein to enter the endoplasmic reticulum. The protein can therefore follow a canonical secretion pathway, and is glycosylated along this path.

Protein Parameters

Since early characterization studies of ApoD, its apparent electrophoretic mobility, density of ApoD-positive fractions and behavior in size exclusion chromatography, suggested the existence of post-translational modifications (glycosylation), a potential for oligomerization, and an association with lipids. The predicted acidic isoelectric point (Table 1) implicates that ApoD polypeptide would have its lowest solubility in aqueous-salt solutions at the pH of acidic organelles in the cell, while at neutral pH the ApoD polypeptide would show a net negative charge. ApoD displays four conserved cysteine residues, while an additional cysteine (Cys116) is present only in humans (absent even in other primates) and allows for inter-molecular disulfide bond formation (Figures 2A, 3B,E; Table 1).

FIGURE 3
www.frontiersin.org

Figure 3. Molecular features of ApoD. (A,B) Graycolored space-filled views of the human ApoD tertiary structure (modelled from PDB ID:2HZQ) showing charged residues in A (positive, red; negative, blue) and hydrophobic residues in B (orange). Side view of the β-barrel (left image; curved arrows point to the pocket entrance) and top view (right image) looking into the hydrophobic pocket (asterisk). (C–E) Human ApoD (PDB ID:2HZQ) side and top views with highlighted relevant residues. Colored residues in (C) are the antioxidant Met93 (blue); the human-specific unpaired Cys116 (purple); the conserved ligand binding pocket Trp127 (green); and the two glycosylated Asn45/Asn78 (yellow). Pink-colored residues in D are the four cysteines forming two intramolecular disulfide bonds. Orange-colored residues in E are those forming three hydrophobic loops around the pocket entrance. (F) Space-filled view of human ApoD with reported oligosaccharides linked to Asn45 and Asn78, as modelled by GlyProt (see Methods). (G) Cartoon representations of human ApoD dimers formed by hydrophobic patches (orange) or by intermolecular Cys116 disulfide bonds (pink). Variations of the particular configuration shown are possible. Dashed lines delineate the ligand pocket. (H) Representation of the best supported tetrameric structure of human ApoD found in BCF. Asterisks mark the ligand pocket accessible in all subunits (two facing back). Oligosaccharides shown in red. (I) Cartoon illustration of a side view of human ApoD with AA (red) and HpETE (blue) positioned into the hydrophobic pocket (marked by a dashed line) and interacting with the Met93-containing hydrophobic patch respectively. (J,K) Cartoon illustration of human ApoD interacting with higher-order lipid structures via the hydrophobic patches at rim of the pocket; (J) HDL particle; (K) Unilamellar vesicle (liposome).

References contributing to this section are listed in Reference Collection 1, Supplementary Table 2.

Protein Structure

The ApoD 3D crystal structure has been solved for the human protein after modification of several residues that rendered the protein prone to aggregation. The unique human Cys116 is close to one of the hydrophobic loops, and was also mutated to facilitate crystallization. The structure reveals a typical Lipocalin fold (Skerra, 2000) composed of an eight-stranded β-barrel structure with an adjacent C-terminal α-helix. It has a closed end, and an open end with access to a pocket able to bind mostly hydrophobic ligands. Two intra-molecular disulfide bonds stabilize the structure. Three out of four loops at the barrel open-end are hydrophobic, making these regions candidate for interaction with hydrophobic surfaces, and contain residues relevant for ApoD antioxidant properties (see section Protein Physiology). Two N-glycosylation sites (Figures 2A, 3F) are located on the side and bottom of the calyx, away from the ligand-binding pocket opening. Figures 3A,B show a surface representation of the ApoD monomer structure with charged or hydrophobic surface highlighted in color. Other relevant residues are shown in Figures 3C–E.

The presence of a ligand inside the pocket did not modify the general crystal structure of ApoD. When explored by amide hydrogen-deuterium exchange mass spectrometry (HDX-MS) or small-angle X-ray scattering (SAXS) in solution, interesting conformational changes elicited by ligand binding were detected, resulting in further ordering of the already stable Lipocalin fold. ApoD structure is also stable upon protein oxidation with H2O2. Dynamic information extrapolated from the crystal structure has allowed further modeling of ApoD binding to small ligands, lipoprotein particles or membranes. These studies help to understand a methionine-dependent lipid antioxidant mechanism (see below) and to study the influence of glycosylation on these functional properties. In addition, the ApoD monomer crystal structure, combined with modeled glycosylation conformations, was used to generate coherent models for the conformations of ApoD oligomers (Figures 3G,H) later confirmed experimentally (see below).

References contributing to this section are listed in Reference Collection 2, Supplementary Table 2.

Protein Glycosylation

As mentioned above, sugars were soon revealed to be linked to ApoD, with a relevant carbohydrate contribution (~15–22%) to its apparent molecular weight. Two asparagine sites were experimentally demonstrated to be glycosylated, and in silico studies of human ApoD revealed no interference of sugars with binding pocket access. Figure 3F depicts a model of the N-linked oligosaccharides. The Asn45 glycosylation site is conserved in birds and mammals, but the second glycosylation site shows variations in position (Figure 2A). In ApoD of human plasma, Asn45 contains primarily trisialo-triantennary oligosaccharides, and Asn78 contains fucosylated disialo-biantennary oligosaccharides. The presence of negatively charged sialic acid in native ApoD sugar moiety contributes to its net negative charge in neutral pH environments.

Interesting variations of ApoD carbohydrate moiety have been reported between species (e.g., humans vs. other primates or rodents), between various tissues in a single species (brain tissue, cerebrospinal fluid, inner ear perilymph or plasma), within a tissue, or between health and disease conditions. Patterns of glycosylation have also been demonstrated to be sex-dependent (e.g., BCF in females, or axillary secretion in males). For example, less glycosylated forms of ApoD are present in mouse/human brain tissue compared to plasma, with differences in both terminal sialic acid and core N-linked oligosaccharides. A clear increase in α2-3 sialoglycosylation of plasma ApoD distinguishes, with high sensitivity, children with Autism Spectrum Disorder from healthy controls. Within a single tissue (cerebrospinal fluid; CSF) there is also variegation in the degree of ApoD sialylation. These variations generate size and charge heterogeneities with potential functional consequences worth exploring.

References contributing to this section are listed in Reference Collection 3, Supplementary Table 2.

Protein Oligomerization

Covalent and non-covalent homodimers and tetramers of ApoD have been detected in various experimental and biological systems. All studies of ApoD oligomerization have been focused so far on the human protein. Crystallization of bacterial recombinant human ApoD revealed that the protein tends to aggregate due to hydrophobic surface patches. This property could promote self-association or association with lipid-based structures in vivo (Figures 3J,K).

Homodimers due to intermolecular disulfide bonds, evidenced by comparing electrophoretic mobility under reducing/non-reducing conditions, have been detected in urine and tear fluids. Experiments with sulfhydryl-trapping reagents during handling indicate that new disulfide bonds were not introduced along the experimental procedure. However, other forms of oligomerization are possible and compatible with these results, resulting from non-covalent stable binding between ApoD monomers. Figures 3G,H summarize the different forms of ApoD self-interaction.

Exposure of human ApoD to oxidized lipids promote dimerization and further oligomerization, in a way dependent on the oxidation state of particular methionine residues (Met93, see below), as demonstrated with recombinant ApoD, mutated at specific Met residues, and produced in a human cell line. Oxidation-dependent ApoD dimerization is resistant to guanidine hydrochloride (GuHCl) but not to urea, indicating that it is based on non-covalent intermolecular bonds. This property has allowed to detect ApoD dimers in specific brain regions of Alzheimer's disease patients (e.g., hippocampus, but not cerebellum) that also correlate with disease progression. In contrast, GuHCl extracts from healthy control brains show only monomeric forms of ApoD.

A tetrameric stable form of native ApoD in BCF, but not in plasma or CSF, has been demonstrated and characterized by multi-angle laser light scattering, analytical ultracentrifugation, HDX-MS and SAXS. Experimental data using progesterone as a ligand and the native ~100 kDa ApoD tetramer from BCF, supports a particular tetramer conformation among those predicted by molecular modeling, where the binding pocket opening is accessible and the sugar moieties do not interfere in the inter-subunit interface (Figure 3H). Monomers interact with each other through the C-terminal α-helix and three β-sheets in close proximity, while glycosylated surfaces and Met93 are exposed in the tetramer. Oligomerization does not preclude ligand binding, and is not significantly altered upon binding of various ligands (biliverdin, palmitic acid, progesterone and sphingomyelin) or by in vitro protein oxidation with H2O2. The putative contribution of intermolecular disulfide bonds in the tetramer (involving human Cys116) has not been explored.

References contributing to this section are listed in Reference Collection 4, Supplementary Table 2.

Small Ligand Binding

The ability to bind progesterone was a defining feature of the most abundant protein in BCF, therefore named progesterone-binding cyst protein (PBCP) or gross cystic disease fluid protein 24 (GCDFP-24). Later on, this protein was demonstrated to be identical to ApoD purified from plasma HDL particles. Ligand-protein interaction at the ApoD binding pocket induces conformational changes leading to a more ordered structure, but does not result in major structural changes or altered oligomerization. These dynamic changes, though subtle, might have implications for ApoD interactions with other proteins or lipoprotein particles. Ligand binding reports are grouped in Reference Collections 5, 6, Supplementary Table 2.

Progesterone accommodation in the pocket involves a tryptophan residue heavily conserved in the Lipocalin family (Trp127 in human ApoD; Figures 2A, 3C), whose fluorescence (Ex. λ = 295 nm) changes upon binding. This element in the pocket makes Trp-fluorescence titration a valid method to test a variety of ligands for ApoD (Table 2). All in vitro ligand-binding experiments have been performed with the human protein, using either recombinant ApoD (expressed by bacteria or eukaryotic cells) or native protein purified from BCF or plasma HDL. Arachidonic acid (AA) shows the highest affinity, while various AA derivatives (e.g., prostaglandins, 12-HETE or 5,15-diHETE) show no binding by Trp-fluorescence titration.

TABLE 2
www.frontiersin.org

Table 2. ApoD ligand binding in vitro assays.

Cholesterol, a reasonable candidate because of its high presence in plasma lipoprotein particles, has been repeatedly tested, and reported to have no binding, or a very low affinity one (Table 2). A series of works (Reference Collection 6, Supplementary Table 2) demonstrate that ApoD has no cholesterol-transfer activity, a hypothesis originated by ApoD co-purification with lecithin-cholesterol acyltransferase (LCAT), whose activity is in fact modulated by ApoD (see section Protein Physiology) by a mechanism discarding ApoD as a cholesterol provider for LCAT.

Only one ligand has been identified bound to ApoD and extracted from the protein after purification from a natural source. E-3-methyl-2-hexenoic acid (E-3M2H), a male axillary precursor of odorants, was identified by gas chromatography-mass spectrometry (GC/MS) after temperature/pH switch and chloroform extraction from purified ApoD.

Interestingly, various ligands (e.g., bilirubin or E-3M2H) whose interaction with ApoD has been demonstrated by a different technique, do not alter Trp-fluorescence, raising the possibility of other sites of interaction. Molecular dynamics simulations infer flexible binding of oxidized derivatives of AA (5s-, 12s-, and 15s-HpETE) around the conserved Met93 at one of the hydrophobic patches at the entrance of the pocket. This particular form of lipoperoxide binding to ApoD is not expected to produce changes in fluorescence of Trp-127, located at the bottom of the binding pocket. A proof of interaction is experimentally supported by site-directed mutagenesis combined with HPLC-detection of reduced lipids (HETEs) after exposure to ApoD. This interaction underlies the antioxidant activity of ApoD (see below). Figure 3I summarizes in cartoon form this new view of small ligand-binding sites of ApoD, not restricted to the Lipocalin pocket.

Protein-Protein Interactions

Interactions of ApoD to higher-order lipid structures, like lipoprotein particles or cellular membranes, are particularly relevant since they determine the range of sites and biological contexts where ApoD function can be performed. They might depend on protein-protein or protein-lipid contacts.

As mentioned above, co-purification of ApoD with LCAT might indicate the potential for a protein-protein interaction in nascent HDL particles, but a clear demonstration of ApoD-LCAT complex is not available. In contrast, a clear protein-protein interaction does account for human ApoD presence in HDL particles. An intermolecular disulfide link between ApoD Cys116 and ApoA-II Cys6 has been demonstrated by peptide digestion followed by sequencing and mass spectrometry (MS). This interaction, however, is an exclusive property of human ApoD due to its unique unpaired cysteine.

A putative disulfide-linked ApoD-ApoB100 complex was also proposed, but evidence is based on predictions from electrophoretic mobility in reducing/non-reducing conditions and immunoblot detection with anti-ApoD antibodies only, or with antibodies raised against LDL particles. An almost full characterization of 23 out of 25 cysteine residues in ApoB-100 by MS-analysis and peptide sequencing found no bonds with ApoD (Yang et al., 1990), strongly arguing against a disulfide-mediated interaction. Alternative mechanisms of ApoD interactions with plasma lipoprotein particles are therefore open to consideration.

Other potential interactions of ApoD have been explored with classic two-hybrid systems, where protein-protein contact takes place in the cell nuclei or cytoplasm, both requiring ectopic expression of ApoD in non-native biological compartments unsuitable for disulfide linked proteins (see sections Protein Structure and Cellular Trafficking). Alternatively, co-immunoprecipitation in vitro with or without crosslinking agents has been a method of choice. Using these approaches, ApoD has been proposed to interact with the extracellular glycoprotein Osteopontin (OPN), the intracellular domain of the Leptin Receptor (OB-Rb), the transmembrane glycoprotein Basigin (BSG), and the Scavenger receptor class B type 1 (SRB1).

The weak interaction reported between ApoD and the intracellular domain of OB-Rb, combined with its presumed topology within the cell, should discard this finding as a biologically relevant interaction for ApoD unless it is replicated. For membrane proteins such as BSG and SRB1, proposed as putative membrane receptors for ApoD, co-localization by confocal imaging is often used as additional evidence. However, protein-complexes are below the resolution of standard co-labeling techniques, and methods relying on distance-dependent energy transfer, super-resolution or immunoelectron microscopy would be desirable as further evidence in relevant in vivo conditions. Other candidate ApoD receptors (LDLR and CXCR-4) are predicted from physiological contexts, where downstream consequences of ApoD exposure are modified by antagonists of these receptors. However, a direct interaction with these receptors has not been explored.

References contributing to this section are listed in Reference Collection 7, Supplementary Table 2.

Binding to Lipid-Rich Structures

The presence of ApoD in plasma lipoprotein particles lies at the base of its discovery in humans. ApoD was initially visualized as a “thin-line” polypeptide in immune-double diffusion analyses of plasma HDL particles, and was then identified as a low-abundance component of HDL3 particles (defined as small-dense HDLs, d = 1.12–1.27 g/ml). Analysis of HDLs separated by electrophoretic mobility in non-denaturing PAGE followed by in-gel trypsinization, identified ApoD within the HDL-α2 type, in a 1:100 ratio with respect to ApoA-I. The presence of ApoD in HDLs has been confirmed also in human CSF and in baboon and mouse plasma. Additionally, plasma ApoB-100 positive LDL particles contain ApoD as well, but at lower concentrations (~8 ng ApoD/μg LDL vs. ~69 ng ApoD/μg HDL3). The generalized interaction with different lipoparticles in several species suggests that ApoD-lipoparticle interactions must rely on a mechanism independent of ApoD-ApoA-II disulfide bond, a human HDL rarity. The fact that ApoD-LDL interaction is prevented by detergents, and do not take place with recombinant ApoD where hydrophobic surface residues have been mutated (to favor crystallization), suggests a hydrophobicity-dependent ApoD-lipoparticle binding mechanism (Figure 3J).

Also, direct binding of ApoD to unilamellar phospholipid vesicles (liposomes) further demonstrates its ability to bind to lipidic structures without requiring a protein-protein interaction. These unilamellar vesicles represent a simplified version of the outer phospholipid layer of HDLs, LDLs or a membrane bilayer (Figure 3K). In addition, ApoD has recently been identified in extracellular vesicles, characterized by the presence of CD81, CD63, and flotillin-1, and a density of d = 1.17–1.23 g/ml. The hydrophobic patches of ApoD at the entrance of the binding pocket are the likely site of interaction with liposomes or biological membranes, as indicated by experiments combining ApoD capacity to reduce oxidized liposomes with mutagenesis of Met residues that in fact contribute to the hydrophobicity of those patches.

The knowledge accrued on ApoD protein structure, its glycosylation and oligomerization properties, as well as its interactions with small ligands and other lipidic structures are relevant for its physiological roles in lipid management, and should help to get a global picture of how these molecular properties are put to work in various physiological contexts.

References contributing to this section are listed in Reference Collection 8, Supplementary Table 2.

Gene Data and Genomic Properties

Chromosomal Position and Gene Structure

The gene coding for ApoD locates in an autosomic chromosome that shows ample synteny in chordates (Sanchez et al., 2006), reflecting a strong evolutionary conservation of this genomic region (Figure 2B). The ApoD gene shows a standard metazoan exon-intron architecture, with a coding sequence interspersed in four exons that is conserved in chordates (Sánchez et al., 2003). Moreover, the gene upstream and downstream untranslated regions (UTRs) are also composed of several exons, mainly in the 5′-UTR, a property well-preserved in mammals (Mejias et al., 2019) (Figure 2C).

References contributing to this section are listed in Reference Collection 9, Supplementary Table 3.

Transcriptional Control of Gene Expression

The promoter region and elements controlling the expression of ApoD have being studied in detail for the human gene. The human promoter shows a canonic TATA-box upstream of the transcription start site. Several promoter elements and nuclear factors have been predicted to potentially regulate ApoD transcription in a number of organisms.

Experimental proof of a regulatory potential of human ApoD has been gathered for SRE1, AP-1, APR-3, NFκB, PARP1, HnRNP-U, and APEX-1 in cultured cells subjected to inflammation (LPS) and metabolic stress (serum deprivation). Also, the transactivator TAp73 mediates ApoD expression upon cell differentiation. The mouse ApoD promoter region has been recently assessed experimentally, and an alternative promoter region has been related to OS-induced ApoD expression.

DNA methylation, inferred from the CpG content of the gene promoter region, is also an important regulatory mechanism for ApoD transcription, with an inverse relationship between level of DNA methylation and ApoD gene transcription. This gene regulation mechanism has been shown in different physiological or pathological contexts: in esophageal, colorectal and astrocytic cancers, in the expression profile defining Th17 lymphocytes, and for the androgen receptor-response in male sexual development.

References contributing to this section are listed in Reference Collection 10, Supplementary Table 3.

Post-transcriptional and Translational Control of Gene Expression

The mRNA 3′-UTR is known to influence its stability and translation efficiency. ApoD 3′-UTRs show a high degree of conservation in mammals, and display shorter lengths and higher G+C content than those observed in average mammalian gene UTRs. These differences have been proposed to underlie a tight regulatory control of ApoD translation. In this context, a number of miRNAs have been predicted to control ApoD translation, possibly by binding to the 3′-UTR. Some of these miRNAs, like miR-229b-3p, miR-423-3p, and miR-490-3p, have been experimentally tested and implicated in the post-transcriptional downregulation of ApoD expression in rat male reproductive system upon metabolic dysfunction.

The 5′-UTR of ApoD also presents relevant properties for the regulation of ApoD expression. It is rich in short-tandem repeats (STR), specifically in primates. Long stretches of STRs are predicted to affect transcription and translation, which might have contributed to the neurodevelopmental changes that underlie primate evolution. Furthermore, mammalian ApoD genes show several alternative 5′-UTRs forms, possibly arising from alternative splicing. The alternative 5′-UTRs of the mouse ApoD gene have been experimentally tested and shown to underlie differential protein expression in several mouse tissues, with a particular 5′-UTR variant being strongly induced upon OS. Moreover, in silico analyses of these 5′-UTR variants in mouse and human ApoD show upstream initiation codons, upstream open reading frames, and predicted secondary structures that suggest a tight control on ApoD gene expression.

References contributing to this section are listed in Reference Collection 11, Supplementary Table 3.

Gene Polymorphisms

In terms of genetic variation for the ApoD gene, over 4,600 variants have been found in the GRCH38.p12 (annotation Release 109) assembly of the human genome, while 187 are reported in the short variants (dbSNP) and structural variants (dbVar) databases. Six variants that involve missense, intron insertions and 3′-UTR insertions, are predicted to involve molecular consequences. Some of these variants have been linked with variable support to human cancer, metabolic or neurological diseases (see Supplementary Table 18, and section ApoD-Disease Relationships), but a final proof of their clinical significance is currently missing.

References contributing to this section are listed in Reference Collection 12, Supplementary Table 3.

Regulation of Expression

A total of 186 primary publications (Figure 4A) were labeled with the regulation of expression (RE) tag for this systematic review (details recorded in Supplementary Tables 917). We combined our analysis with current data compiled in human and mouse expression atlases (see Methods section; Supplementary Figures 1, 2).

FIGURE 4
www.frontiersin.org

Figure 4. Publications on ApoD expression and disease relationships. (A) Distribution of publications describing ApoD mRNA expression or protein presence in vivo, distributed by physiological systems and in cell cultures (primary cells or cell lines). (B) Publications with information on ApoD relationship to disease (expression changes triggered by disease or treatments, or association of ApoD gene variants with disease).

ApoD in Body Fluids

Since its discovery in plasma HDL particles, ApoD protein and/or mRNA have been found in almost every organ, tissue or fluid. In addition to plasma, ApoD protein is present CSF, perilymph, urine, and secretions from exocrine glands (sweat, tears and mammary secretions) (Supplementary Table 9). The cellular origin of ApoD protein in each of these body fluids is not fully elucidated. With the exception of Th17 lymphocytes, blood cells in general do not express ApoD mRNA, and liver and intestine (major sites of HDL biogenesis) are among the ApoD low-expressing tissues both in humans and mice (Supplementary Figures 1, 2). Plasma ApoD protein (~128 mg/l) is approximately 25 times the concentration of CSF ApoD (~5 mg/l) in healthy adult men, and they are uncorrelated, suggesting that a separate pool of ApoD protein is managed in these barrier-separated compartments.

Avian egg fluids are also rich in ApoD, with the interesting property that egg white ApoD positively correlates with egg freshness.

References contributing to this section are listed in Reference Collection 13, Supplementary Table 4.

Tissue and Cellular Expression Patterns and Response to Stimuli

The analysis of tissue expression pattern leads to a general conclusion: in spite of its wide distribution, ApoD is never ubiquitously expressed, never in all cell types in a tissue, or at all times in a given cell type. ApoD is expressed in most tissues with a salt-and-pepper spatiotemporal pattern, suggesting a fine control that depends on particular physiological cell states. Furthermore, all tissues bear ApoD-expressing cells and cells able to endocytose ApoD protein from the extracellular milieu (see section Cellular Trafficking). These expression features, along with ApoD being a very stable protein, result in a high protein abundance when measured in high-throughput analyses, and in a lack of exact fit between mRNA and protein expression in a given tissue or cell (Supplementary Figure 1; Reference Collections 1416; Supplementary Table 4). While tissues as the female breast present high levels of ApoD mRNA and protein, organs like the liver show high abundance of ApoD protein, but barely detectable ApoD mRNA both in human and mice. At the other end of the spectrum, blood cells and immune system-related organs are among those with low levels or no expression of ApoD, either mRNA or protein.

Organs and tissues involved in both male and female reproductive physiology express ApoD (Supplementary Table 10). The high expression of ApoD in breast has been located to the glandular epithelium (Supplementary Figure 1), and breast cysts accumulate high amounts of ApoD protein, making BCF a useful experimental source of native ApoD protein. ApoD mRNA is detected at all stages of the spermatogenesis process in testis and in ovarian theca cells. Along the female cycle, stromal and epithelial cells of the endometrium express ApoD mRNA and protein during the secretory phase. ApoD is also expressed during corpus luteum maturation in the ovary. Gestation alters ApoD abundance in plasma as well (Supplementary Table 9), with a decrease during a healthy pregnancy followed by a fast recovery if the mother breastfeeds her baby. ApoD expression in breast secretions and skin is also altered upon establishment of menopause.

These temporal patterns of expression are due to hormone regulation, as demonstrated by both in vivo and in vitro studies (Supplementary Tables 10, 16, 17; Reference Collection 17). Upregulation of ApoD by androgens is well documented in different preparations like breast explants, male genital fibroblasts or primary epithelial cells from male axillary apocrine glands. This regulation is mediated by nuclear androgen receptor (AR), and ApoD is being used as an AR activity assay (Figure 5B). Estrogens and progesterone, alone or in combination, also change ApoD expression in several experimental settings, with more variation in the final outcome depending on cell type (e.g., breast cancer cell lines up-regulate ApoD upon exposure to 17β-estradiol, while prostate cancer cell lines down-regulate it, Supplementary Table 17). Sex hormone-regulation of ApoD is also present in birds, in the context of oviposition cycles or egg fertilization, thus representing relevant biological stimuli for ApoD spatiotemporal regulation throughout evolution (Figure 5, Supplementary Table 10).

FIGURE 5
www.frontiersin.org

Figure 5. Factors and pathways regulating the expression of ApoD. (A) Diverse stimuli regulate ApoD expression in a variety of cells and physiological conditions. (B) Summary of upstream regulatory pathways regulating ApoD expression where elements of the signaling cascade have been identified.

Expression in the nervous system (Supplementary Table 11) has been amply explored for ApoD, with primary publications doubling those devoted to other tissues or systems (Figure 4). All evidences support a prominent and consistent ApoD expression in the nervous system, where myelinating glial cells (oligodendrocytes and Schwann cells) constitute the main sites of expression in control conditions, followed by a more disseminated expression in astrocytes. ApoD protein abundance in the nervous system is accounted for by the fact that ApoD associates to myelin itself, a structure representing a large proportion of the vertebrate nervous system volume. As mentioned above, only subsets of cells express ApoD at a given time or location for each cell type (Reference Collection 16, Supplementary Table 4).

In addition to glial cells, ApoD has been found in meninges and the vascular system of the nervous system (Supplementary Table 11), particularly in pial and perivascular cells (mural cells or pericytes) associated to the capillary beds. During mouse embryogenesis, ApoD has been detected in both pericytes and endothelial cells, and mRNA expression in the latter is under the control of Wnt/β-catenin signaling during the time interval of blood-brain-barrier formation (Figure 5B). ApoD-positive pericytes and other perivascular cells are also reported in the adult nervous system. RNAseq analyses of acutely isolated cortical cells show endothelial cells as second to myelinating oligodendrocytes in ApoD enrichment.

Although neuronal expression has been subject to debate (Supplementary Table 11), unambiguous detection of ApoD mRNA in neurons has been reported only in the developing brain. In contrast, detection of ApoD protein in some neurons has been reported at various ages in healthy control situations, while this finding is more abundant upon aging or disease. Neuronal uptake of ApoD upon disease has a certain degree of specificity. It is frequently found in the brain of Alzheimer's, but not in Parkinson's disease patients. Animal models of brain traumatic injury, stroke and Niemann-Pick type A disease do show neurons that have internalized ApoD protein, while they are not found in the Niemann-Pick type C mouse model. Transfer of ApoD from astroglial cells to neurons has been demonstrated in cell culture preparations, and shown to be mediated by extracellular vesicles (Supplementary Table 17; see section Cellular Trafficking).

A well-established fact with strong support from different studies is that ApoD expression increases throughout brain aging (Reference Collection 18, Supplementary Tables 4, 11), a pattern conserved in several species analyzed with just one exception: a study documenting a decrease in ApoD mRNA in the aging avian hippocampus. A higher ApoD expression in cortex and brainstem in comparison with hippocampus or cerebellum are well-supported regional differences within the brain (Supplementary Table 11). In the highly-expressing prefrontal cortex, the increase of ApoD mRNA and protein throughout life positively correlates with proteins involved in antioxidant defense.

The expression data obtained from healthy individuals is coherent with an ApoD gene response to diverse experimental stress or injury paradigms (Reference Collections 1920, Supplementary Table 4) that include oxidative stress (OS), peripheral nerve or traumatic brain injury, kainate excitotoxicity, damage by middle cerebral artery occlusion or by viral infection and experimental inflammation. All of the above results in increased ApoD expression in vivo. This ApoD stress response is mostly, but not exclusively, documented in the nervous system (e.g., OS-triggered upregulation is also observed in the cardiovascular system). These patterns of response can be extended to the many disease situations reviewed in section ApoD-Disease Relationships. In addition to the abundant correlative data from human diseases, experiments in animal models of disease analyzed in vivo, primary cell cultures and cell lines support a major conclusion: ApoD is a key player in the endogenous response to a variety of potentially harmful stimuli. The damage and stress responsive p73/p63 and JNK pathways have been demonstrated to up-regulate ApoD (Supplementary Tables 16, 17 and Figure 5), while the particular signaling cascades regulating ApoD upon other stress or inflammation inducers (e.g., H2O2, UV light or LPS) remains to be elucidated. Not all stressful conditions trigger ApoD expression (Supplementary Table 17), underscoring the specificity of pathways regulating ApoD (Figure 5). Moreover, a fine regulation of ApoD upon OS seems necessary, since it involves various non-exclusive mechanisms like DNA demethylation, the use of alternative promoters or 5′-UTR specific mRNA variants (see section Gene Data and Genomic Properties).

Nutritional and metabolic states also regulate ApoD expression (Reference Collection 21, Supplementary Table 4), and ApoD upregulation under caloric restriction or ADCY5 loss-of-function seems to be part of a common signature leading to lifespan extension. Curiously, these results derived from in vivo studies agree with ApoD upregulation upon serum starvation in cell culture systems (Reference Collection 22, Supplementary Table 4). New studies on how metabolic switches can modulate ApoD in different contexts, and searching for the specific signaling pathways that trigger ApoD expression are therefore valuable. A particular lipid-managing pathway is known to control ApoD expression: ApoD is a target gene for LXR in liver, skeletal muscle, adipocytes and endothelial cells, thus becoming part of the response to oxysterol stimulation.

Pathways involved in development and cell differentiation are also known to regulate ApoD expression (Reference Collection 23; Supplementary Tables 4, 16, 17). In addition to its regulation by the Wnt/β-catenin pathway mentioned above, ApoD is downstream of Sox9 during chondrogenic differentiation, and of PACAP/Erk signaling during adipocyte differentiation. Also, particular cell-cell interactions regulate ApoD expression in one of the cellular partners, like endothelial-mural cell interactions relevant during the angiogenesis process. In this scenario, ApoD is downregulated in mural cells by contact-dependent (Notch-3) and contact-independent mechanisms.

Finally, confluency and senescence in cell cultures also trigger ApoD expression (Reference Collection 22, Supplementary Table 4). These culture conditions parallel steady-state situations of cells in their physiological tissue environment and the in vivo upregulation by aging, respectively. Both conditions concur with a halt in cell division, as it is also the case for serum starvation conditions. The good prognosis of some types of cancers where ApoD increases, also relates its expression to low cell-division rate (see section ApoD-Disease Relationships). Retinoic acid induction of ApoD expression, mediated specifically by RARα in breast cancer cells, correlates with the anti-proliferative action of this signaling pathway. However, the potential role of ApoD in regulating cell division (see section Protein Physiology) must be dependent on the physiological/pathological context. For example, in the model of pericyte-endothelium interactions mentioned above, mural cells decrease ApoD expression upon interaction with endothelial cells, when they would stop dividing to generate mature capillary structures.

Figure 5 summarizes stimuli regulating ApoD expression and the particular upstream signaling pathways known to date.

ApoD-Disease Relationships

The reports in this section either study the expression of ApoD in response to disease and therapies, or evaluate association of ApoD gene variants with disease. Information was accrued from a total of 216 primary publications (Figure 4B; details in Supplementary Table 18).

That ApoD is part to the endogenous response to a wide range of diseases, with diverse primary causes, is uncontentious. Data support the existence of common factors underlying diverse disease situations that cause ApoD expression changes, and OS is the strongest candidate. The ApoD upregulation upon experimental stress or injury reviewed above is coherent with prominent examples concurring in the nervous system (Reference Collection 27, Supplementary Table 5), where 85% of 66 reports on degenerative/psychiatric diseases or naturally occurring injury identify an over-expression of ApoD. Exceptions are the down-regulation observed in neurotransmission-centered diseases, like depression and a DOPA-decarboxylase deficiency.

Cancer is the other major disease where changes in ApoD expression have been analyzed (82 reports). A clear negative correlation between ApoD expression and malignancy has been found in nervous system tumors, fibrosarcomas, breast, colorectal, hepatic, renal and cervical cancers. The general association of a good prognosis with high ApoD expression strongly suggests a protective anti-tumoral function for this Lipocalin. While studies of prostate cancer have not evidenced unambiguously such a pattern, some studies show regional ApoD expression differences (high in juxta-tumoral tissue) that are still compatible with a defensive tissue response to neoplastic transformation. ApoD tumor-suppressing activity has been experimentally tested and an inverse relationship between ApoD promoter methylation, ApoD expression and outcome is supported by various reports (Supplementary Table 18). Whether a common mechanism of ApoD function can promote survival of damaged postmitotic cells in neurodegenerative diseases, and also prevents proliferation of cancerous cells deserves further analysis.

Cardiovascular and metabolic diseases (particularly diabetes) as well as infection or injury, are also accompanied by ApoD upregulation. Again, OS might be a common link to ApoD response to these diseases, for instance in atherosclerotic plaques depending on disease progression, or upon oxidative degradation of glycated proteins in diabetes.

In contrast to the many diseases where ApoD expression changes have been reported, few genetic variations of ApoD have been widely or robustly linked to disease risk or prognosis (see section Gene Polymorphisms and Supplementary Table 18). Among the few cases reported, it is striking that most of them occur in non-coding sequences (introns or UTRs) revealing that pathogenic variations in ApoD protein sequence must be too deleterious to survive in extant populations.

Cellular Trafficking

The consistent finding of ApoD in body fluids and the signal sequence present in the translated polypeptide indicate that ApoD is exocytosed from cells expressing the protein. A consistent set of experimental work supports the association of ApoD to the rough endoplasmic reticulum (ER), the signal peptide removal in the protein sorting process, the N-linked oligosaccharide modification carried out in RER-Golgi, and a secretion of the mature glycoprotein to the extracellular environment in several tissues and cultured cells. All these data make ApoD a typical soluble extracellular protein undergoing a canonical secretory pathway, a consensus attained by subcellular localization prediction algorithms and data present in gene ontology databases (see Methods). Additionally, subcellular traffic of ApoD can also lead to its exportation out of the cell in different formats that include ApoD tetramers, HDL-associated ApoD and extracellular vesicle-associated ApoD (Figure 6).

FIGURE 6
www.frontiersin.org

Figure 6. Schematic representation of ApoD subcellular traffic. A model of an ApoD-expressing cell is represented. Canonical exocytosis through the RER-Golgi pathway generates the mature, glycosylated (red dots) protein. The tetrameric form identified in the breast cyst fluid is represented as the format detected in extracellular fluids. Once at the plasma membrane, ApoD can be endocytosed (by non-expressing cells as well) and targeted to lysosomes and autophagolysosomes. When endolysosomes develop into multivesicular bodies, ApoD would be carried on the outer surface of exosomes. Finally, ApoD can be transferred to HDL during their biogenesis, or during their lipid-efflux activity (upon HDL-receptor interaction).

However, several reports have interpreted their findings about ApoD biological roles on the basis of protein partitioning in cytoplasm and/or nuclear compartments. Aside of technical issues questioning those results, some reports use overexpression of fusion-tagged proteins, which are known to undergo unnatural compartmentalization or degradation. Also, strategies based on in vitro interaction assays that were designed for proteins naturally occurring in the cytoplasmic or nuclear compartments (like the classic two-hybrid assays) preclude the finding of functionally relevant interactions for ApoD. An alleged cytosolic ApoD would likely be non-glycosylated and improperly folded in the absence of its intramolecular disulfide bonds.

Intracellular Traffic

A number of studies have shown the presence of ApoD in RER and vesicular compartments of different eukaryotic cells. Immunoelectron microscopy (EM) of nervous system cell types has unambiguously identified ApoD in the outer nuclear membrane-RER membrane complex and in lysosomes. Lysosomes isolated from placental cells, monitored with biochemical techniques, contain ApoD. Likewise, clathrin-coated vesicles isolated from hen's ovaries also contain ApoD.

Recently, the subcellular location of ApoD has been studied in detail in basal conditions and under experimental stimuli, either by immunogold-EM microscopy, or by fluorescence immunocytochemistry combined with established cell compartment markers and monitored by well-documented standardized confocal microscopy. These studies have detected the presence of ApoD in RER, the Golgi apparatus, endosomes, lysosomes, autophagosomes, multivesicular bodies, as well as in the outer side of plasma membrane, clathrin-coated vesicles and caveolae. The protein does not localize to mitochondria or peroxisomes, and has not been immunolocalized inside cell nuclei. Lysosomal ApoD localization has been demonstrated in astrocytes, oligodendrocytes, Schwann cells, fibroblasts, and neurons.

Although those experiments were performed at a fixed time point, serial-time experiments demonstrated that the presence of ApoD in the endosome-lysosome compartment is stable and dynamically enriched upon OS exposure. Long-lasting lysosomal location of ApoD depends on its glycosylation and hydrophobicity, as mutated bacterial recombinant ApoD is maintained in lysosomes only transiently. Targeting of ApoD to the lysosomal compartment occurs not only in ApoD-expressing cells, but also in non-expressing neurons upon exposure to exogenous ApoD or when co-cultured with astrocytes. The fact that ApoD is a stable component of subsets of lysosomes at a given time, connects many of the apparently diverse physiological roles of ApoD (see section Protein Physiology).

References contributing to this section are listed in Reference Collection 33, Supplementary Table 6.

Exocytosis

It is undoubtedly established, as discussed above, that ApoD is being secreted to the extracellular environment following a canonical secretory pathway (Figure 6). However, whether this pathway leads to actual secretion of ApoD in monomeric form has not been established. In addition, extracellular vesicles (EVs) constitute an alternative exocytotic path for ApoD (see section Binding to Lipid-Rich Structures). Proteomic analyses have identified ApoD in EVs from human plasma and CSF. Experimental characterization of EVs produced by a human astroglial cell line and by mouse primary astrocytes, identified the ApoD-positive vesicles as exosomes originated from multivesicular bodies, according to their size (~100 nm), density (1.17–1.23 g/ml) and molecular markers. When exported by glial cells in exosomes, ApoD must be located on the external surface of these EVs (Figure 6).

A third mechanism by which ApoD becomes extracellular is by traveling in HDL particles. ApoD-HDL association can take place during HDL biogenesis, or ApoD can associate to HDLs while the lipoparticles bind to cell membranes and perform their lipid efflux activity. However, these mechanistic details and the particular subcellular origin of the HDL-associated ApoD detected in body fluids need to be investigated. The plasma membrane location of both ApoA-I dependent HDL biogenesis (Denis et al., 2008) and ABCA1-dependent cholesterol efflux activity (Phillips, 2018), makes it a likely location for the origin of ApoD-positive HDL particles (Figure 6).

References contributing to this section are listed in Reference Collection 34, Supplementary Table 6.

Endocytosis

The immunolocalization of ApoD in cells not expressing the gene (see section Tissue and Cellular Expression Patterns and Response to Stimuli), as well as the internalization of ApoD by cells cultured in the presence of its native or recombinant forms, are the experimental basis supporting the endocytosis of this Lipocalin. It takes place both under control conditions and in response to specific biological stimuli. ApoD endocytosis appears as a general property of this protein, as it has been reported in birds and mammals. Particularly, in glia-neuron co-cultures ApoD is found to be exclusively transported in EVs from astrocytes to neurons, where it gets internalized. The current view of several extracellular formats of ApoD (HDL, EVs or tetramers in solution) makes it worth to study whether different membrane interaction mechanisms or endocytosis paths are used for ApoD internalization.

ApoD association to the extracellular side of the plasma membrane is coherent with both, its traffic from RER to plasma membrane by the canonical exocytotic path and with its cell contact before internalization. ApoD-plasma membrane interaction has been experimentally demonstrated and is currently considered an established localization for ApoD in human cells (https://www.proteinatlas.org/ENSG00000189058-APOD/cell). Whether ApoD-membrane association is mediated by protein-protein or protein-lipid interactions requires further research (see sections Protein-Protein Interactions and Binding to Lipid-Rich Structures). Figure 6 summarizes ApoD intra and extracellular traffic as currently known.

References contributing to this section are listed in Reference Collection 35, Supplementary Table 6.

Tissue and Organ Function

The reports tagged in this section were selected because they study the function of ApoD by experimentally altering ApoD natural expression levels, or by subjecting cells or tissues to defined concentrations of the protein in a controlled experimental situation. A critical review of these reports aims at uncovering common and distinct roles for ApoD in different physiological organ and cellular systems.

ApoD Functions in Cardiovascular System

The process of angiogenesis has been a focus of interest to study the role of ApoD, given its reported expression by blood vessel mural cells (MCs: smooth muscle cells and pericytes). Both in embryonic development and during the remodeling process of wound healing, ApoD increased expression is causally linked to undifferentiated mural cells migration, though is not consistently related to cell proliferation (as it is often found in cancer cells; see section ApoD-Disease Relationships). A crosstalk between endothelial cells (ECs) and MCs governs the switch of the angiogenic cellular process from a proliferative/migratory state to a differentiation state, characterized by quiescence and adhesion. This switch is essential for vessel morphogenesis. Blood vessel angiogenesis involves reactive oxygen species (ROS), EC-derived PDGF-BB, JAGGED1 and NO, as well as MC-expressed NOTCH3. These secreted and cell contact-mediated signaling downregulates ApoD in MCs, a process linked to blood vessel maturation. Experimental manipulation of ApoD levels demonstrates that ApoD regulates the adhesion of MCs to the extracellular matrix, and low levels of ApoD promote Zyxin- and Vinculin-positive focal adhesion contacts. Concordant effects have been reported in retinal choroid vessels: ApoD-KO mice show neovascularization with increased permeability.

Myocardial cells show slight expression of ApoD under normal circumstances, but the tissue surrounding an injured/infarcted area promptly upregulates ApoD. The study of this process in ApoD-KO mice indicates that ApoD is associated to protection from cell death in the injured tissue. This ApoD protective role on cardiomyocytes is dependent on a proper protein fold and strongly correlates with its antioxidant activity (see sections Binding to Lipid-Rich Structures and Protein Physiology). In this experimental paradigm, the protective activity is attained by increasing ApoD in plasma, though it is unclear whether ApoD levels are also elevated in the infarcted tissue. We thus propose that ApoD function in myocardial tissue protection and remodeling might be based on: (1) A modulation of cell viability in cardiomyocytes and vessel ECs, possibly due to internalization of plasma-derived ApoD, and/or (2) a regulation of cell differentiation related to the angiogenic response described above, organized by ECs and MCs.

References contributing to this section are listed in Reference Collection 36, Supplementary Table 7.

Roles of ApoD in Metabolism Regulation

The role of ApoD in metabolism has been analyzed in vivo by using two different ApoD-KO mouse lines and a transgenic mouse (hApoD-Tg) driving the expression of human ApoD under the control of the human THY1 gene. This hApoD-Tg mouse ectopically expresses hApoD mostly in neurons, but the protein is present in plasma and other organs physiologically relevant to metabolism. Also, adenovirus-driven liver production of mouse ApoD has been used as a paradigm of acute overexpression, leading to elevated protein levels in plasma.

The metabolic consequences of altering ApoD levels have been evaluated mostly in plasma and liver, although also in retina, and measured in a variety of experimental settings: fasting or non-fasting conditions, different feeding diets, and different sex or age of animals. No clear pattern can be extracted for the role of ApoD on carbohydrate metabolism, where reports describe varied outcomes on glucose tolerance or insulin resistance depending on experimental conditions. Some consistency is observed in the effects on triglycerides (TG): Loss of ApoD leads to decreased hepatic TG content and increased plasma TG, while overexpression leads to elevated TG levels in liver and unaltered or decreased triglyceridemia (depending on the strategy used for ApoD overexpression). On the other hand, variations in plasma cholesterol levels are also reported, with various outcomes upon ApoD loss or overexpression. Also, association of ApoD polymorphisms have been found with both increased and decreased HDL-cholesterol species. The finding of ApoD being able to mediate binding of HDL to LDL, and of HDL particles to actively dividing carcinoma cells, suggests that it can regulate lipid traffic indirectly by influencing lipoparticle dynamics. Variations in local physiological contexts of this traffic mechanism might contribute in very different ways to the final systemic outputs measured in the experimental settings studied in vivo. However, more work is needed to derive definitive evidence for understanding the role of ApoD in lipid and carbohydrate metabolism. So far, the relevant results indicate that the functional relationship of ApoD with various metabolic parameters is, at most, indirect and dependent on other physiological conditions.

References contributing to this section are listed in Reference Collection 37, Supplementary Table 7.

ApoD Functions in Skeletal System

Bone cells, from bone marrow stem cells (MSCs) to osteoblasts, are reported to express ApoD in cell culture systems (Supplementary Tables 16, 17), and two reports have focused on testing the effects of experimental manipulations of ApoD levels on bone formation and remodeling. Relevant sex and hormone-related patterns have been found using ApoD-KO or hApoD-Tg mice and cell culture systems. With both approaches ApoD appears as an osteogenic factor. Lack of ApoD in mice reduces bone volume and thickness. These effects are observed in trabecular and cortical bone in females, but only in cortical bone in males. Enhanced bone turnover in female ApoD-KO mice is indicated by increased osteoblast surface and osteoclast numbers. Primary MSCs from ApoD-KO mice have lower survival and proliferation, and increased osteoclastogenesis, but an uptake of exogenous hApoD partially reverts their osteogenic potential. When osteoporosis is modeled by glucocorticoid (dexamethasone) treatment after osteogenic induction of MSCs, overexpression of ApoD reverts the effects of dexamethasone, as measured by PI3K/Akt pathway activity and downstream osteogenic gene expression, thus promoting the osteogenic process. Osteogenesis is accompanied by SOD and catalase upregulation, and oxidative damage is associated with glucocorticoid-induced osteoporosis, thus linking ApoD function in this context to its antioxidant activity.

References contributing to this section are listed in Reference Collection 38, Supplementary Table 7.

ApoD Functions in the Nervous System

As presented above, the current evidence supports a general view in which non-neuronal cells become the source of ApoD in response to different stimuli, and neurons count on the Lipocalin for its cellular functions by internalizing ApoD. Neurotransmission is one of those functions modulated by ApoD. Analysis of downstream effects in gene expression in the brain of ApoD-KO or hApoD-Tg mice, reveal an enrichment of genes related to synaptic transmission. Particularly, changes in glutamate, somatostatin, dopamine and acetylcholine neurotransmission have been independently documented by receptor binding assays, HPLC determination of neurotransmitters or their catabolites, or receptor immunodetection. These effects might underlie the behavioral phenotypes related to locomotor function, motor and spatial learning, and retinal function observed in mice with altered expression levels of ApoD.

ApoD addition to cultured neurons results in neuritogenesis and synaptogenesis, which are crucial during neuronal development and underlie neuronal plasticity of established circuits. Neuritogenesis is promoted in immature neurons in culture by the combined addition of ApoD and retinoic acid in the absence of serum. Experiments combining ApoD addition with receptor antagonists indicate that ApoD-dependent neurite extension can be mediated by LDLR, and possibly also by CXCR4 activation, pathways known to be involved in neuronal differentiation.

Another general function of ApoD, extensively analyzed in loss-of-function and transgenic mice, is its role in the glial response to dyshomeostatic changes in the nervous system due to oxidative, metabolic or traumatic stresses. Many studies have reported an acute regulation of ApoD expression under these insults, either experimental or triggered by disease (see sections Regulation of Expression and ApoD-Disease Relationships), supporting an overall neuroprotective role now widely accepted as a functional label for this Lipocalin. Both astrocytes and oligodendrocytes express and secrete ApoD in response to stress. The protein exerts an autocrine and paracrine neural tissue protection, which results in functional preservation of OS-challenged dopaminergic systems, of neurons affected by kainate excitotoxicity or suffering from Aβ-related degeneration. Astrocytes, although not an abundant source of ApoD in basal conditions, quickly respond to OS with a JNK-dependent expression of ApoD, which is secreted to the extracellular milieu as cargo on the surface of extracellular vesicles (Figure 6). The protein is internalized by glial and neuronal cells, improving their viability thanks to a control by ApoD of OS-dependent lipid peroxide accumulation. Moreover, a surge of ApoD in a stressed neural tissue behaves as an off-signal limiting the dimension and duration of gliosis and inflammation. The inflammatory response is linked to OS due to increased PLA2 expression and AA production, among other factors. Quenching of AA is proposed as part of this inflammation control by ApoD (see section Protein Physiology).

A long-lasting homeostasis maintenance role for ApoD has been also proposed in the process of physiological aging of the nervous system, where this protein has been shown as the most consistently overexpressed in primates and rodents. Also, life-expanding strategies in model organisms, like caloric restriction, promote ApoD expression not only in the nervous system but also in cardiac and skeletal muscle (Supplementary Table 11). The homeostatic role predicted by the expression pattern is supported by the phenotypes exhibited by aged ApoD-KO mice, which do not display altered lifespan but do present signs of early neurodegeneration at 3 months of age, with oxidative damage and proteostasis defects in cortex and hippocampus. These alterations underlie cognitive defects and a hyperkinetic phenotype evident in old (21 months) ApoD-KO mice.

The predominant expression of ApoD in myelinating cells under control conditions (oligodendrocytes in CNS and Schwann cells in PNS; see section Regulation of Expression) has prompted experimental studies, using cultured primary cells and ApoD-KO and hApoD-Tg mice, that clearly support the implication of ApoD in the myelination process during development, in the lifelong maintenance of the myelin sheath, and in the remyelination that occurs in response to environmental insults. These processes have been analyzed in the mouse brain corpus callosum as well as in the peripheral sciatic nerve. ApoD is required for a proper and timely response to a crush injury in PNS nerves, helping to recover locomotor function. ApoD promotes myelin clearance and regulates angiogenesis and macrophages recruitment to the wound site, processes that are essential for subsequent axonal regeneration and remyelination. ApoD contributes to optimize myelin clearance, carried out by transdifferentiated Schwann cells and infiltrating macrophages, through two complementary actions: control of lipid-mediated inflammatory signaling and optimization of the phagocytosis process itself. Data indicates that ApoD regulates and control the tissue levels of AA and lysophosphatidylcholine (both in vitro-demonstrated ApoD ligands, Table 2). They are needed for an adequate cytokine inflammatory response and recruitment of bone marrow-derived macrophages. Although macrophages do not express ApoD, the levels of this Lipocalin in the injured nerve environment influence their phagocytic activity, since myelin-associated ApoD is phagocytosed as well. Flow cytometry experiments with primary macrophages demonstrated that ApoD affects the initiation and efficacy of phagocytosis.

A dynamic spatiotemporal regulation of ApoD expression is apparent in myelinating cells, with a prominent increase at the height of postnatal myelination followed by continuous rise throughout life. The absence of ApoD results in a defective and irreversible compaction, mostly in the extracellular leaflet of both CNS and PNS myelin. This altered myelin structure results in a decreased conduction velocity, reported for the sciatic nerve, and compromises motor learning tasks. As downstream effects, both the mTORC1-dependent lipogenic switch and the ERK-mediated growth pathways are altered in the absence of ApoD. A lack of myelin compaction is due to inadequate removal of myelin glycocalyx, mostly affecting gangliosides GM1–2b, GD1b, and GT1b content and distribution. This role of ApoD on glycocalyx physiology was demonstrated to be linked to the adequate subcellular localization of lysosomal and plasma membrane sialidase (Neu1 and Neu3) and of the regulatory Fyn kinase. This mechanism requires preservation of lysosomal membrane integrity (see section Protein Physiology).

References contributing to this section are listed in Reference Collection 39, Supplementary Table 7.

Protein Physiology

In this final section we aim at discussing the available knowledge, derived from state-of-the-art research critically assessed in this review, to give a plausible answer to the central question posed in the Introduction: In order to achieve its pleiotropic roles, does ApoD moonlight between different biochemical functions when expressed in different contexts, or instead ApoD displays a distinctive biochemical role that works on varied physiological situations?

The presence of ApoD in extracellular formats such as lipoprotein particles and exosomes evidences its ability to associate to higher-order lipid structures. According to our systematic search no unambiguous evidence exists for the secretion of protein monomers in native conditions. Non-denaturing electrophoresis analysis of CSF revealed only high-molecular weight ApoD oligomers, while in plasma it has been repeatedly identified in lipoprotein particles preparations. In the particular case of BCF, where an extremely high concentration of ApoD is produced, the protein assembles in tetramers through protein-protein interactions. Finally, ApoD secreted by cultured astrocytes is internalized by neurons only if the conditioned extracellular media has not been depleted of extracellular vesicles. A protein region encompassing the first three β-strands, highly conserved in chordates (Figure 2A), and several hydrophobic patches located at the protein pocket entrance (Figure 3E) are proposed to underlie ApoD self-association and ApoD-lipid structure interactions, respectively.

These results shed doubts on a view of ApoD widely cited (a functional tag in most databases for this Lipocalin) as a “lipid transporter,” a task that a secreted globular monomer could easily achieve.

Unquestionably though, it is the ability of ApoD to bind small hydrophobic ligands of varied shapes inside its β-barrel pocket. However, when free ligands (e.g., AA) have been mechanistically related to ApoD function, binding data are compatible with a buffering or quenching function, or a very local shuttling of the ligand at the most, rather than to a generalized long-range ligand transport between cells. A curious case is the expression of ApoD in feather follicles of pheasants, only in skin areas with specific plumage colors, suggestive of a pigment-retention function. Similarly, the ligand bound to ApoD in sweat from human axilla could be the source of slowly released volatile odor molecules. These ligand-retention functions compare well with that of crustacean ApoD homologs, also linked to their carapace coloration (Wade et al., 2009).

In a different context, ligand shuttling has been repeatedly proposed for ApoD in the cholesterol transfer to LCAT. However, ApoD has been demonstrated not to bind cholesterol, not to contribute to LCAT-cholesterol transfer and not to show a direct interaction with LCAT. On the contrary, experimental data suggests that ApoD exerts “stabilizing effects” on LCAT activity. After reviewing the relevant information on this issue, we propose a different view that can guide new testable hypotheses: ApoD binds lysophosphatidylcholine (LPC), a LCAT reaction product that exerts a negative feedback on LCAT activity. By quenching LPC, ApoD would maintain LCAT activity over a wide range of LPC product concentration. This specific LPC quenching function is compatible with the small amounts of ApoD recovered from HDLs, since only a transitory presence of ApoD might be needed when LCAT is adding cholesterol to the lipoparticle. As for the HDL-LDL interaction (see section Roles of ApoD in Metabolism Regulation), the putative consequences of ApoD presence in HDLs on cholesterol management in the organism would therefore be of an indirect nature, and could explain the lack of correlation between ApoD and cholesterol content in many physiological or pathological situations.

A fundamental advance in defining ApoD molecular function was its role in organismal protection against OS, achieved by a control of the magnitude of lipid peroxidation, measured at tissue or cellular levels. This role has received strong experimental support from in vitro biochemical assays, cellular experimental systems, and in vivo experiments with animal models where ApoD expression was manipulated. Moreover, further validation for this role comes from experimental approaches testing the expression of human ApoD in evolutionary distant organisms. Overexpression of human ApoD in Drosophila increases lifespan in both normal and pro-oxidative experimental conditions. Also, replacement in plants of the native chloroplast Lipocalin (LCNP) by human ApoD, targeted to thylakoids, rescues drought and OS sensitivity of the mutant. Lipid peroxidation control is evidenced in both reports as the mechanism mediating the organism response.

An ApoD antioxidant mechanism has been demonstrated using oxidized AA-derivatives in solution or auto-oxidized liposomes. ApoD is able to reduce free radical-generating lipid hydroperoxides to inert lipid hydroxides. In this reaction, the residue Met93 exposed on one of the surface hydrophobic patches of the protein (Figure 3C) is converted to Met93-sulfoxide. This residue is preserved in ApoD chordate orthologs (Figure 2A) and contributes to the functional differentiation of ApoD from its closest Lipocalin relative, RBP4, where that position is occupied by charged (Lys or Arg) residues (Diez-Hermano et al., 2021). To maintain ApoD antioxidant activity, the action of a methionine sulfoxide reductase (MRS) would be required. However, oxidized ApoD tends to self-associate. Interesting data from Alzheimer's disease brain samples reveal that hippocampal (but not cerebellar) MRS levels decrease with disease progression, while ApoD oligomerization increases. This suggests that the ApoD redox cycling might be blocked if Met93 does not return to its native form and the protein self-associates. This effect sets an upper limit to ApoD antioxidant activity, since it would result in the consumption of ApoD-Met93. Whether this depletion triggers a feedback regulatory loop promoting ApoD gene expression under OS situations in different physiological and pathological contexts would be an interesting aspect to explore. In this context, we must keep in mind that ApoD structure is stable under pro-oxidative situations, making it suitable for the biological contexts where ApoD function is beneficial (from neurodegenerative conditions to cancer). Also, ApoD ligand binding ability is preserved at low pH and its glycosylation prevents a rapid degradation, both good assets to perform its ligand binding and antioxidant functions inside the endolysosomal compartment.

The direct antioxidant activity of ApoD and its demonstrated stable location in the lysosomal compartment put forward a new view of ApoD protein physiology that holds high explanatory power in the understanding of a number of apparently varied ApoD roles.

ApoD control of redox state can be performed directly on both, cell membranes and lipoprotein particles. The unilamellar vesicles where ApoD-reducing activity has been demonstrated are a good experimental model for both types of lipid-based structures. Lipid peroxide products are mainly derived from cellular membranes, which are a major target for cell-generated ROS. The ability of ApoD to keep low levels of membrane-originated lipid peroxides, together with the positive correlation of ApoD content in HDLs with their antioxidant capacity, and the promotion of HDL-LDL interaction by exogenously-added native ApoD, support the protective action of ApoD in both types of lipidic structures (membranes and lipoparticles). Additional evidence comes from the existence of ApoD insect homologs stably anchored to cell membranes (Ganfornina et al., 1995; Ruiz García, 2013), which suggests that membrane interaction is part of an ancestral ApoD property.

In addition to the immediate effects on the redox state of membranes and other lipid structures, ApoD can give rise to indirect effects when performing its antioxidant function in the lysosome. The lysosome is considered a “lipid-controlling” cellular hub. ApoD maintenance of lysosomal membrane redox balance and integrity results in the control of plasma membrane composition. This is for example the case for plasma membrane glycolipids, with important consequences for membrane-membrane interactions like those required in the process of myelin compaction. Lysosomal membrane stability can, by extension, influence the lipid export/import balance in cells, another way of ApoD indirectly conditioning the organism lipid metabolism. Altered ApoD expression in response to mutations of the lysosomal cholesterol transporters (as in Niemann-Pick type C disease) supports this notion. Plasma membrane modulation is also coherent with the observed correlation of ApoD content in HDLs and their ABCA1-dependent cholesterol efflux capability in macrophages, or the subtle changes in lipid content in lipoprotein particles of subjects with ApoD polymorphisms. Through its influence on membranes and lipoparticle dynamics, without a need of binding cholesterol, ApoD can modulate its flux within and between cells.

Additionally, the lysosome is a “cell death/survival controller” by its fundamental recycling, detoxifying and proteostatic functions. Lysosomal ApoD would condition whether a failure in the lysosomal compartment takes place upon a wide array of disease/injury situations, thus contributing to the final cell fate. This ApoD-dependent cell fate decision can be extended to developmental processes as well.

Finally, a role of ApoD in innate immunity has been frequently reported, while no mechanistic link to the protein physiology was proposed. We suggest that ApoD, with its lysosomal optimization mechanism, can modulate the efficiency of phagocytic cells, like it has been demonstrated in injury-recruited macrophages, therefore influencing many of the maintenance and immune responses of the organism.

This view makes us to propose that ApoD lipid-binding properties are more related to management of lipid-based structures composition (membranes or lipoparticles) and a control of their redox state, than to lipid transport. Whether similar membrane-stabilizing properties endow ApoD-positive exosomes with resistance properties to be efficient cargo transporters in disease or tissue damage situations, would be worth studying.

A different aspect of ApoD physiology scarcely studied is the role of its demonstrated N-linked glycosylation, which has been proven to be tissue and species specific, and to be essential for both, ApoD interaction with lipoparticles and for its cellular localization in the endolysosomal compartment. This is particularly important because of the association between redox signaling and glycan profiles, which in turn could affect several signaling pathways (Khoder-Agha and Kietzmann, 2021). In relation to this, modulation of signaling pathways by ApoD has been confirmed in endothelial cells and osteoblasts (PI3K-Akt pathway) and nervous tissue (pERK). How ApoD controls signaling cascades is open to discussion. Although several protein candidates have been proposed as ApoD membrane receptor, no clear demonstration is available for a receptor-mediated signaling transduction. Alternatively, ApoD might not require a protein receptor and trigger a unique signaling cascade. Instead, it could be working as a quencher of lipid modulators (e.g., AA), or conditioning the membrane partitioning of signaling complexes that are known to be dependent on membrane lipids distribution.

In summary, the available information supports a parsimonious hypothesis for the biological function of ApoD, with a unique biochemical role related to the management and redox state of lipid cellular and extracellular structures. This proposition is compatible with the wealth of experimental results showing that multiple stimuli in varied cellular contexts trigger ApoD expression with a tight spatiotemporal regulatory control. The protein can then become associated with the challenged membranes or being exported to the extracellular milieu to act in a paracrine fashion. Both direct and indirect downstream effects, depending on the cell type affected, would explain pleiotropy at the organismal level with a single biochemical function.

The proposed unique molecular mechanism also explains ApoD biological role in response to tissue/organ damage and disease, where homeostatic maintenance is disturbed and ApoD will contribute to restore the equilibrium through tissue repair/reconstruction. Under this paradigm, we can also explain ApoD roles in organismal developmental processes implying building-deconstruction cycles. Figure 7 summarizes the new view on ApoD physiology. References contributing to this section are listed in Reference collection 40, Supplementary Table 8.

FIGURE 7
www.frontiersin.org

Figure 7. ApoD physiology summary. (A) Schematic view of the lipid-managing biochemical function of ApoD. The lipid structure depicted can equally represent the surface of a lipoprotein particle, extracellular vesicle or cellular membrane. ApoD antioxidant activity can be maintained by redox cycling, requiring a reductase activity, or the cycle can terminate by oligomerization of oxidized ApoD. (B) Summary of global tissue function of ApoD, where it contributes to the turnover and maintenance of tissues and organs. This equilibrium is reached after developmental processes in which ApoD is also involved, and switches to a different state upon disease, injury or physiological aging.

Future Goals for ApoD Biology

In spite of the explanatory power of our proposed biological role for ApoD, many questions keep been unresolved and many others are likely to arise, which can spur and guide new research programs. A few of them follow:

(1) To explore the functional relationship between the protein antioxidant capacity and the pocket ligand binding. In this respect, the hypothesis of ApoD working on oxidized lipid “whiskers” (Greenberg et al., 2008; Del Caño-Espinel, 2014) on cell membrane bilayers or lipoparticles is appealing and worth contrasting.

(2) To test whether ApoD downstream effects on signaling pathways rely on a canonical receptor-mediated transduction, or alternatively they depend on the modulation of the lipid context of signaling elements (e.g., PI3K). Findings in Drosophila reveal that loss of an ApoD homolog alters PI3K association to the plasma membrane (Hull-Thompson et al., 2009).

(3) To test whether oligomeric vs. monomeric forms of ApoD underlie its managing function on cell membranes or lipoparticles.

(4) To characterize the extent of ApoD redox cycle, maintaining antioxidant ApoD activity thanks to the intervention of reductases, and the implications of a potential upper limit to this mechanism due to ApoD oligomerization. This aspect can be key to fully understand ApoD function in aging and disease.

(5) To analyze the effects of differential glycosylation on ApoD interactions and functions.

(6) Recent studies on a Drosophila homolog (Yin et al., 2021) point to lipid droplets as another higher-order lipid structure susceptible to be modulated by ApoD. Searching for lipid droplet-managing functions of vertebrate ApoD is therefore pertinent.

(7) At a more general tissue/organ level, several functions are relevant to be studied in more depth, such as the ApoD role on feather and skin physiology, neuronal synaptic function, and metabolism.

(8) Finally, it is worth to analyze the potential exchange between the nervous system and systemic pools of extracellular ApoD in its different formats, not only to fully understand its roles in the organism, but also for a potential therapeutic use of ApoD in nervous system diseases.

ApoD Systematic Review. Reference Collections

Reference Collection 1 (McConathy and Alaupovic, 1973, 1976, 1986; Camato et al., 1989; Kamboh et al., 1989; Holmquist, 1990; Weinberg, 1994; Yang et al., 1994; Holzfeind et al., 1995; Terrisse et al., 2001; Salvatore et al., 2007).

Reference Collection 2 (Eichinger et al., 2007; Oakley et al., 2012; Kielkopf et al., 2018, 2019, 2021).

Reference Collection 3 (McConathy and Alaupovic, 1976, 1986; Bojanovski et al., 1980; Yang et al., 1994; Schindler et al., 1995; Zeng et al., 1996; Sun et al., 1998; Perdomo and Henry Dong, 2009; Li et al., 2016; Qin et al., 2017).

Reference Collection 4 (Blanco-Vaca and Pownall, 1993; Holzfeind et al., 1995; Patel et al., 1997; Böttcher et al., 2000; Nasreen et al., 2006; Bhatia et al., 2012, 2013; Kielkopf et al., 2018, 2019, 2021).

Reference Collection 5 (Haagensen et al., 1979; Lea, 1988; Dilley et al., 1990; Peitsch and Boguski, 1990; Morais Cabral et al., 1995; Zeng et al., 1996; Goessling and Zucker, 2000; Vogt and Skerra, 2001; Breustedt et al., 2006; Akiba et al., 2011; Oakley et al., 2012; Ruiz et al., 2013; García-Mateo et al., 2014; Kielkopf et al., 2019, 2021).

Reference Collection 6 (Fielding and Fielding, 1980; Morton and Zilversmit, 1981; Mahadevan and Soloff, 1983; Steyrer and Kostner, 1988; Camato et al., 1989; Francone et al., 1989; Holmquist, 1989; Sánchez et al., 1992a).

Reference Collection 7 (Utermann et al., 1980; Camato et al., 1989; Yang et al., 1990, 1994; Blanco-Vaca et al., 1992; Liu et al., 2001; Jin et al., 2006; Najyb et al., 2015; Lai et al., 2017).

Reference Collection 8 (McConathy and Alaupovic, 1973, 1976, 1986; Bojanovski et al., 1980; Borghini et al., 1995; Böttcher et al., 2000; Goessling and Zucker, 2000; Perdomo and Henry Dong, 2009; Bhatia et al., 2012; Sreckovic et al., 2013; Braesch-Andersen et al., 2014; Singh et al., 2016; Pascua-Maestro et al., 2018).

Reference Collection 9 (Drayna et al., 1986, 1987; Warden et al., 1992; Provost et al., 1995; Séguin et al., 1995; Cofer and Ross, 1996; Yoshida et al., 1996; Diez-Hermano et al., 2020; Sałkowska et al., 2020).

Reference Collection 10 (Lambert et al., 1993; Yoshida et al., 1996; Do Carmo et al., 2002, 2007; Yamashita et al., 2002; van den Boom et al., 2006; Sasaki et al., 2009; Levros et al., 2010, 2013; Tapia et al., 2011; Ammerpohl et al., 2013; Bajo-Grañeras et al., 2013; Namdar-Aligoodarzi et al., 2015; Diez-Hermano et al., 2020; Sałkowska et al., 2020; Wu et al., 2021).

Reference Collection 11 (Provost et al., 1995; Séguin et al., 1995; Namdar-Aligoodarzi et al., 2015; Lim et al., 2016; Diez-Hermano et al., 2020).

Reference Collection 12 (Drayna et al., 1987; Eichner et al., 1989; Kamboh et al., 1989; Baker et al., 1994; Vijayaraghavan et al., 1994; Desai et al., 2002, 2003; Helisalmi et al., 2004; Hansen et al., 2006; Zhang et al., 2006; Chen et al., 2008; Shibata et al., 2013; Lövkvist et al., 2014; Guven et al., 2019).

Reference Collection 13 (Holmquist, 1990; Holmquist and Vesterberg, 1991; Vizoso et al., 1992, 1994; Blanco-Vaca et al., 1994; Thalmann et al., 1994; Holzfeind et al., 1995; Vieira et al., 1995; Holmquist et al., 1996; Sun et al., 1998; Suresh et al., 1998; Harding et al., 2000; Koch et al., 2001; Yao and Vieira, 2002; Baechle et al., 2006; Do Carmo et al., 2009a; Perdomo and Henry Dong, 2009; Perdomo et al., 2010; Liu et al., 2013; Sreckovic et al., 2013; Ali et al., 2014; Mattsson et al., 2014; Csosz et al., 2015; Waldner et al., 2018; Xu et al., 2018;Kopylov et al., 2020).

Reference Collection 14 (Drayna et al., 1986; Provost et al., 1990, 1991b, 1995; Smith et al., 1990; Spreyer et al., 1990; Escribano et al., 1995; Gillen et al., 1995; Holzfeind et al., 1995; Schaeren-Wiemers et al., 1995; Séguin et al., 1995; Cofer and Ross, 1996; Yoshida et al., 1996; Zeng et al., 1996; Hardardóttir et al., 1997; Suresh et al., 1998; Franz et al., 1999; Montpied et al., 1999; Ong et al., 1999; Terrisse et al., 1999; Zhou et al., 2000; Belloir et al., 2001; Liu et al., 2001; Thomas et al., 2001a,c; Kao et al., 2002; Sánchez et al., 2002; del Valle et al., 2003; Tomarev et al., 2003; Ganfornina et al., 2005, 2008; Hildebrand et al., 2005; Yang et al., 2005; Del Signore et al., 2006; Chen et al., 2007, 2011; Molnár et al., 2007; Zhang et al., 2007, 2014; Befort et al., 2008; Do Carmo et al., 2008, 2009a; Loerch et al., 2008; Rickhag et al., 2008; Collins-Racie et al., 2009; Perdomo and Henry Dong, 2009; Schäfer et al., 2009; Bianchi-Frias et al., 2010; Navarro et al., 2010b; Qin et al., 2010; Bajo-Grañeras et al., 2011a, 2013; Walsh et al., 2012; Germeyer et al., 2013; Manousaki et al., 2013; Edlow et al., 2014; García-Mateo et al., 2014, 2018; Hatzirodos et al., 2015; Jeong et al., 2015; Sanchez et al., 2015; Zheng et al., 2015; Zhu et al., 2015; Gao et al., 2016; Lim et al., 2016; Tao et al., 2017; Carnes et al., 2018; Desmarais et al., 2018; Piórkowska et al., 2018; Flores et al., 2019; Jensen et al., 2019; Diez-Hermano et al., 2020).

Reference Collection 15 (Bouma et al., 1988; Boyles et al., 1990a,b; Escribano et al., 1995; Navarro et al., 1998, 2004, 2008, 2010a,b; Suresh et al., 1998; Franz et al., 1999; Ong et al., 1999, 2002; Kalman et al., 2000; Belloir et al., 2001; Hu et al., 2001; Thomas et al., 2001b; del Valle et al., 2003; Khan et al., 2003; Desai et al., 2005; Hildebrand et al., 2005; Del Signore et al., 2006; Ordoñez et al., 2006; Chen et al., 2007, 2011, 2013; Schröder et al., 2007; Ganfornina et al., 2008; Rickhag et al., 2008; Do Carmo et al., 2009a; Jansen et al., 2009; Qin et al., 2010; Bajo-Grañeras et al., 2011a, 2013; Delattre et al., 2012; Ordóñez et al., 2012; Germeyer et al., 2013; García-Mateo et al., 2014, 2018; Labrie et al., 2015; Martínez-Pinilla et al., 2015; Sanchez et al., 2015; Zheng et al., 2015; Li et al., 2016; Lim et al., 2016; Soria et al., 2017; Desmarais et al., 2018; Flores et al., 2019; Jensen et al., 2019; El-Darzi et al., 2020; Pascua-Maestro et al., 2020).

Reference Collection 16 (Bouma et al., 1988; Boyles et al., 1990a; Schaeren-Wiemers et al., 1995; Vieira et al., 1995; Zeng et al., 1996; Ong et al., 1997; Franz et al., 1999; Kalman et al., 2000; Belloir et al., 2001; Hu et al., 2001; Navarro et al., 2004; Desai et al., 2005; Ganfornina et al., 2005; Hildebrand et al., 2005; Ishii et al., 2005; Loerch et al., 2008; Rickhag et al., 2008; Song et al., 2009; Bajo-Grañeras et al., 2013; Germeyer et al., 2013; García-Mateo et al., 2014; Martínez-Pinilla et al., 2015; Zheng et al., 2015; Li et al., 2016; Flores et al., 2019; Kendal et al., 2020; Pascua-Maestro et al., 2020; Sałkowska et al., 2020;Schlotter et al., 2020).

Reference Collection 17 (Simard et al., 1990, 1991, 1992; Sugimoto et al., 1994; Patel et al., 1995; Harding et al., 2000; Zhou et al., 2000; Yao and Vieira, 2002; Appari et al., 2009; Do Carmo et al., 2009a; Wang et al., 2010; Chen et al., 2011; Tapia et al., 2011; Delattre et al., 2012; Eigeliene et al., 2012; Pérez et al., 2012; Germeyer et al., 2013; Jeong et al., 2015; Tao et al., 2017; Kfir et al., 2018; Hornig et al., 2019; Igarashi et al., 2020).

Reference Collection 18 (Loerch et al., 2008; de Magalhães et al., 2009; Kim et al., 2009; Bianchi-Frias et al., 2010; Navarro et al., 2010b, 2013; Martínez et al., 2012, 2013; Ordóñez et al., 2012; Yan et al., 2012; Sanchez et al., 2015; Martineau et al., 2016; Kosarussavadi et al., 2017; García-Mateo et al., 2018; Waldner et al., 2018; Yu et al., 2020).

Reference Collection 19 (Do Carmo et al., 2007; Ganfornina et al., 2008; Nowicki et al., 2009; Sasaki et al., 2009; Bajo-Grañeras et al., 2011a,b, 2013; Martínez et al., 2013; Dassati et al., 2020; Diez-Hermano et al., 2020; Martínez-Pinilla et al., 2021).

Reference Collection 20 (Blais et al., 1994, 1995; Gillen et al., 1995; Hardardóttir et al., 1997; Ong et al., 1997; Montpied et al., 1999; Trieu and Uckun, 2000; Kim et al., 2001; Leung et al., 2004; Del Signore et al., 2006; Do Carmo et al., 2007, 2008; Rickhag et al., 2008; Qin et al., 2010; Chia et al., 2011; Puntambekar et al., 2011; García-Mateo et al., 2014; Najyb et al., 2017; Scalf et al., 2019; Schlotter et al., 2020).

Reference Collection 21 (Suresh et al., 1998; Hargreaves et al., 1999; Hummasti et al., 2004; Bujalska et al., 2006; Cha et al., 2006; Zhang et al., 2007; Perdomo et al., 2010; Kosacka et al., 2011; Ali et al., 2014; Edlow et al., 2014; Zheng et al., 2015; Lim et al., 2016; Lai et al., 2017; Lambert et al., 2017; Desmarais et al., 2018; Piórkowska et al., 2018; Xu et al., 2018;Saadane et al., 2019).

Reference Collection 22 (Provost et al., 1991b; López-Boado et al., 1994, 1996; Do Carmo et al., 2002, 2007; Kang et al., 2003a; Sarjeant et al., 2003; Bianchi-Frias et al., 2010; Levros et al., 2010; Bajo-Grañeras et al., 2011a; Braesch-Andersen et al., 2014; Martineau et al., 2016).

Reference Collection 23 (Tew et al., 2007; Kosacka et al., 2009, 2011; Pajaniappan et al., 2011; Jensen et al., 2019).

Reference Collection 24 (López-Boado et al., 1997; Thomas et al., 2001a, 2003b; O'Donnell et al., 2003; Befort et al., 2008; Dassati et al., 2020).

Reference Collection 25 (Provost et al., 1991a; Patel et al., 1995; Suresh et al., 1998; Do Carmo et al., 2002; Kao et al., 2002; Kang et al., 2003a; Sarjeant et al., 2003; Leung et al., 2004; Ishii et al., 2005; Bujalska et al., 2006; Fujita et al., 2007; Tew et al., 2007; Kosacka et al., 2009; Pajaniappan et al., 2011; Germeyer et al., 2013; García-Mateo et al., 2014; Martineau et al., 2016; Flores et al., 2019; Saadane et al., 2019; Igarashi et al., 2020; Pascua-Maestro et al., 2020).

Reference Collection 26 (Simard et al., 1990, 1991, 1992; Blais et al., 1994, 1995; López-Boado et al., 1994, 1996, 1997; Sugimoto et al., 1994; Do Carmo et al., 2002, 2007; Sarjeant et al., 2003; Hummasti et al., 2004; Leung et al., 2004; Cha et al., 2006; Tew et al., 2007; Kosacka et al., 2009, 2011; Sasaki et al., 2009; Bianchi-Frias et al., 2010; Levros et al., 2010; Martínez et al., 2012, 2013; Bajo-Grañeras et al., 2013; Braesch-Andersen et al., 2014; Martínez-Pinilla et al., 2015, 2021; Martineau et al., 2016; Lai et al., 2017; Pascua-Maestro et al., 2018; Flores et al., 2019; Dassati et al., 2020).

Reference Collection 27 (Boyles et al., 1990a; Spreyer et al., 1990; Gillen et al., 1995; Yoshida et al., 1996; Dandoy-Dron et al., 1998; Suresh et al., 1998; Terrisse et al., 1998, 1999; Franz et al., 1999; Kalman et al., 2000; Trieu and Uckun, 2000; Belloir et al., 2001; Kim et al., 2001; Lieuallen et al., 2001; Reindl et al., 2001; Thomas et al., 2001b,c, 2003a,d; Mahadik et al., 2002; Ong et al., 2002; Desai et al., 2003, 2005; Glöckner and Ohm, 2003; Kang et al., 2003b; Navarro et al., 2003, 2018; Saha and Rangarajan, 2003; Helisalmi et al., 2004; Li et al., 2005, 2006, 2015; Yao et al., 2005; Hansen et al., 2006; Ordoñez et al., 2006; Rickhag et al., 2006, 2008; Zhang et al., 2006; Almgren et al., 2008; Chen et al., 2008; Dean et al., 2008; Do Carmo et al., 2008; Song et al., 2009; Ruscher et al., 2010; Kroksveen et al., 2012, 2013; Ordóñez et al., 2012; Martínez et al., 2013; Shibata et al., 2013; García-Mateo et al., 2014; Lövkvist et al., 2014; Mattsson et al., 2014; Martínez-Pinilla et al., 2015; Oláh et al., 2015; Del Valle et al., 2016; Lee et al., 2016; Przybycien-Szymanska et al., 2016; Qin et al., 2017; Waldner et al., 2018; Bhatia et al., 2019; Guven et al., 2019; Jiang et al., 2019; Khoonsari et al., 2019; Kuiperij et al., 2019; Perrotte et al., 2019; Zou et al., 2019; Pascua-Maestro et al., 2020; Watanabe et al., 2020; Zhou et al., 2020; Tristán-Noguero et al., 2021; Vardi et al., 2021).

Reference Collection 28 (Balbín et al., 1990; Mazoujian and Haagensen, 1990; Simard et al., 1990, 1991, 1992; Søreide et al., 1991a,b, 1994; Sánchez et al., 1992a,b; Díez-Itza et al., 1994; Aspinall et al., 1995; Chen et al., 1995; Lane et al., 1995; Hall et al., 1996, 2004; López-Boado et al., 1996, 1997; Osundeko et al., 1997; Zhang et al., 1998; Clements et al., 1999; Serra Díaz et al., 1999; Lamelas et al., 2000; Rodríguez et al., 2000; Satoh et al., 2000; Serra et al., 2000; Vázquez et al., 2000; Rojo et al., 2001; Selim et al., 2001; Gutmann et al., 2002; Hunter et al., 2002, 2005a,b; Yamashita et al., 2002; Alvarez et al., 2003, 2004; Linn et al., 2003; Miranda et al., 2003; Alexander et al., 2004; Ashida et al., 2004; Baris et al., 2004; West et al., 2004; Kristensen et al., 2005; Lynch et al., 2005; Ogawa et al., 2005; Ricci et al., 2005; Utsunomiya et al., 2005; Colin et al., 2006; Doane et al., 2006; Jin et al., 2006; van den Boom et al., 2006; Bianchi-Frias et al., 2007, 2010; Carreño et al., 2007; Gonzalez et al., 2007; Vizoso et al., 2007; Yokoo et al., 2007; Galamb et al., 2008; Lisovsky et al., 2008; Quaresima et al., 2008; Song et al., 2008; Fontaine et al., 2009; Søiland et al., 2009a,b; Jinawath et al., 2010; Bidinotto et al., 2012; Palmerini et al., 2012; Bajo-Grañeras et al., 2013; Wang et al., 2013; Jeong et al., 2015; Allina et al., 2016; Sandim et al., 2016; Klebaner et al., 2017; Hu et al., 2018; Cury et al., 2019; Li et al., 2019; Mohammed et al., 2019; Jankovic-Karasoulos et al., 2020; Jiang et al., 2020; Mirza and Abdel-Dayem, 2020; Lopez-Nunez et al., 2021; Wu et al., 2021).

Reference Collection 29 (Wiklund et al., 1980; James et al., 1986; Lin et al., 2001, 2019; Sarjeant et al., 2003; Wei et al., 2008; Perdomo and Henry Dong, 2009; Tsukamoto et al., 2013; Pleva et al., 2015; Cheow et al., 2016; Pamir et al., 2016; Sulkava et al., 2017; Contreras-Duarte et al., 2018; Nazarenko et al., 2018; Caparosa et al., 2019; Ng et al., 2019; Sun et al., 2019; Schlotter et al., 2020).

Reference Collection 30 (Curry et al., 1977; Albers et al., 1981; Cheung et al., 1993; Baker et al., 1994; Vijayaraghavan et al., 1994; Hansen et al., 2004; Iqbal et al., 2008; Navarro et al., 2010a; Edlow et al., 2014; Manjunatha et al., 2016; Ravnsborg et al., 2016; Brahimaj et al., 2017; Herzig et al., 2017; Desmarais et al., 2018; Saadane et al., 2019; Kopylov et al., 2020; Santana et al., 2020).

Reference Collection 31 (Bujalska et al., 2006; Appari et al., 2009; Chen et al., 2011; Allegra et al., 2012; Ammerpohl et al., 2013; Zhu et al., 2015; Salami et al., 2019; Tanase-Nakao et al., 2019; Geng et al., 2020).

Reference Collection 32 (Albers et al., 1984; Haffner et al., 1985; Holmquist et al., 1993; Gottsch et al., 2003; Zenkel et al., 2005; Flatscher-Bader and Wilce, 2006; Tew et al., 2007; Collins-Racie et al., 2009; Schäfer et al., 2009; Sun et al., 2013; Aregger et al., 2014; Hueging et al., 2015; Kliuchnikova et al., 2016; Starodubtseva et al., 2016; Soria et al., 2017; Deng et al., 2018; Forster et al., 2019; Ponnikorn et al., 2019;Liu et al., 2020).

Reference Collection 33 (Bouma et al., 1988; Patel et al., 1995; Navarro et al., 1998, 2004; Suresh et al., 1998; Ong et al., 1999; Hu et al., 2001; Leung et al., 2004; Schröder et al., 2007; Søiland et al., 2009a,b; Ordóñez et al., 2012; Martínez-Pinilla et al., 2015; Klebaner et al., 2017; Pascua-Maestro et al., 2017, 2020; Schlotter et al., 2020).

Reference Collection 34 (Simard et al., 1990, 1991, 1992; Blais et al., 1994; Patel et al., 1995; Suresh et al., 1998; Do Carmo et al., 2007; Sreckovic et al., 2013; Cheow et al., 2016; Pamir et al., 2016; Przybycien-Szymanska et al., 2016; Pascua-Maestro et al., 2017, 2018).

Reference Collection 35 (Vieira et al., 1995; Sarjeant et al., 2003; Thomas et al., 2003c; Hildebrand et al., 2005; Najyb et al., 2015, 2017; Pascua-Maestro et al., 2017, 2018, 2020; Martínez-Pinilla et al., 2021).

Reference Collection 36 (Sarjeant et al., 2003; Pajaniappan et al., 2011; Tsukamoto et al., 2013; Lai et al., 2017).

Reference Collection 37 (Desai et al., 2002; Do Carmo et al., 2009b; Perdomo et al., 2010; Jiménez-Palomares et al., 2011; Ali et al., 2014; Braesch-Andersen et al., 2014; Labrie et al., 2015; Desmarais et al., 2019; El-Darzi et al., 2020).

Reference Collection 38 (Martineau et al., 2016; Yu et al., 2020).

Reference Collection 39 (Hildebrand et al., 2005; Thomas and Yao, 2007; Do Carmo et al., 2008; Ganfornina et al., 2008, 2010; He et al., 2009; Kosacka et al., 2009; Rajput et al., 2009; Boer et al., 2010; Bajo-Grañeras et al., 2011a,b; Kumar, 2012; Martínez et al., 2012; Ruiz et al., 2013; García-Mateo et al., 2014, 2018; Li et al., 2015; Sanchez et al., 2015; Najyb et al., 2017; Bhatia et al., 2019; El-Darzi et al., 2020; Pascua-Maestro et al., 2020).

Reference Collection 40 (Steyrer and Kostner, 1988; Yamashita et al., 2002; Sarjeant et al., 2003; Thomas et al., 2003c; Jin et al., 2006; Eichinger et al., 2007; Thomas and Yao, 2007; Muffat et al., 2008; He et al., 2009; Kosacka et al., 2009; Sasaki et al., 2009; Ganfornina et al., 2010; Bajo-Grañeras et al., 2011a; Pajaniappan et al., 2011; Bhatia et al., 2012, 2013; Martínez et al., 2012; Oakley et al., 2012; Sreckovic et al., 2013; Braesch-Andersen et al., 2014; García-Mateo et al., 2014, 2018; Najyb et al., 2015; Gao et al., 2016; Martineau et al., 2016; Pamir et al., 2016; Lai et al., 2017; Pascua-Maestro et al., 2017, 2018, 2020; Kielkopf et al., 2018, 2019, 2021; El-Darzi et al., 2020;Henri and Rumeau, 2021).

Data Availability Statement

The original contributions presented in the study are included in the article/Supplementary Materials, further inquiries can be directed to the corresponding author.

Author Contributions

MG and DS: conceptualization, writing, review, and editing. Both authors contributed to the article and approved the submitted version.

Funding

This work was supported by a Ministerio de Ciencia e Innovacion grant PID2019-110911RB-I00 to MG and DS.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's Note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Acknowledgments

We gratefully thank Dr. C. Lillo (Salamanca University, Spain), Dr. G. Perdomo (IBGM-CSIC, Spain), and Dr. A. Skerra (Technische Universität München, Germany) for their expert judgement about electron microscopy, metabolism, and protein biochemistry data reviewed in this work. We also want to thank Mrs. Piedad Prado from the University of Valladolid Medical Library for her help in compiling digital copies of some publications needed for this review. Finally, we want to thank the reviewers of the manuscript for their helpful and enriching comments.

Supplementary Material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fphys.2021.738991/full#supplementary-material

Supplementary Figure 1. ApoD mRNA and protein expression summary of the Human Protein Atlas.

Supplementary Figure 2. ApoD mRNA and protein expression summary of the Mouse Gene Expression Database.

Supplementary Table 1. Complete ApoD Reference Library.

Supplementary Tables 2–8. References organized by subheadings within each tag (MP, GD, RE, DR, CT, TOF, and PP).

Supplementary Tables 9–15. Analysis of primary papers describing ApoD mRNA expression or protein detection in vivo in different physiological systems: body fluids, reproductive system, nervous system, skin and glands, musculoskeletal system, cardiovascular and immune systems, and other functional systems (digestive system, liver, pancreas, adipose tissue, kidney).

Supplementary Tables 16, 17. Analysis of primary papers describing ApoD mRNA expression or protein detection in primary cultures and cell lines.

Supplementary Table 18. Analysis of primary papers describing ApoD relationships to disease (expression regulation by disease or treatment, genetic variations associated to disease).

Abbreviations

AA: arachidonic acid; BCF: breast cyst fluid; CSF: cerebrospinal fluid; E-3M2H: E-3-methyl-2-hexenoic acid; ECs: endothelial cells; EM: electron microscopy; EVs: extracellular vesicles; GuHCl: guanidine hydrochloride; HDX-MS: amide hydrogen-deuterium exchange mass spectrometry; LPC: lysophosphatidylcholine; LPS: bacterial lipopolysaccharide; MCs: blood vessel mural cells; MSCs: bone marrow stem cells; OS: oxidative stress; RER: rough endoplasmic reticulum; ROS: reactive oxygen species; SAXS: small-angle X-ray scattering; STR: short-tandem repeats; TG: triglycerides; UTRs: gene untranslated regions.

References

Akiba, S., Arai, N., Kusuoku, H., Takagi, Y., Hagura, T., Takeuchi, K., et al. (2011). The N-terminal amino acid of apolipoprotein D is putatively covalently bound to 3-hydroxy-3-methyl hexanoic acid, a key odour compound in axillary sweat. Int. J. Cosmet. Sci. 33, 283–286. doi: 10.1111/j.1468-2494.2010.00636.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Albers, J. J., Cheung, M. C., Ewens, S. L., and Tollefson, J. H. (1981). Characterization and immunoassay of apolipoprotein D. Atherosclerosis 39, 395–409. doi: 10.1016/0021-9150(81)90025-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Albers, J. J., Taggart, H. M., Applebaum-Bowden, D., Haffner, S., Chesnut, C. H., and Hazzard, W. R. (1984). Reduction of lecithin-cholesterol acyltransferase, apolipoprotein D and the Lp(a) lipoprotein with the anabolic steroid stanozolol. Biochim. Biophys. Acta 795, 293–296. doi: 10.1016/0005-2760(84)90078-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Alexander, H., Stegner, A. L., Wagner-Mann, C., Du Bois, G. C., Alexander, S., and Sauter, E. R. (2004). Proteomic analysis to identify breast cancer biomarkers in nipple aspirate fluid. Clin. Cancer Res. 10, 7500–7510. doi: 10.1158/1078-0432.CCR-04-1002

PubMed Abstract | CrossRef Full Text | Google Scholar

Ali, K., Abo-Ali, E. M., Kabir, M. D., Riggins, B., Nguy, S., Li, L., et al. (2014). A Western-fed diet increases plasma HDL and LDL-cholesterol levels in apoD-/- mice. PLoS ONE 9:e115744. doi: 10.1371/journal.pone.0115744

PubMed Abstract | CrossRef Full Text | Google Scholar

Allegra, A., Marino, A., Peregrin, P. C., Lama, A., García-Segovia, A., Forte, G. I., et al. (2012). Endometrial expression of selected genes in patients achieving pregnancy spontaneously or after ICSI and patients failing at least two ICSI cycles. Reprod. Biomed. Online 25, 481–491. doi: 10.1016/j.rbmo.2012.07.019

PubMed Abstract | CrossRef Full Text | Google Scholar

Allina, D. O., Andreeva, Y. Y., Zavalishina, L. E., Moskvina, L. V., and Frank, G. A. (2016). Estimation of the diagnostic potential of APOD, PTOV1, and EPHA4 for prostatic neoplasms. Arkh. Patol. 78, 9–14. doi: 10.17116/patol20167859-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Almgren, M., Nyengaard, J. R., Persson, B., and Lavebratt, C. (2008). Carbamazepine protects against neuronal hyperplasia and abnormal gene expression in the megencephaly mouse. Neurobiol. Dis. 32, 364–376. doi: 10.1016/j.nbd.2008.07.025

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez, M. L., Barbón, J. J., González, L. O., Abelairas, J., Boto, A., and Vizoso, F. J. (2003). Apolipoprotein D expression in retinoblastoma. Ophthalmic Res. 35, 111–116. doi: 10.1159/000069130

PubMed Abstract | CrossRef Full Text | Google Scholar

Alvarez, M. L., Barbón, J. J., González, L. O., Lamelas, M. L., Vázquez, J., and Vizoso, F. J. (2004). Expression of two androgen-induced proteins (pepsinogen C and apolipoprotein d) in epithelial skin cancers of the eyelids. Ophthalmologica 218, 115–119. doi: 10.1159/000076147

PubMed Abstract | CrossRef Full Text | Google Scholar

Ammerpohl, O., Bens, S., Appari, M., Werner, R., Korn, B., Drop, S. L. S., et al. (2013). Androgen receptor function links human sexual dimorphism to DNA methylation. PLoS ONE 8:e73288. doi: 10.1371/journal.pone.0073288

PubMed Abstract | CrossRef Full Text | Google Scholar

Appari, M., Werner, R., Wünsch, L., Cario, G., Demeter, J., Hiort, O., et al. (2009). Apolipoprotein D (APOD) is a putative biomarker of androgen receptor function in androgen insensitivity syndrome. J. Mol. Med. 87, 623–632. doi: 10.1007/s00109-009-0462-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Aregger, F., Uehlinger, D. E., Witowski, J., Brunisholz, R. A., Hunziker, P., Frey, F. J., et al. (2014). Identification of IGFBP-7 by urinary proteomics as a novel prognostic marker in early acute kidney injury. Kidney Int. 85, 909–919. doi: 10.1038/ki.2013.363

PubMed Abstract | CrossRef Full Text | Google Scholar

Ashida, S., Nakagawa, H., Katagiri, T., Furihata, M., Iiizumi, M., Anazawa, Y., et al. (2004). Molecular features of the transition from prostatic intraepithelial neoplasia (PIN) to prostate cancer: genome-wide gene-expression profiles of prostate cancers and PINs. Cancer Res. 64, 5963–5972. doi: 10.1158/0008-5472.CAN-04-0020

PubMed Abstract | CrossRef Full Text | Google Scholar

Aspinall, J. O., Bentel, J. M., Horsfall, D. J., Haagensen, D. E., Marshall, V. R., and Tilley, W. D. (1995). Differential expression of apolipoprotein-D and prostate specific antigen in benign and malignant prostate tissues. J. Urol. 154, 622–628. doi: 10.1016/S0022-5347(01)67123-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Baechle, D., Flad, T., Cansier, A., Steffen, H., Schittek, B., Tolson, J., et al. (2006). Cathepsin D is present in human eccrine sweat and involved in the postsecretory processing of the antimicrobial peptide DCD-1L. J. Biol. Chem. 281, 5406–5415. doi: 10.1074/jbc.M504670200

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajo-Grañeras, R., Crespo-Sanjuan, J., García-Centeno, R. M., Garrote-Adrados, J. A., Gutierrez, G., García-Tejeiro, M., et al. (2013). Expression and potential role of apolipoprotein D on the death-survival balance of human colorectal cancer cells under oxidative stress conditions. Int. J. Colorectal Dis. 28, 751–766. doi: 10.1007/s00384-012-1616-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajo-Grañeras, R., Ganfornina, M. D., Martín-Tejedor, E., and Sanchez, D. (2011a). Apolipoprotein D mediates autocrine protection of astrocytes and controls their reactivity level, contributing to the functional maintenance of paraquat-challenged dopaminergic systems. Glia 59, 1551–1566. doi: 10.1002/glia.21200

PubMed Abstract | CrossRef Full Text | Google Scholar

Bajo-Grañeras, R., Sanchez, D., Gutierrez, G., González, C., Do Carmo, S., Rassart, E., et al. (2011b). Apolipoprotein D alters the early transcriptional response to oxidative stress in the adult cerebellum. J. Neurochem. 117, 949–960. doi: 10.1111/j.1471-4159.2011.07266.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Baker, W. A., Hitman, G. A., Hawrami, K., McCarthy, M. I., Riikonen, A., Tuomilehto-Wolf, E., et al. (1994). Apolipoprotein D gene polymorphism: a new genetic marker for type 2 diabetic subjects in Nauru and south India. Diabet. Med. 11, 947–952. doi: 10.1111/j.1464-5491.1994.tb00252.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Balbín, M., Freije, J. M., Fueyo, A., Sánchez, L. M., and López-Otín, C. (1990). Apolipoprotein D is the major protein component in cyst fluid from women with human breast gross cystic disease. Biochem. J. 271, 803–807. doi: 10.1042/bj2710803

PubMed Abstract | CrossRef Full Text | Google Scholar

Baris, O., Savagner, F., Nasser, V., Loriod, B., Granjeaud, S., Guyetant, S., et al. (2004). Transcriptional profiling reveals coordinated up-regulation of oxidative metabolism genes in thyroid oncocytic tumors. J. Clin. Endocrinol. Metab. 89, 994–1005. doi: 10.1210/jc.2003-031238

PubMed Abstract | CrossRef Full Text | Google Scholar

Befort, K., Filliol, D., Darcq, E., Ghate, A., Matifas, A., Lardenois, A., et al. (2008). Gene expression is altered in the lateral hypothalamus upon activation of the mu opioid receptor. Ann. N. Y. Acad. Sci. 1129, 175–184. doi: 10.1196/annals.1417.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Belloir, B., Kövari, E., Surini-Demiri, M., and Savioz, A. (2001). Altered apolipoprotein D expression in the brain of patients with Alzheimer disease. J. Neurosci. Res. 64, 61–69. doi: 10.1002/jnr.1054

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatia, S., Jenner, A. M., Li, H., Ruberu, K., Spiro, A. S., Shepherd, C. E., et al. (2013). Increased apolipoprotein D dimer formation in Alzheimer's disease hippocampus is associated with lipid conjugated diene levels. J. Alzheimers Dis. 35, 475–486. doi: 10.3233/JAD-122278

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatia, S., Kim, W. S., Shepherd, C. E., and Halliday, G. M. (2019). Apolipoprotein D upregulation in Alzheimer's disease but not frontotemporal dementia. J. Mol. Neurosci. 67, 125–132. doi: 10.1007/s12031-018-1217-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Bhatia, S., Knoch, B., Wong, J., Kim, W. S., Else, P. L., Oakley, A. J., et al. (2012). Selective reduction of hydroperoxyeicosatetraenoic acids to their hydroxy derivatives by apolipoprotein D: implications for lipid antioxidant activity and Alzheimer's disease. Biochem. J. 442, 713–721. doi: 10.1042/BJ20111166

PubMed Abstract | CrossRef Full Text | Google Scholar

Bianchi-Frias, D., Pritchard, C., Mecham, B. H., Coleman, I. M., and Nelson, P. S. (2007). Genetic background influences murine prostate gene expression: implications for cancer phenotypes. Genome Biol. 8:R117. doi: 10.1186/gb-2007-8-6-r117

PubMed Abstract | CrossRef Full Text | Google Scholar

Bianchi-Frias, D., Vakar-Lopez, F., Coleman, I. M., Plymate, S. R., Reed, M. J., and Nelson, P. S. (2010). The effects of aging on the molecular and cellular composition of the prostate microenvironment. PLoS ONE 5:e12501. doi: 10.1371/journal.pone.0012501

PubMed Abstract | CrossRef Full Text | Google Scholar

Bidinotto, L. T., de Cicco, R. L., Vanegas, J. E., Santucci-Pereira, J., Vanden Heuvel, J. P., Washington, S., et al. (2012). Fish oil alters tamoxifen-modulated expression of mRNAs that encode genes related to differentiation, proliferation, metastasis, and immune response in rat mammary tumors. Nutr. Cancer 64, 991–999. doi: 10.1080/01635581.2012.712736

PubMed Abstract | CrossRef Full Text | Google Scholar

Blais, Y., Sugimoto, K., Carriere, M. C., Haagensen, D. E., Labrie, F., and Simard, J. (1994). Potent stimulatory effect of interleukin-1 alpha on apolipoprotein D and gross cystic disease fluid protein-15 expression in human breast-cancer cells. Int. J. Cancer 59, 400–407. doi: 10.1002/ijc.2910590319

PubMed Abstract | CrossRef Full Text | Google Scholar

Blais, Y., Sugimoto, K., Carrière, M. C., Haagensen, D. E., Labrie, F., and Simard, J. (1995). Interleukin-6 inhibits the potent stimulatory action of androgens, glucocorticoids and interleukin-1 alpha on apolipoprotein D and GCDFP-15 expression in human breast cancer cells. Int. J. Cancer 62, 732–737. doi: 10.1002/ijc.2910620614

PubMed Abstract | CrossRef Full Text | Google Scholar

Blanco-Vaca, F., Gaubatz, J. W., Bren, N., Kottke, B. A., Morrisett, J. D., and Guevara, J. (1994). Identification and quantification of apolipoproteins in addition to apo[a] and apo B-100 in human lipoprotein[a]. Chem. Phys. Lipids 67–68, 35–42. doi: 10.1016/0009-3084(94)90122-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Blanco-Vaca, F., and Pownall, H. J. (1993). Disulfide linked dimers of apolipoprotein D in urine. Electrophoresis 14, 1086–1087. doi: 10.1002/elps.11501401175

PubMed Abstract | CrossRef Full Text | Google Scholar

Blanco-Vaca, F., Via, D. P., Yang, C. Y., Massey, J. B., and Pownall, H. J. (1992). Characterization of disulfide-linked heterodimers containing apolipoprotein D in human plasma lipoproteins. J. Lipid Res. 33, 1785–1796. doi: 10.1016/S0022-2275(20)41336-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Boer, S., Sanchez, D., Reinieren, I., van den Boom, T., Udawela, M., Scarr, E., et al. (2010). Decreased kainate receptors in the hippocampus of apolipoprotein D knockout mice. Prog. Neuropsychopharmacol. Biol. Psychiatry 34, 271–278. doi: 10.1016/j.pnpbp.2009.11.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Bojanovski, D., Alaupovic, P., McConathy, W. J., and Kelly, J. L. (1980). Isolation and partial characterization of apolipoprotein D and lipoprotein D from baboon plasma. FEBS Lett. 112, 251–254. doi: 10.1016/0014-5793(80)80191-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Borghini, I., Barja, F., Pometta, D., and James, R. W. (1995). Characterization of subpopulations of lipoprotein particles isolated from human cerebrospinal fluid. Biochim. Biophys. Acta 1255, 192–200. doi: 10.1016/0005-2760(94)00232-N

PubMed Abstract | CrossRef Full Text | Google Scholar

Böttcher, A., Schlosser, J., Kronenberg, F., Dieplinger, H., Knipping, G., Lackner, K. J., et al. (2000). Preparative free-solution isotachophoresis for separation of human plasma lipoproteins: apolipoprotein and lipid composition of HDL subfractions. J. Lipid Res. 41, 905–915. doi: 10.1016/S0022-2275(20)32032-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Bouma, M. E., de Bandt, J. P., Ayrault-Jarrier, M., Burdin, J., Verthier, N., and Raisonnier, A. (1988). Immunoperoxidase localization of apolipoprotein D in human enterocytes and hepatocytes. Scand. J. Gastroenterol. 23, 477–483. doi: 10.3109/00365528809093897

PubMed Abstract | CrossRef Full Text | Google Scholar

Boyles, J. K., Notterpek, L. M., and Anderson, L. J. (1990a). Accumulation of apolipoproteins in the regenerating and remyelinating mammalian peripheral nerve. Identification of apolipoprotein D, apolipoprotein A-IV, apolipoprotein E, and apolipoprotein A-I. J. Biol. Chem. 265, 17805–17815.

PubMed Abstract | Google Scholar

Boyles, J. K., Notterpek, L. M., Wardell, M. R., and Rall, S. C. (1990b). Identification, characterization, and tissue distribution of apolipoprotein D in the rat. J. Lipid Res. 31, 2243–2256.

PubMed Abstract | Google Scholar

Braesch-Andersen, S., Beckman, L., Paulie, S., and Kumagai-Braesch, M. (2014). ApoD mediates binding of HDL to LDL and to growing T24 carcinoma. PLoS ONE 9:e115180. doi: 10.1371/journal.pone.0115180

PubMed Abstract | CrossRef Full Text | Google Scholar

Brahimaj, A., Ligthart, S., Ikram, M. A., Hofman, A., Franco, O. H., Sijbrands, E. J. G., et al. (2017). Serum levels of apolipoproteins and incident type 2 diabetes: a prospective cohort study. Diabetes Care 40, 346–351. doi: 10.2337/dc16-1295

PubMed Abstract | CrossRef Full Text | Google Scholar

Breustedt, D. A., Schönfeld, D. L., and Skerra, A. (2006). Comparative ligand-binding analysis of ten human lipocalins. Biochim. Biophys. Acta 1764, 161–173. doi: 10.1016/j.bbapap.2005.12.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Bujalska, I. J., Quinkler, M., Tomlinson, J. W., Montague, C. T., Smith, D. M., and Stewart, P. M. (2006). Expression profiling of 11beta-hydroxysteroid dehydrogenase type-1 and glucocorticoid-target genes in subcutaneous and omental human preadipocytes. J. Mol. Endocrinol. 37, 327–340. doi: 10.1677/jme.1.02048

PubMed Abstract | CrossRef Full Text | Google Scholar

Camato, R., Marcel, Y. L., Milne, R. W., Lussier-Cacan, S., and Weech, P. K. (1989). Protein polymorphism of a human plasma apolipoprotein D antigenic epitope. J. Lipid Res. 30, 865–875. doi: 10.1016/S0022-2275(20)38304-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Caparosa, E. M., Sedgewick, A. J., Zenonos, G., Zhao, Y., Carlisle, D. L., Stefaneanu, L., et al. (2019). Regional molecular signature of the symptomatic atherosclerotic carotid plaque. Neurosurgery 85, E284–E293. doi: 10.1093/neuros/nyy470

PubMed Abstract | CrossRef Full Text | Google Scholar

Carnes, M. U., Allingham, R. R., Ashley-Koch, A., and Hauser, M. A. (2018). Transcriptome analysis of adult and fetal trabecular meshwork, cornea, and ciliary body tissues by RNA sequencing. Exp. Eye Res. 167, 91–99. doi: 10.1016/j.exer.2016.11.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Carreño, G., Del Casar, J. M., Corte, M. D., González, L. O., Bongera, M., Merino, A. M., et al. (2007). Local recurrence after mastectomy for breast cancer: analysis of clinicopathological, biological and prognostic characteristics. Breast Cancer Res. Treat. 102, 61–73. doi: 10.1007/s10549-006-9310-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Cha, M.-H., Kim, I.-C., Lee, B.-H., and Yoon, Y. (2006). Baicalein inhibits adipocyte differentiation by enhancing COX-2 expression. J. Med. Food 9, 145–153. doi: 10.1089/jmf.2006.9.145

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Li, Y., Du, J., Cao, Y., and Li, X. (2011). Increased JNK1 activity contributes to the upregulation of ApoD in the apocrine secretory gland cells from axillary osmidrosis. Mol. Cell. Biochem. 354, 311–316. doi: 10.1007/s11010-011-0830-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Yang, G., Li, Y., Li, X., and Du, J. (2013). Expression of apolipoprotein D and androgen receptor in axillary osmidrosis and its molecular mechanism. Int. J. Clin. Exp. Med. 6, 497–503.

PubMed Abstract | Google Scholar

Chen, J., Heerdt, B. G., and Augenlicht, L. H. (1995). Presence and instability of repetitive elements in sequences the altered expression of which characterizes risk for colonic cancer. Cancer Res. 55, 174–180.

PubMed Abstract | Google Scholar

Chen, Y., Jia, L., Wei, C., Wang, F., Lv, H., and Jia, J. (2008). Association between polymorphisms in the apolipoprotein D gene and sporadic Alzheimer's disease. Brain Res. 1233, 196–202. doi: 10.1016/j.brainres.2008.07.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y.-W., Gregory, C. M., Scarborough, M. T., Shi, R., Walter, G. A., and Vandenborne, K. (2007). Transcriptional pathways associated with skeletal muscle disuse atrophy in humans. Physiol. Genomics 31, 510–520. doi: 10.1152/physiolgenomics.00115.2006

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheow, E. S. H., Cheng, W. C., Lee, C. N., de Kleijn, D., Sorokin, V., and Sze, S. K. (2016). Plasma-derived extracellular vesicles contain predictive biomarkers and potential therapeutic targets for Myocardial Ischemic (MI) injury. Mol. Cell Proteomics 15, 2628–2640. doi: 10.1074/mcp.M115.055731

PubMed Abstract | CrossRef Full Text | Google Scholar

Cheung, M. C., Mendez, A. J., Wolf, A. C., and Knopp, R. H. (1993). Characterization of apolipoprotein A-I- and A-II-containing lipoproteins in a new case of high density lipoprotein deficiency resembling Tangier disease and their effects on intracellular cholesterol efflux. J. Clin. Invest. 91, 522–529. doi: 10.1172/JCI116231

PubMed Abstract | CrossRef Full Text | Google Scholar

Chia, W.-J., Dawe, G. S., and Ong, W.-Y. (2011). Expression and localization of the iron-siderophore binding protein lipocalin 2 in the normal rat brain and after kainate-induced excitotoxicity. Neurochem. Int. 59, 591–599. doi: 10.1016/j.neuint.2011.04.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Clements, J. A., Rohde, P., Allen, V., Hyland, V. J., Samaratunga, M. L., Tilley, W. D., et al. (1999). Molecular detection of prostate cells in ejaculate and urethral washings in men with suspected prostate cancer. J. Urol. 161, 1337–1343. doi: 10.1016/S0022-5347(01)61680-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Cofer, S., and Ross, S. R. (1996). The murine gene encoding apolipoprotein D exhibits a unique expression pattern as compared to other species. Gene 171, 261–263. doi: 10.1016/0378-1119(96)00099-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Colin, C., Baeza, N., Bartoli, C., Fina, F., Eudes, N., Nanni, I., et al. (2006). Identification of genes differentially expressed in glioblastoma versus pilocytic astrocytoma using Suppression Subtractive Hybridization. Oncogene 25, 2818–2826. doi: 10.1038/sj.onc.1209305

PubMed Abstract | CrossRef Full Text | Google Scholar

Collins-Racie, L. A., Yang, Z., Arai, M., Li, N., Majumdar, M. K., Nagpal, S., et al. (2009). Global analysis of nuclear receptor expression and dysregulation in human osteoarthritic articular cartilage: reduced LXR signaling contributes to catabolic metabolism typical of osteoarthritis. Osteoarthr. Cartil. 17, 832–842. doi: 10.1016/j.joca.2008.12.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Contreras-Duarte, S., Chen, P., Andía, M., Uribe, S., Irarrázaval, P., and Kopp, S.. (2018). Attenuation of atherogenic apo B-48-dependent hyperlipidemia and high density lipoprotein remodeling induced by vitamin C and E combination and their beneficial effect on lethal ischemic heart disease in mice. Biol. Res. 51:34. doi: 10.1186/s40659-018-0183-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Csosz, É., Emri, G., Kalló, G., Tsaprailis, G., and Tozsér, J. (2015). Highly abundant defense proteins in human sweat as revealed by targeted proteomics and label-free quantification mass spectrometry. J. Eur. Acad. Dermatol. Venereol. 29, 2024–2031. doi: 10.1111/jdv.13221

PubMed Abstract | CrossRef Full Text | Google Scholar

Curry, M. D., McConathy, W. J., and Alaupovic, P. (1977). Quantitative determination of human apolipoprotein D by electroimmunoassay and radial immunodiffusion. Biochim. Biophys. Acta 491, 232–241. doi: 10.1016/0005-2795(77)90059-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Cury, S. S., de Moraes, D., Freire, P. P., de Oliveira, G., Marques, D. V. P., Fernandez, G. J., et al. (2019). Tumor transcriptome reveals high expression of IL-8 in non-small cell lung cancer patients with low pectoralis muscle area and reduced survival. Cancers 11:1251. doi: 10.3390/cancers11091251

PubMed Abstract | CrossRef Full Text | Google Scholar

Dandoy-Dron, F., Guillo, F., Benboudjema, L., Deslys, J. P., Lasmézas, C., Dormont, D., et al. (1998). Gene expression in scrapie. Cloning of a new scrapie-responsive gene and the identification of increased levels of seven other mRNA transcripts. J. Biol. Chem. 273, 7691–7697. doi: 10.1074/jbc.273.13.7691

PubMed Abstract | CrossRef Full Text | Google Scholar

Dassati, S., Schweigreiter, R., Buechner, S., and Waldner, A. (2020). Celecoxib promotes survival and upregulates the expression of neuroprotective marker genes in two different in vitro models of Parkinson's disease. Neuropharmacology 194:108378. doi: 10.1016/j.neuropharm.2020.108378

PubMed Abstract | CrossRef Full Text | Google Scholar

de Magalhães, J. P., Curado, J., and Church, G. M. (2009). Meta-analysis of age-related gene expression profiles identifies common signatures of aging. Bioinformatics 25, 875–881. doi: 10.1093/bioinformatics/btp073

PubMed Abstract | CrossRef Full Text | Google Scholar

Dean, B., Digney, A., Sundram, S., Thomas, E., and Scarr, E. (2008). Plasma apolipoprotein E is decreased in schizophrenia spectrum and bipolar disorder. Psychiatry Res. 158, 75–78. doi: 10.1016/j.psychres.2007.05.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Del Caño-Espinel, M. del. (2014). Relación de la apolipoproteína D y sus homólogos en Drosophila con las membranas biológicas: Estudio de su función en diferentes procesos celulares y de su localización y efectos sobre las balsas lipídicas (Ph.D. thesis). Valladolid, Spain. doi: 10.35376/10324/7752

CrossRef Full Text | Google Scholar

Del Signore, A., De Sanctis, V., Di Mauro, E., Negri, R., Perrone-Capano, C., and Paggi, P. (2006). Gene expression pathways induced by axotomy and decentralization of rat superior cervical ganglion neurons. Eur. J. Neurosci. 23, 65–74. doi: 10.1111/j.1460-9568.2005.04520.x

PubMed Abstract | CrossRef Full Text | Google Scholar

del Valle, E., Navarro, A., Astudillo, A., and Tolivia, J. (2003). Apolipoprotein D expression in human brain reactive astrocytes. J. Histochem. Cytochem. 51, 1285–1290. doi: 10.1177/002215540305101005

PubMed Abstract | CrossRef Full Text | Google Scholar

Del Valle, E., Navarro, A., Martínez-Pinilla, E., Torices, S., and Tolivia, J. (2016). Apo J and Apo D: complementary or antagonistic roles in Alzheimer's disease? J. Alzheimers Dis. 53, 639–650. doi: 10.3233/JAD-160032

PubMed Abstract | CrossRef Full Text | Google Scholar

Delattre, C., Winstall, E., Lessard, C., Donovan, M., Simonetti, L., Minondo, A.-M., et al. (2012). Proteomic analysis identifies new biomarkers for postmenopausal and dry skin. Exp. Dermatol. 21, 205–210. doi: 10.1111/j.1600-0625.2011.01434.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, P., Zeng, J., Li, J., Feng, W., Chen, J., and Zeng, Y. (2018). Screening of serum protein markers for avascular osteonecrosis of femoral head differentially expressed after treatment with yuanshi shengmai chenggu tablets. Biomed. Res. Int. 2018:5692735. doi: 10.1155/2018/5692735

PubMed Abstract | CrossRef Full Text | Google Scholar

Denis, M., Landry, Y. D., and Zha, X. (2008). ATP-binding cassette A1-mediated lipidation of apolipoprotein A-I occurs at the plasma membrane and not in the endocytic compartments. J. Biol. Chem. 283, 16178–16186. doi: 10.1074/jbc.M709597200

PubMed Abstract | CrossRef Full Text | Google Scholar

Desai, P. P., Bunker, C. H., Ukoli, F. A. M., and Kamboh, M. I. (2002). Genetic variation in the apolipoprotein D gene among African blacks and its significance in lipid metabolism. Atherosclerosis 163, 329–338. doi: 10.1016/S0021-9150(02)00012-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Desai, P. P., Hendrie, H. C., Evans, R. M., Murrell, J. R., DeKosky, S. T., and Kamboh, M. I. (2003). Genetic variation in apolipoprotein D affects the risk of Alzheimer disease in African-Americans. Am. J. Med. Genet. B Neuropsychiatr. Genet. 116B, 98–101. doi: 10.1002/ajmg.b.10798

PubMed Abstract | CrossRef Full Text | Google Scholar

Desai, P. P., Ikonomovic, M. D., Abrahamson, E. E., Hamilton, R. L., Isanski, B. A., Hope, C. E., et al. (2005). Apolipoprotein D is a component of compact but not diffuse amyloid-beta plaques in Alzheimer's disease temporal cortex. Neurobiol. Dis. 20, 574–582. doi: 10.1016/j.nbd.2005.04.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Desmarais, F., Bergeron, K.-F., Lacaille, M., Lemieux, I., Bergeron, J., Biron, S., et al. (2018). High ApoD protein level in the round ligament fat depot of severely obese women is associated with an improved inflammatory profile. Endocrine 61, 248–257. doi: 10.1007/s12020-018-1621-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Desmarais, F., Bergeron, K.-F., Rassart, E., and Mounier, C. (2019). Apolipoprotein D overexpression alters hepatic prostaglandin and omega fatty acid metabolism during the development of a non-inflammatory hepatic steatosis. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 1864, 522–531. doi: 10.1016/j.bbalip.2019.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

Diez-Hermano, S., Ganfornina, M. D., Skerra, A., Gutierrez, G., and Sanchez, D. (2021). An evolutionary perspective of the Lipocalin protein family. Front. Physiol. 12:718983. doi: 10.3389/fphys.2021.718983

PubMed Abstract | CrossRef Full Text | Google Scholar

Diez-Hermano, S., Mejias, A., Sanchez, D., Gutierrez, G., and Ganfornina, M. D. (2020). Control of the neuroprotective Lipocalin Apolipoprotein D expression by alternative promoter regions and differentially expressed mRNA 5' UTR variants. PLoS ONE 15:e0234857. doi: 10.1371/journal.pone.0234857

PubMed Abstract | CrossRef Full Text | Google Scholar

Díez-Itza, I., Vizoso, F., Merino, A. M., Sánchez, L. M., Tolivia, J., Fernández, J., et al. (1994). Expression and prognostic significance of apolipoprotein D in breast cancer. Am. J. Pathol. 144, 310–320.

PubMed Abstract | Google Scholar

Dilley, W. G., Haagensen, D. E., Cox, C. E., and Wells, S. A. (1990). Immunologic and steroid binding properties of the GCDFP-24 protein isolated from human breast gross cystic disease fluid. Breast Cancer Res. Treat. 16, 253–260. doi: 10.1007/BF01806333

PubMed Abstract | CrossRef Full Text | Google Scholar

Do Carmo, S., Forest, J.-C., Giguère, Y., Masse, A., Lafond, J., and Rassart, E. (2009a). Modulation of Apolipoprotein D levels in human pregnancy and association with gestational weight gain. Reprod. Biol. Endocrinol. 7:92. doi: 10.1186/1477-7827-7-92

PubMed Abstract | CrossRef Full Text | Google Scholar

Do Carmo, S., Fournier, D., Mounier, C., and Rassart, E. (2009b). Human apolipoprotein D overexpression in transgenic mice induces insulin resistance and alters lipid metabolism. Am. J. Physiol. Endocrinol. Metab. 296, E802–E811. doi: 10.1152/ajpendo.90725.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Do Carmo, S., Jacomy, H., Talbot, P. J., and Rassart, E. (2008). Neuroprotective effect of apolipoprotein D against human coronavirus OC43-induced encephalitis in mice. J. Neurosci. 28, 10330–10338. doi: 10.1523/JNEUROSCI.2644-08.2008

PubMed Abstract | CrossRef Full Text | Google Scholar

Do Carmo, S., Levros, L.-C., and Rassart, E. (2007). Modulation of apolipoprotein D expression and translocation under specific stress conditions. Biochim. Biophys. Acta 1773, 954–969. doi: 10.1016/j.bbamcr.2007.03.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Do Carmo, S., Séguin, D., Milne, R., and Rassart, E. (2002). Modulation of apolipoprotein D and apolipoprotein E mRNA expression by growth arrest and identification of key elements in the promoter. J. Biol. Chem. 277, 5514–5523. doi: 10.1074/jbc.M105057200

PubMed Abstract | CrossRef Full Text | Google Scholar

Doane, A. S., Danso, M., Lal, P., Donaton, M., Zhang, L., Hudis, C., et al. (2006). An estrogen receptor-negative breast cancer subset characterized by a hormonally regulated transcriptional program and response to androgen. Oncogene 25, 3994–4008. doi: 10.1038/sj.onc.1209415

PubMed Abstract | CrossRef Full Text | Google Scholar

Drayna, D., Fielding, C., McLean, J., Baer, B., Castro, G., Chen, E., et al. (1986). Cloning and expression of human apolipoprotein D cDNA. J. Biol. Chem. 261, 16535–16539. doi: 10.1016/S0021-9258(18)66599-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Drayna, D., Scott, J. D., and Lawn, R. (1987). Multiple RFLPs at the human apolipoprotein D (APOD) locus. Nucleic Acids Res. 15:9617. doi: 10.1093/nar/15.22.9617

PubMed Abstract | CrossRef Full Text | Google Scholar

Edlow, A. G., Vora, N. L., Hui, L., Wick, H. C., Cowan, J. M., and Bianchi, D. W. (2014). Maternal obesity affects fetal neurodevelopmental and metabolic gene expression: a pilot study. PLoS ONE 9:e88661. doi: 10.1371/journal.pone.0088661

PubMed Abstract | CrossRef Full Text | Google Scholar

Eichinger, A., Nasreen, A., Kim, H. J., and Skerra, A. (2007). Structural insight into the dual ligand specificity and mode of high density lipoprotein association of apolipoprotein D. J. Biol. Chem. 282, 31068–31075. doi: 10.1074/jbc.M703552200

PubMed Abstract | CrossRef Full Text | Google Scholar

Eichner, J. E., Kuller, L. H., Ferrell, R. E., and Kamboh, M. I. (1989). Phenotypic effects of apolipoprotein structural variation on lipid profiles. IV. Apolipoprotein polymorphisms in a small group of black women from the healthy women study. Genet. Epidemiol. 6, 681–689. doi: 10.1002/gepi.1370060605

PubMed Abstract | CrossRef Full Text | Google Scholar

Eigeliene, N., Elo, T., Linhala, M., Hurme, S., Erkkola, R., and Härkönen, P. (2012). Androgens inhibit the stimulatory action of 17β-estradiol on normal human breast tissue in explant cultures. J. Clin. Endocrinol. Metab. 97, E1116–E1127. doi: 10.1210/jc.2011-3228

PubMed Abstract | CrossRef Full Text | Google Scholar

El-Darzi, N., Mast, N., Petrov, A. M., Dao, T., Astafev, A. A., Saadane, A., et al. (2020). Studies of ApoD-/- and ApoD-/-ApoE-/- mice uncover the APOD significance for retinal metabolism, function, and status of chorioretinal blood vessels. Cell. Mol. Life Sci. 78, 963–983. doi: 10.1007/s00018-020-03546-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Escribano, J., Ortego, J., and Coca-Prados, M. (1995). Isolation and characterization of cell-specific cDNA clones from a subtractive library of the ocular ciliary body of a single normal human donor: transcription and synthesis of plasma proteins. J. Biochem. 118, 921–931. doi: 10.1093/jb/118.5.921

PubMed Abstract | CrossRef Full Text | Google Scholar

Fielding, P. E., and Fielding, C. J. (1980). A cholesteryl ester transfer complex in human plasma. Proc. Natl. Acad. Sci. U.S.A. 77, 3327–3330.

PubMed Abstract | Google Scholar

Flatscher-Bader, T., and Wilce, P. A. (2006). Chronic smoking and alcoholism change expression of selective genes in the human prefrontal cortex. Alcohol. Clin. Exp. Res. 30, 908–915. doi: 10.1111/j.1530-0277.2006.00106.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Flores, R., Jin, X., Chang, J., Zhang, C., Cogan, D. G., Schaefer, E. J., et al. (2019). LCAT, ApoD, and ApoA1 expression and review of cholesterol deposition in the cornea. Biomolecules 9:785. doi: 10.3390/biom9120785

PubMed Abstract | CrossRef Full Text | Google Scholar

Fontaine, J.-F., Mirebeau-Prunier, D., Raharijaona, M., Franc, B., Triau, S., Rodien, P., et al. (2009). Increasing the number of thyroid lesions classes in microarray analysis improves the relevance of diagnostic markers. PLoS ONE 4:e7632. doi: 10.1371/journal.pone.0007632

PubMed Abstract | CrossRef Full Text | Google Scholar

Forster, C. S., Haffey, W. D., Bennett, M., Greis, K. D., and Devarajan, P. (2019). Identification of urinary CD44 and prosaposin as specific biomarkers of urinary tract infections in children with neurogenic bladders. Biomark Insights 14, 1–7. doi: 10.1177/1177271919835570

PubMed Abstract | CrossRef Full Text | Google Scholar

Francone, O. L., Gurakar, A., and Fielding, C. (1989). Distribution and functions of lecithin:cholesterol acyltransferase and cholesteryl ester transfer protein in plasma lipoproteins. Evidence for a functional unit containing these activities together with apolipoproteins A-I and D that catalyzes the esterification and transfer of cell-derived cholesterol. J. Biol. Chem. 264, 7066–7072. doi: 10.1016/S0021-9258(18)83541-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Franz, G., Reindl, M., Patel, S. C., Beer, R., Unterrichter, I., Berger, T., et al. (1999). Increased expression of apolipoprotein D following experimental traumatic brain injury. J. Neurochem. 73, 1615–1625. doi: 10.1046/j.1471-4159.1999.0731615.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Fujita, T., Iwata, T., Shiba, H., Igarashi, A., Hirata, R., Takeda, K., et al. (2007). Identification of marker genes distinguishing human periodontal ligament cells from human mesenchymal stem cells and human gingival fibroblasts. J. Periodont. Res. 42, 283–286. doi: 10.1111/j.1600-0765.2006.00944.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Galamb, O., Sipos, F., Solymosi, N., Spisák, S., Krenács, T., Tóth, K., et al. (2008). Diagnostic mRNA expression patterns of inflamed, benign, and malignant colorectal biopsy specimen and their correlation with peripheral blood results. Cancer Epidemiol. Biomarkers Prev. 17, 2835–2845. doi: 10.1158/1055-9965.EPI-08-0231

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganfornina, M. D., Do Carmo, S., Lora, J. M., Torres-Schumann, S., Vogel, M., Allhorn, M., et al. (2008). Apolipoprotein D is involved in the mechanisms regulating protection from oxidative stress. Aging Cell 7, 506–515. doi: 10.1111/j.1474-9726.2008.00395.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganfornina, M. D., Do Carmo, S., Martínez, E., Tolivia, J., Navarro, A., Rassart, E., et al. (2010). ApoD, a glia-derived apolipoprotein, is required for peripheral nerve functional integrity and a timely response to injury. Glia 58, 1320–1334. doi: 10.1002/glia.21010

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganfornina, M. D., Gutiérrez, G., and Bastiani, M.S. D. (2000). A phylogenetic analysis of the lipocalin protein family. Mol. Biol. Evol. 17, 114–126. doi: 10.1093/oxfordjournals.molbev.a026224

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganfornina, M. D., Sánchez, D., and Bastiani, M. J. (1995). Lazarillo, a new GPI-linked surface lipocalin, is restricted to a subset of neurons in the grasshopper embryo. Development 121, 123–134. doi: 10.1242/dev.121.1.123

PubMed Abstract | CrossRef Full Text | Google Scholar

Ganfornina, M. D., Sánchez, D., Pagano, A., Tonachini, L., Descalzi-Cancedda, F., and Martínez, S. (2005). Molecular characterization and developmental expression pattern of the chicken apolipoprotein D gene: implications for the evolution of vertebrate lipocalins. Dev. Dyn. 232, 191–199. doi: 10.1002/dvdy.20193

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, G.-Q., Song, L.-S., Tong, B., and Li, G.-P. (2016). Expression levels of GSTA2 and APOD genes might be associated with carotenoid coloration in golden pheasant (Chrysolophus pictus) plumage. Zool. Res. 37, 144–150. doi: 10.13918/j.issn.2095-8137.2016.3.144

PubMed Abstract | CrossRef Full Text | Google Scholar

García-Mateo, N., Ganfornina, M. D., Montero, O., Gijón, M. A., Murphy, R. C., and Sanchez, D. (2014). Schwann cell-derived Apolipoprotein D controls the dynamics of post-injury myelin recognition and degradation. Front. Cell Neurosci 8:374. doi: 10.3389/fncel.2014.00374

PubMed Abstract | CrossRef Full Text | Google Scholar

García-Mateo, N., Pascua-Maestro, R., Pérez-Castellanos, A., Lillo, C., Sanchez, D., and Ganfornina, M. D. (2018). Myelin extracellular leaflet compaction requires apolipoprotein D membrane management to optimize lysosomal-dependent recycling and glycocalyx removal. Glia 66, 670–687. doi: 10.1002/glia.23274

PubMed Abstract | CrossRef Full Text

Geng, Y.-J., Li, T., Wen, L., Gao, H., Yuan, J., Liu, W.-C., et al. (2020). Percutaneous interstitial nd:YAG laser therapy for axillary osmidrosis. Lasers Surg. Med. 52, 639–646. doi: 10.1002/lsm.23187

PubMed Abstract | CrossRef Full Text | Google Scholar

Germeyer, A., Capp, E., Schlicksupp, F., Jauckus, J., von Rango, U., von Wolff, M., et al. (2013). Cell-type specific expression and regulation of apolipoprotein D and E in human endometrium. Eur. J. Obstet. Gynecol. Reprod. Biol. 170, 487–491. doi: 10.1016/j.ejogrb.2013.06.043

PubMed Abstract | CrossRef Full Text | Google Scholar

Gillen, C., Gleichmann, M., Spreyer, P., and Müller, H. W. (1995). Differentially expressed genes after peripheral nerve injury. J. Neurosci. Res. 42, 159–171. doi: 10.1002/jnr.490420203

PubMed Abstract | CrossRef Full Text | Google Scholar

Glöckner, F., and Ohm, T. G. (2003). Hippocampal apolipoprotein D level depends on Braak stage and APOE genotype. Neuroscience 122, 103–110. doi: 10.1016/S0306-4522(03)00529-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Goessling, W., and Zucker, S. D. (2000). Role of apolipoprotein D in the transport of bilirubin in plasma. Am. J. Physiol. Gastrointest. Liver Physiol. 279, G356–365. doi: 10.1152/ajpgi.2000.279.2.G356

PubMed Abstract | CrossRef Full Text | Google Scholar

Gonzalez, L. O., Corte, M. D., Junquera, S., Bongera, M., Rodriguez, J. C., and Vizoso, F. J. (2007). Expression of androgen receptor and two androgen-induced proteins (apolipoprotein D and pepsinogen C) in ductal carcinoma in situ of the breast. Histopathology 50, 866–874. doi: 10.1111/j.1365-2559.2007.02687.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Gottsch, J. D., Bowers, A. L., Margulies, E. H., Seitzman, G. D., Kim, S. W., Saha, S., et al. (2003). Serial analysis of gene expression in the corneal endothelium of Fuchs' dystrophy. Invest. Ophthalmol. Vis. Sci. 44, 594–599. doi: 10.1167/iovs.02-0300

PubMed Abstract | CrossRef Full Text | Google Scholar

Greenberg, M. E., Li, X.-M., Gugiu, B. G., Gu, X., Qin, J., Salomon, R. G., et al. (2008). The lipid whisker model of the structure of oxidized cell membranes. J. Biol. Chem. 283, 2385–2396. doi: 10.1074/jbc.M707348200

PubMed Abstract | CrossRef Full Text | Google Scholar

Gutmann, D. H., Hedrick, N. M., Li, J., Nagarajan, R., Perry, A., and Watson, M. A. (2002). Comparative gene expression profile analysis of neurofibromatosis 1-associated and sporadic pilocytic astrocytomas. Cancer Res. 62, 2085–2091.

PubMed Abstract | Google Scholar

Guven, G., Vurgun, E., Bilgic, B., Hanagasi, H., Gurvit, H., Ozer, E., et al. (2019). Association between selected cholesterol-related gene polymorphisms and Alzheimer's disease in a Turkish cohort. Mol. Biol. Rep. 46, 1701–1707. doi: 10.1007/s11033-019-04619-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Haagensen, D. E. Jr., Mazoujian, G., Dilley, W. G., Pedersen, C. E., Kister, S. J., and Wells, S. A. Jr. (1979). Breast gross cystic disease fluid analysis. i. isolation and radioimmunoassay for a major component protein. JNCI 62, 239–247.

PubMed Abstract | Google Scholar

Haffner, S. M., Applebaum-Bowden, D., Wahl, P. W., Hoover, J. J., Warnick, G. R., Albers, J. J., et al. (1985). Epidemiological correlates of high density lipoprotein subfractions, apolipoproteins A-I, A-II, and D, and lecithin cholesterol acyltransferase. Effects of smoking, alcohol, and adiposity. Arteriosclerosis 5, 169–177. doi: 10.1161/01.ATV.5.2.169

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, R. E., Aspinall, J. O., Horsfall, D. J., Birrell, S. N., Bentel, J. M., Sutherland, R. L., et al. (1996). Expression of the androgen receptor and an androgen-responsive protein, apolipoprotein D, in human breast cancer. Br. J. Cancer 74, 1175–1180. doi: 10.1038/bjc.1996.513

PubMed Abstract | CrossRef Full Text | Google Scholar

Hall, R. E., Horsfall, D. J., Stahl, J., Vivekanandan, S., Ricciardelli, C., Stapleton, A. M. F., et al. (2004). Apolipoprotein-D: a novel cellular marker for HGPIN and prostate cancer. Prostate 58, 103–108. doi: 10.1002/pros.10343

PubMed Abstract | CrossRef Full Text | Google Scholar

Hansen, L., Gaster, M., Oakeley, E. J., Brusgaard, K., Damsgaard Nielsen, E.-M., Beck-Nielsen, H., et al. (2004). Expression profiling of insulin action in human myotubes: induction of inflammatory and pro-angiogenic pathways in relationship with glycogen synthesis and type 2 diabetes. Biochem. Biophys. Res. Commun. 323, 685–695. doi: 10.1016/j.bbrc.2004.08.146

PubMed Abstract | CrossRef Full Text | Google Scholar

Hansen, T., Hemmingsen, R. P., Wang, A. G., Olsen, L., Timm, S., Søeby, K., et al. (2006). Apolipoprotein D is associated with long-term outcome in patients with schizophrenia. Pharmacogenomics J. 6, 120–125. doi: 10.1038/sj.tpj.6500350

PubMed Abstract | CrossRef Full Text | Google Scholar

Hardardóttir, I., Sipe, J., Moser, A. H., Fielding, C. J., Feingold, K. R., and Grünfeld, C. (1997). LPS and cytokines regulate extra hepatic mRNA levels of apolipoproteins during the acute phase response in Syrian hamsters. Biochim. Biophys. Acta 1344, 210–220. doi: 10.1016/S0005-2760(96)00143-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Harding, C., Osundeko, O., Tetlow, L., Faragher, E. B., Howell, A., and Bundred, N. J. (2000). Hormonally-regulated proteins in breast secretions are markers of target organ sensitivity. Br. J. Cancer 82, 354–360. doi: 10.1054/bjoc.1999.0926

PubMed Abstract | CrossRef Full Text | Google Scholar

Hargreaves, D. F., Potten, C. S., Harding, C., Shaw, L. E., Morton, M. S., Roberts, S. A., et al. (1999). Two-week dietary soy supplementation has an estrogenic effect on normal premenopausal breast. J. Clin. Endocrinol. Metab. 84, 4017–4024. doi: 10.1210/jc.84.11.4017

PubMed Abstract | CrossRef Full Text | Google Scholar

Hatzirodos, N., Hummitzsch, K., Irving-Rodgers, H. F., and Rodgers, R. J. (2015). Transcriptome comparisons identify new cell markers for theca interna and granulosa cells from small and large antral ovarian follicles. PLoS ONE 10:e0119800. doi: 10.1371/journal.pone.0119800

PubMed Abstract | CrossRef Full Text | Google Scholar

He, X., Jittiwat, J., Kim, J.-H., Jenner, A. M., Farooqui, A. A., Patel, S. C., et al. (2009). Apolipoprotein D modulates F2-isoprostane and 7-ketocholesterol formation and has a neuroprotective effect on organotypic hippocampal cultures after kainate-induced excitotoxic injury. Neurosci. Lett. 455, 183–186. doi: 10.1016/j.neulet.2009.03.038

PubMed Abstract | CrossRef Full Text | Google Scholar

Helisalmi, S., Hiltunen, M., Vepsäläinen, S., Iivonen, S., Corder, E. H., Lehtovirta, M., et al. (2004). Genetic variation in apolipoprotein D and Alzheimer's disease. J. Neurol. 251, 951–957. doi: 10.1007/s00415-004-0470-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Henri, P., and Rumeau, D. (2021). Ectopic expression of human apolipoprotein D in Arabidopsis plants lacking chloroplastic lipocalin partially rescues sensitivity to drought and oxidative stress. Plant Physiol. Biochem. 158, 265–274. doi: 10.1016/j.plaphy.2020.11.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Herzig, K.-H., Leppäluoto, J., Jokelainen, J., Meugnier, E., Pesenti, S., Selänne, H., et al. (2017). Low level activity thresholds for changes in NMR biomarkers and genes in high risk subjects for Type 2 diabetes. Sci. Rep. 7:11267. doi: 10.1038/s41598-017-09753-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Hildebrand, M. S., de Silva, M. G., Klockars, T., Solares, C. A., Hirose, K., Smith, J. D., et al. (2005). Expression of the carrier protein apolipoprotein D in the mouse inner ear. Hear. Res. 200, 102–114. doi: 10.1016/j.heares.2004.08.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmquist, L. (1989). Separation of free and apolipoprotein D-associated human plasma lecithin: cholesterol acyltransferase. J. Biochem. Biophys. Methods 19, 93–103. doi: 10.1016/0165-022X(89)90054-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmquist, L. (1990). Identification and quantification of apolipoprotein D in normal human urine. Electrophoresis 11, 93–94. doi: 10.1002/elps.1150110119

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmquist, L., Fredrikson, S., and Vesterberg, O. (1996). A zone immunoelectrophoresis assay method for quantification of apolipoprotein D in human cerebrospinal fluid. J. Biochem. Biophys. Methods 33, 1–8. doi: 10.1016/0165-022X(95)00041-O

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmquist, L., and Vesterberg, O. (1991). Quantification of apolipoprotein D in human urine by zone immunoelectrophoresis assay: a methodological and clinical study. J. Biochem. Biophys. Methods 23, 315–327. doi: 10.1016/0165-022X(91)90007-J

PubMed Abstract | CrossRef Full Text | Google Scholar

Holmquist, L., Vesterberg, O., and Persson, B. (1993). Apolipoprotein D and alpha 1-microglobulin in human urine: effect of cadmium exposure. Int. Arch. Occup. Environ. Health 64, 469–472. doi: 10.1007/BF00381093

PubMed Abstract | CrossRef Full Text | Google Scholar

Holzfeind, P., Merschak, P., Dieplinger, H., and Redl, B. (1995). The human lacrimal gland synthesizes apolipoprotein D mRNA in addition to tear prealbumin mRNA, both species encoding members of the lipocalin superfamily. Exp. Eye Res. 61, 495–500. doi: 10.1016/S0014-4835(05)80145-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Hornig, N. C., Demiri, J., Rodens, P., Murga Penas, E. M., Caliebe, A., Eckstein, A. K., et al. (2019). Reduced androgen receptor expression in genital skin fibroblasts from patients with 45,X/46,XY mosaicism. J. Clin. Endocrinol. Metab. 104, 4630–4638. doi: 10.1210/jc.2019-00108

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, C., Zhou, Y., Liu, C., and Kang, Y. (2018). A novel scoring system for gastric cancer risk assessment based on the expression of three CLIP4 DNA methylation-associated genes. Int. J. Oncol. 53, 633–643. doi: 10.3892/ijo.2018.4433

PubMed Abstract | CrossRef Full Text | Google Scholar

Hu, C. Y., Ong, W. Y., Sundaram, R. K., Chan, C., and Patel, S. C. (2001). Immunocytochemical localization of apolipoprotein D in oligodendrocyte precursor-like cells, perivascular cells, and pericytes in the human cerebral cortex. J. Neurocytol. 30, 209–218. doi: 10.1023/A:1012797623620

PubMed Abstract | CrossRef Full Text | Google Scholar

Hueging, K., Weller, R., Doepke, M., Vieyres, G., Todt, D., Wölk, B., et al. (2015). Several human liver cell expressed apolipoproteins complement HCV virus production with varying efficacy conferring differential specific infectivity to released viruses. PLoS ONE 10:e0134529. doi: 10.1371/journal.pone.0134529

PubMed Abstract | CrossRef Full Text | Google Scholar

Hull-Thompson, J., Muffat, J., Sanchez, D., Walker, D. W., Benzer, S., Ganfornina, M. D., et al. (2009). Control of metabolic homeostasis by stress signaling is mediated by the lipocalin NLaz. PLoS Genet. 5:e1000460. doi: 10.1371/journal.pgen.1000460

PubMed Abstract | CrossRef Full Text | Google Scholar

Hummasti, S., Laffitte, B. A., Watson, M. A., Galardi, C., Chao, L. C., Ramamurthy, L., et al. (2004). Liver X receptors are regulators of adipocyte gene expression but not differentiation: identification of apoD as a direct target. J. Lipid Res. 45, 616–625. doi: 10.1194/jlr.M300312-JLR200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hunter, S., Weiss, S., Ou, C.-Y., Jaye, D., Young, A., Wilcox, J., et al. (2005b). Apolipoprotein D is down-regulated during malignant transformation of neurofibromas. Hum. Pathol. 36, 987–993. doi: 10.1016/j.humpath.2005.06.018

PubMed Abstract | CrossRef Full Text | Google Scholar

Hunter, S., Young, A., Olson, J., Brat, D. J., Bowers, G., Wilcox, J. N., et al. (2002). Differential expression between pilocytic and anaplastic astrocytomas: identification of apolipoprotein D as a marker for low-grade, non-infiltrating primary CNS neoplasms. J. Neuropathol. Exp. Neurol. 61, 275–281. doi: 10.1093/jnen/61.3.275

PubMed Abstract | CrossRef Full Text | Google Scholar

Hunter, S. B., Varma, V., Shehata, B., Nolen, J. D. L., Cohen, C., Olson, J. J., et al. (2005a). Apolipoprotein D expression in primary brain tumors: analysis by quantitative RT-PCR in formalin-fixed, paraffin-embedded tissue. J. Histochem. Cytochem. 53, 963–969. doi: 10.1369/jhc.4A6530.2005

PubMed Abstract | CrossRef Full Text | Google Scholar

Igarashi, M., Masunaga, Y., Hasegawa, Y., Kinjo, K., Miyado, M., Saitsu, H., et al. (2020). Nonsense-associated altered splicing of MAP3K1 in two siblings with 46,XY disorders of sex development. Sci. Rep. 10:17375. doi: 10.1038/s41598-020-74405-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Iqbal, M. J., Higginbotham, A., Chickris, N., Bollaert, M., Rockway, S., and Banz, W. J. (2008). A combination of CLA-DAG oil modifies the diabetic phenotype in male Zucker diabetic fatty rats. Horm. Metab. Res. 40, 262–268. doi: 10.1055/s-2008-1058063

PubMed Abstract | CrossRef Full Text | Google Scholar

Ishii, M., Koike, C., Igarashi, A., Yamanaka, K., Pan, H., Higashi, Y., et al. (2005). Molecular markers distinguish bone marrow mesenchymal stem cells from fibroblasts. Biochem. Biophys. Res. Commun. 332, 297–303. doi: 10.1016/j.bbrc.2005.04.118

PubMed Abstract | CrossRef Full Text | Google Scholar

James, R. W., Martin, B., Pometta, D., Grab, B., and Suenram, A. (1986). Apoprotein D in a healthy, male population and in male myocardial infarction patients and their male, first-degree relatives. Atherosclerosis 60, 49–53. doi: 10.1016/0021-9150(86)90086-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Jankovic-Karasoulos, T., Bianco-Miotto, T., Butler, M. S., Butler, L. M., McNeil, C. M., O'Toole, S. A., et al. (2020). Elevated levels of tumour apolipoprotein D independently predict poor outcome in breast cancer patients. Histopathology 76, 976–987. doi: 10.1111/his.14081

PubMed Abstract | CrossRef Full Text | Google Scholar

Jansen, P. J., Lütjohann, D., Thelen, K. M., von Bergmann, K., van Leuven, F., Ramaekers, F. C. S., et al. (2009). Absence of ApoE upregulates murine brain ApoD and ABCA1 levels, but does not affect brain sterol levels, while human ApoE3 and human ApoE4 upregulate brain cholesterol precursor levels. J. Alzheimers Dis. 18, 319–329. doi: 10.3233/JAD-2009-1150

PubMed Abstract | CrossRef Full Text | Google Scholar

Jensen, L. D., Hot, B., Ramsköld, D., Germano, R. F. V., Yokota, C., Giatrellis, S., et al. (2019). Disruption of the extracellular matrix progressively impairs central nervous system vascular maturation downstream of β-catenin signaling. Arterioscler. Thromb. Vasc. Biol. 39, 1432–1447. doi: 10.1161/ATVBAHA.119.312388

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeong, J., Bae, H., Lim, W., Bazer, F. W., and Song, G. (2015). Diethylstilbestrol regulates expression of avian apolipoprotein D during regression and recrudescence of the oviduct and epithelial-derived ovarian carcinogenesis. Domest. Anim. Endocrinol. 52, 82–89. doi: 10.1016/j.domaniend.2015.03.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, D., Jin, H., Zuo, J., Kong, Y., Zhang, X., Dong, Q., et al. (2020). Potential biomarkers screening to predict side effects of dexamethasone in different cancers. Mol. Genet. Genomic Med. 8:e1160. doi: 10.1002/mgg3.1160

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiang, R., Rong, C., Ke, R., Meng, S., Yan, X., Ke, H., et al. (2019). Differential proteomic analysis of serum exosomes reveals alterations in progression of Parkinson disease. Medicine 98:e17478. doi: 10.1097/MD.0000000000017478

PubMed Abstract | CrossRef Full Text | Google Scholar

Jiménez-Palomares, M., Cózar-Castellano, I., Ganfornina, M. D., Sánchez, D., and Perdomo, G. (2011). Genetic deficiency of apolipoprotein D in the mouse is associated with nonfasting hypertriglyceridemia and hyperinsulinemia. Metab. Clin. Exp. 60, 1767–1774. doi: 10.1016/j.metabol.2011.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, D., El-Tanani, M., and Campbell, F. C. (2006). Identification of apolipoprotein D as a novel inhibitor of osteopontin-induced neoplastic transformation. Int. J. Oncol. 29, 1591–1599. doi: 10.3892/ijo.29.6.1591

PubMed Abstract | CrossRef Full Text | Google Scholar

Jinawath, N., Vasoontara, C., Jinawath, A., Fang, X., Zhao, K., Yap, K.-L., et al. (2010). Oncoproteomic analysis reveals co-upregulation of RELA and STAT5 in carboplatin resistant ovarian carcinoma. PLoS ONE 5:e11198. doi: 10.1371/journal.pone.0011198

PubMed Abstract | CrossRef Full Text | Google Scholar

Kalman, J., McConathy, W., Araoz, C., Kasa, P., and Lacko, A. G. (2000). Apolipoprotein D in the aging brain and in Alzheimer's dementia. Neurol. Res. 22, 330–336. doi: 10.1080/01616412.2000.11740678

PubMed Abstract | CrossRef Full Text | Google Scholar

Kamboh, M. I., Albers, J. J., Majumder, P. P., and Ferrell, R. E. (1989). Genetic studies of human apolipoproteins. IX. Apolipoprotein D polymorphism and its relation to serum lipoprotein lipid levels. Am. J. Hum. Genet. 45, 147–154.

PubMed Abstract | Google Scholar

Kang, M. K., Kameta, A., Shin, K.-H., Baluda, M. A., Kim, H.-R., and Park, N.-H. (2003a). Senescence-associated genes in normal human oral keratinocytes. Exp. Cell Res. 287, 272–281. doi: 10.1016/s0014-4827(03)00061-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Kang, S., Seo, S., Hill, J., Kwon, B., Lee, H., Cho, H., et al. (2003b). Changes in gene expression in latent HSV-1-infected rabbit trigeminal ganglia following epinephrine iontophoresis. Curr. Eye Res. 26, 225–229. doi: 10.1076/ceyr.26.3.225.14894

PubMed Abstract | CrossRef Full Text | Google Scholar

Kao, L. C., Tulac, S., Lobo, S., Imani, B., Yang, J. P., Germeyer, A., et al. (2002). Global gene profiling in human endometrium during the window of implantation. Endocrinology 143, 2119–2138. doi: 10.1210/endo.143.6.8885

PubMed Abstract | CrossRef Full Text | Google Scholar

Kendal, A. R., Layton, T., Al-Mossawi, H., Appleton, L., Dakin, S., Brown, R., et al. (2020). Multi-omic single cell analysis resolves novel stromal cell populations in healthy and diseased human tendon. Sci. Rep. 10:13939. doi: 10.1038/s41598-020-70786-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Kfir, S., Basavaraja, R., Wigoda, N., Ben-Dor, S., Orr, I., and Meidan, R. (2018). Genomic profiling of bovine corpus luteum maturation. PLoS ONE 13:e0194456. doi: 10.1371/journal.pone.0194456

PubMed Abstract | CrossRef Full Text | Google Scholar

Khan, M. M., Parikh, V. V., and Mahadik, S. P. (2003). Antipsychotic drugs differentially modulate apolipoprotein D in rat brain. J. Neurochem. 86, 1089–1100. doi: 10.1046/j.1471-4159.2003.01866.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Khoder-Agha, F., and Kietzmann, T. (2021). The glyco-redox interplay: principles and consequences on the role of reactive oxygen species during protein glycosylation. Redox Biol. 42:101888. doi: 10.1016/j.redox.2021.101888

PubMed Abstract | CrossRef Full Text | Google Scholar

Khoonsari, P. E., Ossipova, E., Lengqvist, J., Svensson, C. I., Kosek, E., Kadetoff, D., et al. (2019). The human CSF pain proteome. J. Proteomics 190, 67–76. doi: 10.1016/j.jprot.2018.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Kielkopf, C. S., Ghosh, M., Anand, G. S., and Brown, S. H. J. (2019). HDX-MS reveals orthosteric and allosteric changes in apolipoprotein-D structural dynamics upon binding of progesterone. Protein Sci. 28, 365–374. doi: 10.1002/pro.3534

PubMed Abstract | CrossRef Full Text | Google Scholar

Kielkopf, C. S., Low, J. K. K., Mok, Y.-F., Bhatia, S., Palasovski, T., Oakley, A. J., et al. (2018). Identification of a novel tetrameric structure for human apolipoprotein-D. J. Struct. Biol. 203, 205–218. doi: 10.1016/j.jsb.2018.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Kielkopf, C. S., Whitten, A. E., Garner, B., and Brown, S. H. J. (2021). Small angle X-ray scattering analysis of ligand-bound forms of tetrameric apolipoprotein-D. Biosci. Rep. 41:BSR20201423. doi: 10.1042/BSR20201423

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D. S., Lee, S. J., Park, S. Y., Yoo, H. J., Kim, S. H., Kim, K. J., et al. (2001). Differentially expressed genes in rat dorsal root ganglia following peripheral nerve injury. Neuroreport 12, 3401–3405. doi: 10.1097/00001756-200110290-00050

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, W. S., Wong, J., Weickert, C. S., Webster, M. J., Bahn, S., and Garner, B. (2009). Apolipoprotein-D expression is increased during development and maturation of the human prefrontal cortex. J. Neurochem. 109, 1053–1066. doi: 10.1111/j.1471-4159.2009.06031.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Klebaner, D., Hamilton-Dutoit, S., Ahern, T., Crawford, A., Jakobsen, T., Cronin-Fenton, D. P., et al. (2017). Apolipoprotein D expression does not predict breast cancer recurrence among tamoxifen-treated patients. PLoS ONE 12:e0171453. doi: 10.1371/journal.pone.0171453

PubMed Abstract | CrossRef Full Text | Google Scholar

Kliuchnikova, A. A., Samokhina, N. I., Ilina, I. Y., Karpov, D. S., Pyatnitskiy, M. A., Kuznetsova, K. G., et al. (2016). Human aqueous humor proteome in cataract, glaucoma, and pseudoexfoliation syndrome. Proteomics 16, 1938–1946. doi: 10.1002/pmic.201500423

PubMed Abstract | CrossRef Full Text | Google Scholar

Koch, S., Donarski, N., Goetze, K., Kreckel, M., Stuerenburg, H. J., Buhmann, C., et al. (2001). Characterization of four lipoprotein classes in human cerebrospinal fluid. J. Lipid Res. 42, 1143–1151. doi: 10.1016/S0022-2275(20)31605-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Kopylov, A. T., Papysheva, O., Gribova, I., Kotaysch, G., Kharitonova, L., Mayatskaya, T., et al. (2020). Molecular pathophysiology of diabetes mellitus during pregnancy with antenatal complications. Sci. Rep. 10:19641. doi: 10.1038/s41598-020-76689-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Kosacka, J., Gericke, M., Nowicki, M., Kacza, J., Borlak, J., and Spanel-Borowski, K. (2009). Apolipoproteins D and E3 exert neurotrophic and synaptogenic effects in dorsal root ganglion cell cultures. Neuroscience 162, 282–291. doi: 10.1016/j.neuroscience.2009.04.073

PubMed Abstract | CrossRef Full Text | Google Scholar

Kosacka, J., Schröder, T., Bechmann, I., Klöting, N., Nowicki, M., Mittag, A., et al. (2011). PACAP up-regulates the expression of apolipoprotein D in 3T3-L1 adipocytes. DRG/3T3-L1 co-cultures study. Neurosci. Res. 69, 8–16. doi: 10.1016/j.neures.2010.09.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Kosarussavadi, S., Pennington, Z. T., Covell, J., Blaisdell, A. P., and Schlinger, B. A. (2017). Across sex and age: learning and memory and patterns of avian hippocampal gene expression. Behav. Neurosci. 131, 483–491. doi: 10.1037/bne0000222

PubMed Abstract | CrossRef Full Text | Google Scholar

Kristensen, V. N., Sørlie, T., Geisler, J., Yoshimura, N., Linegjaerde, O.-C., Glad, I., et al. (2005). Effects of anastrozole on the intratumoral gene expression in locally advanced breast cancer. J. Steroid Biochem. Mol. Biol. 95, 105–111. doi: 10.1016/j.jsbmb.2005.04.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Kroksveen, A. C., Aasebø, E., Vethe, H., Van Pesch, V., Franciotta, D., Teunissen, C. E., et al. (2013). Discovery and initial verification of differentially abundant proteins between multiple sclerosis patients and controls using iTRAQ and SID-SRM. J. Proteomics 78, 312–325. doi: 10.1016/j.jprot.2012.09.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Kroksveen, A. C., Guldbrandsen, A., Vedeler, C., Myhr, K. M., Opsahl, J. A., and Berven, F. S. (2012). Cerebrospinal fluid proteome comparison between multiple sclerosis patients and controls. Acta Neurol. Scand., Suppl. 126, 90–96. doi: 10.1111/ane.12029

PubMed Abstract | CrossRef Full Text | Google Scholar

Kuiperij, H. B., Hondius, D. C., Kersten, I., Versleijen, A. A. M., Rozemuller, A. J. M., Greenberg, S. M., et al. (2019). Apolipoprotein D: a potential biomarker for cerebral amyloid angiopathy. Neuropathol. Appl. Neurobiol. 46, 431–440. doi: 10.1111/nan.12595

PubMed Abstract | CrossRef Full Text | Google Scholar

Kumar, U. (2012). Immunohistochemical distribution of somatostatin and somatostatin receptor subtypes (SSTR1–5) in hypothalamus of ApoD knockout mice brain. J. Mol. Neurosci. 48, 684–695. doi: 10.1007/s12031-012-9792-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Labrie, M., Lalonde, S., Najyb, O., Thiery, M., Daneault, C., Des Rosiers, C., et al. (2015). Apolipoprotein D transgenic mice develop hepatic steatosis through activation of PPARγ and fatty acid uptake. PLoS ONE 10:e0130230. doi: 10.1371/journal.pone.0130230

PubMed Abstract | CrossRef Full Text | Google Scholar

Lai, C.-J., Cheng, H.-C., Lin, C.-Y., Huang, S.-H., Chen, T.-H., Chung, C.-J., et al. (2017). Activation of liver X receptor suppresses angiogenesis via induction of ApoD. FASEB J. 31, 5568–5576. doi: 10.1096/fj.201700374R

PubMed Abstract | CrossRef Full Text | Google Scholar

Lambert, C., Cubedo, J., Padró, T., Sánchez-Hernández, J., Antonijoan, R. M., Perez, A., et al. (2017). Phytosterols and omega 3 supplementation exert novel regulatory effects on metabolic and inflammatory pathways: a proteomic study. Nutrients 9:599. doi: 10.3390/nu9060599

PubMed Abstract | CrossRef Full Text | Google Scholar

Lambert, J., Provost, P. R., Marcel, Y. L., and Rassart, E. (1993). Structure of the human apolipoprotein D gene promoter region. Biochim. Biophys. Acta 1172, 190–192. doi: 10.1016/0167-4781(93)90292-L

PubMed Abstract | CrossRef Full Text | Google Scholar

Lamelas, M. L., Vázquez, J., Enguita, M. I., Rodríguez, J. C., González, L. O., Merino, A. M., et al. (2000). Apolipoprotein D expression in metastasic lymph nodes of breast cancer. Int. J. Surg. Investig. 2, 285–293.

PubMed Abstract | Google Scholar

Lane, D. M., Boatman, K. K., and McConathy, W. J. (1995). Serum lipids and apolipoproteins in women with breast masses. Breast Cancer Res. Treat. 34, 161–169. doi: 10.1007/BF00665788

PubMed Abstract | CrossRef Full Text | Google Scholar

Lea, O. A. (1988). Binding properties of progesterone-binding Cyst protein, PBCP. Steroids 52, 337–338. doi: 10.1016/0039-128X(88)90135-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Lee, M. Y., Kim, E. Y., Kim, S. H., Cho, K.-C., Ha, K., Kim, K. P., et al. (2016). Discovery of serum protein biomarkers in drug-free patients with major depressive disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry 69, 60–68. doi: 10.1016/j.pnpbp.2016.04.009

PubMed Abstract | CrossRef Full Text | Google Scholar

Leung, W. C. Y., Lawrie, A., Demaries, S., Massaeli, H., Burry, A., Yablonsky, S., et al. (2004). Apolipoprotein D and platelet-derived growth factor-BB synergism mediates vascular smooth muscle cell migration. Circ. Res. 95, 179–186. doi: 10.1161/01.RES.0000135482.74178.14

PubMed Abstract | CrossRef Full Text | Google Scholar

Levros, L.-C., Do Carmo, S., Edouard, E., Legault, P., Charfi, C., and Rassart, E. (2010). Characterization of nuclear factors modulating the apolipoprotein D promoter during growth arrest: implication of PARP-1, APEX-1 and ERK1/2 catalytic activities. Biochim. Biophys. Acta 1803, 1062–1071. doi: 10.1016/j.bbamcr.2010.04.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Levros, L.-C., Labrie, M., Charfi, C., and Rassart, E. (2013). Binding and repressive activities of apolipoprotein E3 and E4 isoforms on the human ApoD promoter. Mol. Neurobiol. 48, 669–680. doi: 10.1007/s12035-013-8456-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Repa, J. J., Valasek, M. A., Beltroy, E. P., Turley, S. D., German, D. C., et al. (2005). Molecular, anatomical, and biochemical events associated with neurodegeneration in mice with Niemann-Pick type C disease. J. Neuropathol. Exp. Neurol. 64, 323–333. doi: 10.1093/jnen/64.4.323

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Ruberu, K., Karl, T., and Garner, B. (2016). Cerebral apolipoprotein-D is hypoglycosylated compared to peripheral tissues and is variably expressed in mouse and human brain regions. PLoS ONE 11:e0148238. doi: 10.1371/journal.pone.0148238

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, H., Ruberu, K., Muñoz, S. S., Jenner, A. M., Spiro, A., Zhao, H., et al. (2015). Apolipoprotein D modulates amyloid pathology in APP/PS1 Alzheimer's disease mice. Neurobiol. Aging 36, 1820–1833. doi: 10.1016/j.neurobiolaging.2015.02.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Liu, C., Chen, Y., Gao, C., Wang, M., Ma, X., et al. (2019). Tumor characterization in breast cancer identifies immune-relevant gene signatures associated with prognosis. Front. Genet. 10:1119. doi: 10.3389/fgene.2019.01119

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, X., Miyajima, M., Mineki, R., Taka, H., Murayama, K., and Arai, H. (2006). Analysis of potential diagnostic biomarkers in cerebrospinal fluid of idiopathic normal pressure hydrocephalus by proteomics. Acta Neurochir. 148, 859–864; discussion: 864. doi: 10.1007/s00701-006-0787-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Lieuallen, K., Pennacchio, L. A., Park, M., Myers, R. M., and Lennon, G. G. (2001). Cystatin B-deficient mice have increased expression of apoptosis and glial activation genes. Hum. Mol. Genet. 10, 1867–1871. doi: 10.1093/hmg/10.18.1867

PubMed Abstract | CrossRef Full Text | Google Scholar

Lim, W., Bae, H., and Song, G. (2016). Differential expression of apolipoprotein D in male reproductive system of rats by high-fat diet. Andrology 4, 1115–1122. doi: 10.1111/andr.12250

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, C. S., Ho, H. C., Gholami, S., Chen, K. C., Jad, A., and Lue, T. F. (2001). Gene expression profiling of an arteriogenic impotence model. Biochem. Biophys. Res. Commun. 285, 565–569. doi: 10.1006/bbrc.2001.5191

PubMed Abstract | CrossRef Full Text | Google Scholar

Lin, Y.-S., Chang, T.-H., Shi, C.-S., Wang, Y.-Z., Ho, W.-C., Huang, H.-D., et al. (2019). Liver X receptor/retinoid X receptor pathway plays a regulatory role in pacing-induced cardiomyopathy. J. Am. Heart Assoc. 8:e009146. doi: 10.1161/JAHA.118.009146

PubMed Abstract | CrossRef Full Text | Google Scholar

Linn, S. C., West, R. B., Pollack, J. R., Zhu, S., Hernandez-Boussard, T., Nielsen, T. O., et al. (2003). Gene expression patterns and gene copy number changes in dermatofibrosarcoma protuberans. Am. J. Pathol. 163, 2383–2395. doi: 10.1016/S0002-9440(10)63593-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Lisovsky, M., Hoang, M. P., Dresser, K. A., Kapur, P., Bhawan, J., and Mahalingam, M. (2008). Apolipoprotein D in CD34-positive and CD34-negative cutaneous neoplasms: a useful marker in differentiating superficial acral fibromyxoma from dermatofibrosarcoma protuberans. Mod. Pathol. 21, 31–38. doi: 10.1038/modpathol.3800971

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, H., Anders, F., Funke, S., Mercieca, K., Grus, F., and Prokosch, V. (2020). Proteome alterations in aqueous humour of primary open angle glaucoma patients. Int. J. Ophthalmol. 13, 176–179. doi: 10.18240/ijo.2020.01.24

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Qiu, N., and Ma, M. (2013). Comparative proteomic analysis of hen egg white proteins during early phase of embryonic development by combinatorial peptide ligand library and matrix-assisted laser desorption ionization-time of flight. Poult. Sci. 92, 1897–1904. doi: 10.3382/ps.2012-02986

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Z., Chang, G. Q., and Leibowitz, S. F. (2001). Apolipoprotein D interacts with the long-form leptin receptor: a hypothalamic function in the control of energy homeostasis. FASEB J. 15, 1329–1331. doi: 10.1096/fj.00-0530fje

PubMed Abstract | CrossRef Full Text | Google Scholar

Loerch, P. M., Lu, T., Dakin, K. A., Vann, J. M., Isaacs, A., Geula, C., et al. (2008). Evolution of the aging brain transcriptome and synaptic regulation. PLoS ONE 3:e3329. doi: 10.1371/journal.pone.0003329

PubMed Abstract | CrossRef Full Text | Google Scholar

López-Boado, Y. S., Klaus, M., Dawson, M. I., and López-Otín, C. (1996). Retinoic acid-induced expression of apolipoprotein D and concomitant growth arrest in human breast cancer cells are mediated through a retinoic acid receptor RARalpha-dependent signaling pathway. J. Biol. Chem. 271, 32105–32111. doi: 10.1074/jbc.271.50.32105

PubMed Abstract | CrossRef Full Text | Google Scholar

López-Boado, Y. S., Puente, X. S., Alvarez, S., Tolivia, J., Binderup, L., and López-Otín, C. (1997). Growth inhibition of human breast cancer cells by 1,25-dihydroxyvitamin D3 is accompanied by induction of apolipoprotein D expression. Cancer Res. 57, 4091–4097.

PubMed Abstract | Google Scholar

López-Boado, Y. S., Tolivia, J., and López-Otín, C. (1994). Apolipoprotein D gene induction by retinoic acid is concomitant with growth arrest and cell differentiation in human breast cancer cells. J. Biol. Chem. 269, 26871–26878. doi: 10.1016/S0021-9258(18)47100-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Lopez-Nunez, O., Surrey, L. F., Alaggio, R., Herradura, A., McGough, R. L., and John, I. (2021). Novel APOD-GLI1 rearrangement in a sarcoma of unknown lineage. Histopathology 78, 338–340. doi: 10.1111/his.14235

PubMed Abstract | CrossRef Full Text | Google Scholar

Lövkvist, H., Jönsson, A.-C., Luthman, H., Jood, K., Jern, C., Wieloch, T., et al. (2014). Variations in apolipoprotein D and sigma non-opioid intracellular receptor 1 genes with relation to risk, severity and outcome of ischemic stroke. BMC Neurol. 14:191. doi: 10.1186/s12883-014-0191-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Lynch, C. C., Hikosaka, A., Acuff, H. B., Martin, M. D., Kawai, N., Singh, R. K., et al. (2005). MMP-7 promotes prostate cancer-induced osteolysis via the solubilization of RANKL. Cancer Cell 7, 485–496. doi: 10.1016/j.ccr.2005.04.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahadevan, V., and Soloff, L. A. (1983). A method for isolating human plasma lecithin:cholesterol acyltransferase without using anti-apolipoprotein D, and its characterization. Biochim. Biophys. Acta 752, 89–97. doi: 10.1016/0005-2760(83)90236-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Mahadik, S. P., Khan, M. M., Evans, D. R., and Parikh, V. V. (2002). Elevated plasma level of apolipoprotein D in schizophrenia and its treatment and outcome. Schizophr. Res. 58, 55–62. doi: 10.1016/S0920-9964(01)00378-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Manjunatha, S., Distelmaier, K., Dasari, S., Carter, R. E., Kudva, Y. C., and Nair, K. S. (2016). Functional and proteomic alterations of plasma high density lipoproteins in type 1 diabetes mellitus. Metab. Clin. Exp. 65, 1421–1431. doi: 10.1016/j.metabol.2016.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Manousaki, T., Hull, P. M., Kusche, H., Machado-Schiaffino, G., Franchini, P., Harrod, C., et al. (2013). Parsing parallel evolution: ecological divergence and differential gene expression in the adaptive radiations of thick-lipped Midas cichlid fishes from Nicaragua. Mol. Ecol. 22, 650–669. doi: 10.1111/mec.12034

PubMed Abstract | CrossRef Full Text | Google Scholar

Martineau, C., Najyb, O., Signor, C., Rassart, É., and Moreau, R. (2016). Apolipoprotein D deficiency is associated to high bone turnover, low bone mass and impaired osteoblastic function in aged female mice. Metab. Clin. Exp. 65, 1247–1258. doi: 10.1016/j.metabol.2016.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez, E., Navarro, A., Ordóñez, C., Del Valle, E., and Tolivia, J. (2012). Amyloid-β25-35 induces apolipoprotein D Synthesis and growth arrest in HT22 hippocampal cells. J. Alzheimers Dis. 30, 233–244. doi: 10.3233/JAD-2012-112102

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez, E., Navarro, A., Ordóñez, C., Del Valle, E., and Tolivia, J. (2013). Oxidative stress induces apolipoprotein D overexpression in hippocampus during aging and Alzheimer's disease. J. Alzheimers Dis. 36, 129–144. doi: 10.3233/JAD-130215

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez-Pinilla, E., Navarro, A., Ordóñez, C., del Valle, E., and Tolivia, J. (2015). Apolipoprotein D subcellular distribution pattern in neuronal cells during oxidative stress. Acta Histochem. 117, 536–544. doi: 10.1016/j.acthis.2015.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Martínez-Pinilla, E., Rubio-Sardón, N., Peláez, R., García-Álvarez, E., del Valle, E., Tolivia, J., et al. (2021). Neuroprotective effect of apolipoprotein D in cuprizone-induced cell line models: a potential therapeutic approach for multiple sclerosis and demyelinating diseases. Int. J. Mol. Sci. 22:1260. doi: 10.3390/ijms22031260

PubMed Abstract | CrossRef Full Text | Google Scholar

Mattsson, N., Insel, P., Nosheny, R., Trojanowski, J. Q., Shaw, L. M., Jack, C. R., et al. (2014). Effects of cerebrospinal fluid proteins on brain atrophy rates in cognitively healthy older adults. Neurobiol. Aging 35, 614–622. doi: 10.1016/j.neurobiolaging.2013.08.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Mazoujian, G., and Haagensen, D. E. (1990). The immunopathology of gross cystic disease fluid proteins. Ann. N. Y. Acad. Sci. 586, 188–197. doi: 10.1111/j.1749-6632.1990.tb17806.x

PubMed Abstract | CrossRef Full Text | Google Scholar

McConathy, W. J., and Alaupovic, P. (1973). Isolation and partial characterization of apolipoprotein D: a new protein moiety of the human plasma lipoprotein system. FEBS Lett. 37, 178–182. doi: 10.1016/0014-5793(73)80453-3

PubMed Abstract | CrossRef Full Text | Google Scholar

McConathy, W. J., and Alaupovic, P. (1976). Studies on the isolation and partial characterization of apolipoprotein D and lipoprotein D of human plasma. Biochemistry 15, 515–520. doi: 10.1021/bi00648a010

PubMed Abstract | CrossRef Full Text | Google Scholar

McConathy, W. J., and Alaupovic, P. (1986). Isolation and characterization of other apolipoproteins. Meth. Enzymol. 128, 297–310. doi: 10.1016/0076-6879(86)28075-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Mejias, A., Diez-Hermano, S., Ganfornina, M. D., Gutierrez, G., and Sanchez, D. (2019). Characterization of mammalian Lipocalin UTRs in silico: predictions for their role in post-transcriptional regulation. PLoS ONE 14:e0213206. doi: 10.1371/journal.pone.0213206

PubMed Abstract | CrossRef Full Text | Google Scholar

Miranda, E., Vizoso, F., Martín, A., Quintela, I., Corte, M. D., Seguí, M. E., et al. (2003). Apolipoprotein D expression in cutaneous malignant melanoma. J. Surg. Oncol. 83, 99–105. doi: 10.1002/jso.10245

PubMed Abstract | CrossRef Full Text | Google Scholar

Mirza, Z., and Abdel-Dayem, U. A. (2020). Uncovering potential roles of differentially expressed genes, upstream regulators, and canonical pathways in endometriosis using an in silico genomics approach. Diagnostics 10:416. doi: 10.3390/diagnostics10060416

PubMed Abstract | CrossRef Full Text | Google Scholar

Mohammed, A., Janakiram, N. B., Suen, C., Stratton, N., Lightfoot, S., Singh, A., et al. (2019). Targeting cholecystokinin-2 receptor for pancreatic cancer chemoprevention. Mol. Carcinog. 58, 1908–1918. doi: 10.1002/mc.23084

PubMed Abstract | CrossRef Full Text | Google Scholar

Molnár, A., Gyurján, I., Korpos, E., Borsy, A., Stéger, V., Buzás, Z., et al. (2007). Identification of differentially expressed genes in the developing antler of red deer Cervus elaphus. Mol. Genet. Genomics 277, 237–248. doi: 10.1007/s00438-006-0193-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Montpied, P., de Bock, F., Lerner-Natoli, M., Bockaert, J., and Rondouin, G. (1999). Hippocampal alterations of apolipoprotein E and D mRNA levels in vivo and in vitro following kainate excitotoxicity. Epilepsy Res. 35, 135–146. doi: 10.1016/S0920-1211(99)00003-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Morais Cabral, J. H., Atkins, G. L., Sánchez, L. M., López-Boado, Y. S., López-Otin, C., and Sawyer, L. (1995). Arachidonic acid binds to apolipoprotein D: implications for the protein's function. FEBS Lett. 366, 53–56. doi: 10.1016/0014-5793(95)00484-Q

PubMed Abstract | CrossRef Full Text | Google Scholar

Morton, R. E., and Zilversmit, D. B. (1981). The separation of apolipoprotein D from cholesteryl ester transfer protein. Biochim. Biophys. Acta 663, 350–355. doi: 10.1016/0005-2760(81)90220-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Muffat, J., Walker, D. W., and Benzer, S. (2008). Human ApoD, an apolipoprotein up-regulated in neurodegenerative diseases, extends lifespan and increases stress resistance in Drosophila. Proc. Natl. Acad. Sci. U.S.A. 105, 7088–7093. doi: 10.1073/pnas.0800896105

PubMed Abstract | CrossRef Full Text | Google Scholar

Najyb, O., Brissette, L., and Rassart, E. (2015). Apolipoprotein D internalization is a basigin-dependent mechanism. J. Biol. Chem. 290, 16077–16087. doi: 10.1074/jbc.M115.644302

PubMed Abstract | CrossRef Full Text | Google Scholar

Najyb, O., Do Carmo, S., Alikashani, A., and Rassart, E. (2017). Apolipoprotein D overexpression protects against kainate-induced neurotoxicity in mice. Mol. Neurobiol. 54, 3948–3963. doi: 10.1007/s12035-016-9920-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Namdar-Aligoodarzi, P., Mohammadparast, S., Zaker-Kandjani, B., Talebi Kakroodi, S., Jafari Vesiehsari, M., and Ohadi, M. (2015). Exceptionally long 5' UTR short tandem repeats specifically linked to primates. Gene 569, 88–94. doi: 10.1016/j.gene.2015.05.053

PubMed Abstract | CrossRef Full Text | Google Scholar

Nasreen, A., Vogt, M., Kim, H. J., Eichinger, A., and Skerra, A. (2006). Solubility engineering and crystallization of human apolipoprotein D. Protein Sci. 15, 190–199. doi: 10.1110/ps.051775606

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Alonso, A., Garrido, P., González, C., González Del Rey, C., Ordoñez, C., et al. (2010a). Increase in placental apolipoprotein D as an adaptation to human gestational diabetes. Placenta 31, 25–31. doi: 10.1016/j.placenta.2009.11.002

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Del Valle, E., Astudillo, A., González del Rey, C., and Tolivia, J. (2003). Immunohistochemical study of distribution of apolipoproteins E and D in human cerebral beta amyloid deposits. Exp. Neurol. 184, 697–704. doi: 10.1016/S0014-4886(03)00315-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., del Valle, E., Juárez, A., Martinez, E., Ordóñez, C., Astudillo, A., et al. (2010b). Apolipoprotein D synthesis progressively increases in frontal cortex during human lifespan. Age 32, 85–96. doi: 10.1007/s11357-009-9117-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Del Valle, E., and Tolivia, J. (2004). Differential expression of apolipoprotein d in human astroglial and oligodendroglial cells. J. Histochem. Cytochem. 52, 1031–1036. doi: 10.1369/jhc.3A6213.2004

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Méndez, E., Diaz, C., del Valle, E., Martínez-Pinilla, E., Ordóñez, C., et al. (2013). Lifelong expression of apolipoprotein D in the human brainstem: correlation with reduced age-related neurodegeneration. PLoS ONE 8:e77852. doi: 10.1371/journal.pone.0077852

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Ordóñez, C., Martínez, E., Pérez, C., Astudillo, A., and Tolivia, J. (2008). Apolipoprotein D expression absence in degenerating neurons of human central nervous system. Histol. Histopathol. 23, 995–1001. doi: 10.14670/HH-23.995

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Rioseras, B., Del Valle, E., Martínez-Pinilla, E., Astudillo, A., and Tolivia, J. (2018). Expression Pattern of Myelin-Related Apolipoprotein D in Human Multiple Sclerosis Lesions. Front. Aging Neurosci. 10:254. doi: 10.3389/fnagi.2018.00254

PubMed Abstract | CrossRef Full Text | Google Scholar

Navarro, A., Tolivia, J., Astudillo, A., and del Valle, E. (1998). Pattern of apolipoprotein D immunoreactivity in human brain. Neurosci. Lett. 254, 17–20. doi: 10.1016/S0304-3940(98)00639-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Nazarenko, M. S., Markov, A. V., Sleptsov, A. A., Koroleva, I. A., Sharysh, D. V., Zarubin, A. A., et al. (2018). Comparative analysis of gene expression in vascular cells of patients with advanced atherosclerosis. Biomed. Khim. 64, 416–422. doi: 10.18097/PBMC20186405416

PubMed Abstract | CrossRef Full Text | Google Scholar

Ng, M. W., Angerosa, J., Konstantinov, I. E., Cheung, M. M., and Pepe, S. (2019). Remote ischaemic preconditioning modifies serum apolipoprotein D, met-enkephalin, adenosine, and nitric oxide in healthy young adults. Clin. Exp. Pharmacol. Physiol. 46, 995–1000. doi: 10.1111/1440-1681.13150

PubMed Abstract | CrossRef Full Text | Google Scholar

Nowicki, M., Kosacka, J., Spanel-Borowski, K., and Borlak, J. (2009). Deferoxamine-induced neurite outgrowth and synapse formation in postnatal rat dorsal root ganglion (DRG) cell cultures. Eur. J. Cell Biol. 88, 551–562. doi: 10.1016/j.ejcb.2009.05.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Oakley, A. J., Bhatia, S., Ecroyd, H., and Garner, B. (2012). Molecular dynamics analysis of apolipoprotein-D-lipid hydroperoxide interactions: mechanism for selective oxidation of Met-93. PLoS ONE 7:e34057. doi: 10.1371/journal.pone.0034057

PubMed Abstract | CrossRef Full Text | Google Scholar

O'Donnell, J., Stemmelin, J., Nitta, A., Brouillette, J., and Quirion, R. (2003). Gene expression profiling following chronic NMDA receptor blockade-induced learning deficits in rats. Synapse 50, 171–180. doi: 10.1002/syn.10258

PubMed Abstract | CrossRef Full Text | Google Scholar

Ogawa, K., Utsunomiya, T., Mimori, K., Yamashita, K., Okamoto, M., Tanaka, F., et al. (2005). Genomic screens for genes upregulated by demethylation in colorectal cancer: possible usefulness for clinical application. Int. J. Oncol. 27, 417–426. doi: 10.3892/ijo.27.2.417

PubMed Abstract | CrossRef Full Text | Google Scholar

Oláh, Z., Kálmán, J., Tóth, M. E., Zvara, Á., Sántha, M., Ivitz, E., et al. (2015). Proteomic analysis of cerebrospinal fluid in Alzheimer's disease: wanted dead or alive. J. Alzheimers Dis. 44, 1303–1312. doi: 10.3233/JAD-140141

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, W.-Y., Hu, C.-Y., and Patel, S. C. (2002). Apolipoprotein D in the Niemann-Pick type C disease mouse brain: an ultrastructural immunocytochemical analysis. J. Neurocytol. 31, 121–129. doi: 10.1023/A:1023993405851

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, W. Y., He, Y., Suresh, S., and Patel, S. C. (1997). Differential expression of apolipoprotein D and apolipoprotein E in the kainic acid-lesioned rat hippocampus. Neuroscience 79, 359–367. doi: 10.1016/S0306-4522(96)00608-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ong, W. Y., Lau, C. P., Leong, S. K., Kumar, U., Suresh, S., and Patel, S. C. (1999). Apolipoprotein D gene expression in the rat brain and light and electron microscopic immunocytochemistry of apolipoprotein D expression in the cerebellum of neonatal, immature and adult rats. Neuroscience 90, 913–922. doi: 10.1016/S0306-4522(98)00507-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Ordoñez, C., Navarro, A., Perez, C., Astudillo, A., Martínez, E., and Tolivia, J. (2006). Apolipoprotein D expression in substantia nigra of Parkinson disease. Histol. Histopathol. 21, 361–366. doi: 10.14670/HH-21.361

PubMed Abstract | CrossRef Full Text | Google Scholar

Ordóñez, C., Navarro, A., Pérez, C., Martínez, E., del Valle, E., and Tolivia, J. (2012). Gender differences in apolipoprotein D expression during aging and in Alzheimer disease. Neurobiol. Aging 33, 433.e11–e20. doi: 10.1016/j.neurobiolaging.2011.01.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Osundeko, O., Tetlow, L., Bundred, N., and Gowland, E. (1997). Radioimmunoassay for serum apolipoprotein D, an atypical apolipoprotein: validation and clinical application. Ann. Clin. Biochem. 34(Pt. 5), 537–542. doi: 10.1177/000456329703400508

PubMed Abstract | CrossRef Full Text | Google Scholar

Pajaniappan, M., Glober, N. K., Kennard, S., Liu, H., Zhao, N., and Lilly, B. (2011). Endothelial cells downregulate apolipoprotein D expression in mural cells through paracrine secretion and Notch signaling. Am. J. Physiol. Heart Circ. Physiol. 301, H784–H793. doi: 10.1152/ajpheart.00116.2011

PubMed Abstract | CrossRef Full Text | Google Scholar

Palmerini, E., Gambarotti, M., Staals, E. L., Zanella, L., Sieberova, G., Longhi, A., et al. (2012). Fibrosarcomatous changes and expression of CD34+ and apolipoprotein-D in dermatofibrosarcoma protuberans. Clin. Sarcoma Res. 2:4. doi: 10.1186/2045-3329-2-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Pamir, N., Hutchins, P., Ronsein, G., Vaisar, T., Reardon, C. A., Getz, G. S., et al. (2016). Proteomic analysis of HDL from inbred mouse strains implicates APOE associated with HDL in reduced cholesterol efflux capacity via the ABCA1 pathway. J. Lipid Res. 57, 246–257. doi: 10.1194/jlr.M063701

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascua-Maestro, R., Corraliza-Gomez, M., Fadrique-Rojo, C., Ledesma, M. D., Schuchman, E. H., Sanchez, D., et al. (2020). Apolipoprotein D-mediated preservation of lysosomal function promotes cell survival and delays motor impairment in Niemann-Pick type A disease. Neurobiol. Dis. 144:105046. doi: 10.1016/j.nbd.2020.105046

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascua-Maestro, R., Diez-Hermano, S., Lillo, C., Ganfornina, M. D., and Sanchez, D. (2017). Protecting cells by protecting their vulnerable lysosomes: identification of a new mechanism for preserving lysosomal functional integrity upon oxidative stress. PLoS Genet. 13:e1006603. doi: 10.1371/journal.pgen.1006603

PubMed Abstract | CrossRef Full Text | Google Scholar

Pascua-Maestro, R., González, E., Lillo, C., Ganfornina, M. D., Falcón-Pérez, J. M., and Sanchez, D. (2018). Extracellular vesicles secreted by astroglial cells transport apolipoprotein D to neurons and mediate neuronal survival upon oxidative stress. Front. Cell Neurosci. 12:526. doi: 10.3389/fncel.2018.00526

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, R. C., Lange, D., McConathy, W. J., Patel, Y. C., and Patel, S. C. (1997). Probing the structure of the ligand binding cavity of lipocalins by fluorescence spectroscopy. Protein Eng. 10, 621–625. doi: 10.1093/protein/10.6.621

PubMed Abstract | CrossRef Full Text | Google Scholar

Patel, S. C., Asotra, K., Patel, Y. C., McConathy, W. J., Patel, R. C., and Suresh, S. (1995). Astrocytes synthesize and secrete the lipophilic ligand carrier apolipoprotein D. Neuroreport 6, 653–657. doi: 10.1097/00001756-199503000-00017

PubMed Abstract | CrossRef Full Text | Google Scholar

Peitsch, M. C., and Boguski, M. S. (1990). Is apolipoprotein D a mammalian bilin-binding protein? New Biol. 2, 197–206.

PubMed Abstract | Google Scholar

Perdomo, G., and Henry Dong, H. (2009). Apolipoprotein D in lipid metabolism and its functional implication in atherosclerosis and aging. Aging 1, 17–27. doi: 10.18632/aging.100004

PubMed Abstract | CrossRef Full Text | Google Scholar

Perdomo, G., Kim, D. H., Zhang, T., Qu, S., Thomas, E. A., Toledo, F. G. S., et al. (2010). A role of apolipoprotein D in triglyceride metabolism. J. Lipid Res. 51, 1298–1311. doi: 10.1194/jlr.M001206

PubMed Abstract | CrossRef Full Text | Google Scholar

Pérez, C., Navarro, A., Martínez, E., Ordóñez, C., Del Valle, E., and Tolivia, J. (2012). Age-related changes of apolipoprotein D expression in female rat central nervous system with chronic estradiol treatment. Age 34, 895–904. doi: 10.1007/s11357-011-9286-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Perrotte, M., Le Page, A., Fournet, M., Le Sayec, M., Rassart, É., Fulop, T., et al. (2019). Blood-based redox-signature and their association to the cognitive scores in MCI and Alzheimer's disease patients. Free Radic. Biol. Med. 130, 499–511. doi: 10.1016/j.freeradbiomed.2018.10.452

PubMed Abstract | CrossRef Full Text | Google Scholar

Phillips, M. C. (2018). Is ABCA1 a lipid transfer protein? J. Lipid Res. 59, 749–763. doi: 10.1194/jlr.R082313

PubMed Abstract | CrossRef Full Text | Google Scholar

Piórkowska, K., Zukowski, K., Ropka-Molik, K., Tyra, M., and Gurgul, A. (2018). A comprehensive transcriptome analysis of skeletal muscles in two Polish pig breeds differing in fat and meat quality traits. Genet. Mol. Biol. 41, 125–136. doi: 10.1590/1678-4685-gmb-2016-0101

PubMed Abstract | CrossRef Full Text | Google Scholar

Pleva, L., Kusnierova, P., Plevova, P., Zapletalova, J., Karpisek, M., Faldynova, L., et al. (2015). Increased levels of MMP-3, MMP-9 and MPO represent predictors of in-stent restenosis, while increased levels of ADMA, LCAT, ApoE and ApoD predict bare metal stent patency. Biomed. Pap. Med. Fac. Univ. Palacky Olomouc Czech Repub. 159, 586–594. doi: 10.5507/bp.2015.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Ponnikorn, S., Mongkolrob, R., Klongthalay, S., Roytrakul, S., Srisanga, K., Tungpradabkul, S., et al. (2019). Comparative proteome-wide analysis of bone marrow microenvironment of β-thalassemia/hemoglobin E. Proteomes 7:8. doi: 10.3390/proteomes7010008

PubMed Abstract | CrossRef Full Text | Google Scholar

Provost, P. R., Marcel, Y. L., Milne, R. W., Weech, P. K., and Rassart, E. (1991a). Apolipoprotein D transcription occurs specifically in nonproliferating quiescent and senescent fibroblast cultures. FEBS Lett. 290, 139–141.

PubMed Abstract | Google Scholar

Provost, P. R., Tremblay, Y., el-Amine, M., and Bélanger, A. (1995). Guinea pig apolipoprotein D RNA diversity, and developmental and gestational modulation of mRNA levels. Mol. Cell. Endocrinol. 109, 225–236. doi: 10.1016/0303-7207(95)03506-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Provost, P. R., Villeneuve, L., Weech, P. K., Milne, R. W., Marcel, Y. L., and Rassart, E. (1991b). Localization of the major sites of rabbit apolipoprotein D gene transcription by in situ hybridization. J. Lipid Res. 32, 1959–1970.

PubMed Abstract | Google Scholar

Provost, P. R., Weech, P. K., Tremblay, N. M., Marcel, Y. L., and Rassart, E. (1990). Molecular characterization and differential mRNA tissue distribution of rabbit apolipoprotein D. J. Lipid Res. 31, 2057–2065. doi: 10.1016/S0022-2275(20)42270-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Przybycien-Szymanska, M. M., Yang, Y., and Ashley, W. W. (2016). Microparticle derived proteins as potential biomarkers for cerebral vasospasm post subarachnoid hemorrhage. A preliminary study. Clin. Neurol Neurosurg. 141, 48–55. doi: 10.1016/j.clineuro.2015.12.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Puntambekar, S. S., Davis, D. S., Hawel, L., Crane, J., Byus, C. V., and Carson, M. J. (2011). LPS-induced CCL2 expression and macrophage influx into the murine central nervous system is polyamine-dependent. Brain Behav. Immun. 25, 629–639. doi: 10.1016/j.bbi.2010.12.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, W., Pan, J., Bauman, W. A., and Cardozo, C. P. (2010). Differential alterations in gene expression profiles contribute to time-dependent effects of nandrolone to prevent denervation atrophy. BMC Genomics 11:596. doi: 10.1186/1471-2164-11-596

PubMed Abstract | CrossRef Full Text | Google Scholar

Qin, Y., Chen, Y., Yang, J., Wu, F., Zhao, L., Yang, F., et al. (2017). Serum glycopattern and Maackia amurensis lectin-II binding glycoproteins in autism spectrum disorder. Sci. Rep. 7:46041. doi: 10.1038/srep46041

PubMed Abstract | CrossRef Full Text | Google Scholar

Quaresima, B., Crugliano, T., Gaspari, M., Faniello, M. C., Cosimo, P., Valanzano, R., et al. (2008). A proteomics approach to identify changes in protein profiles in serum of Familial Adenomatous Polyposis patients. Cancer Lett. 272, 40–52. doi: 10.1016/j.canlet.2008.06.021

PubMed Abstract | CrossRef Full Text | Google Scholar

Rajput, P. S., Billova, S., Patel, S. C., Kharmate, G., Somvanshi, R. K., and Kumar, U. (2009). Expression of somatostatin and somatostatin receptor subtypes in Apolipoprotein D (ApoD) knockout mouse brain: an immunohistochemical analysis. J. Chem. Neuroanat. 38, 20–33. doi: 10.1016/j.jchemneu.2009.05.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Ravnsborg, T., Andersen, L. L. T., Trabjerg, N. D., Rasmussen, L. M., Jensen, D. M., and Overgaard, M. (2016). First-trimester multimarker prediction of gestational diabetes mellitus using targeted mass spectrometry. Diabetologia 59, 970–979. doi: 10.1007/s00125-016-3869-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Reindl, M., Knipping, G., Wicher, I., Dilitz, E., Egg, R., Deisenhammer, F., et al. (2001). Increased intrathecal production of apolipoprotein D in multiple sclerosis. J. Neuroimmunol. 119, 327–332. doi: 10.1016/S0165-5728(01)00378-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Ricci, F., Kern, S. E., Hruban, R. H., and Iacobuzio-Donahue, C. A. (2005). Stromal responses to carcinomas of the pancreas: juxtatumoral gene expression conforms to the infiltrating pattern and not the biologic subtype. Cancer Biol. Ther. 4, 302–307. doi: 10.4161/cbt.4.3.1501

PubMed Abstract | CrossRef Full Text | Google Scholar

Rickhag, M., Deierborg, T., Patel, S., Ruscher, K., and Wieloch, T. (2008). Apolipoprotein D is elevated in oligodendrocytes in the peri-infarct region after experimental stroke: influence of enriched environment. J. Cereb. Blood Flow Metab. 28, 551–562. doi: 10.1038/sj.jcbfm.9600552

PubMed Abstract | CrossRef Full Text | Google Scholar

Rickhag, M., Wieloch, T., Gid,ö, G., Elmér, E., Krogh, M., Murray, J., et al. (2006). Comprehensive regional and temporal gene expression profiling of the rat brain during the first 24 h after experimental stroke identifies dynamic ischemia-induced gene expression patterns, and reveals a biphasic activation of genes in surviving tissue. J. Neurochem. 96, 14–29. doi: 10.1111/j.1471-4159.2005.03508.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Rodríguez, J. C., Díaz, M., González, L. O., Sánchez, J., Sánchez, M. T., Merino, A. M., et al. (2000). Apolipoprotein D expression in benign and malignant prostate tissues. Int. J. Surg. Investig. 2, 319–326.

PubMed Abstract | Google Scholar

Rojo, J. V., González, L. O., Lamelas, M. L., Merino, A., and Vizoso, F. (2001). Apolipoprotein D expression in endometrial carcinomas. Acta Obstet. Gynecol. Scand. 80, 158–161. doi: 10.1034/j.1600-0412.2001.080002158.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruiz García, M. (2013). Lazarillo and related lipocalins: ligands and functions (Ph.D. thesis). Universidad de Valladolid, Valladolid, Spain.

Ruiz, M., Sanchez, D., Correnti, C., Strong, R. K., and Ganfornina, M. D. (2013). Lipid-binding properties of human ApoD and Lazarillo-related lipocalins: functional implications for cell differentiation. FEBS J. 280, 3928–3943. doi: 10.1111/febs.12394

PubMed Abstract | CrossRef Full Text | Google Scholar

Ruscher, K., Erickson, A., Kuric, E., Inácio, A. R., and Wieloch, T. (2010). Effects of chronic Clozapine administration on apolipoprotein D levels and on functional recovery following experimental stroke. Brain Res. 1321, 152–163. doi: 10.1016/j.brainres.2010.01.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Saadane, A., Mast, N., Trichonas, G., Chakraborty, D., Hammer, S., Busik, J. V., et al. (2019). Retinal vascular abnormalities and microglia activation in mice with deficiency in cytochrome P450 46A1-mediated cholesterol removal. Am. J. Pathol. 189, 405–425. doi: 10.1016/j.ajpath.2018.10.013

PubMed Abstract | CrossRef Full Text | Google Scholar

Saha, S., and Rangarajan, P. N. (2003). Common host genes are activated in mouse brain by Japanese encephalitis and rabies viruses. J. Gen. Virol. 84, 1729–1735. doi: 10.1099/vir.0.18826-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Salami, M., Bandegi, A. R., Sameni, H. R., Vafaei, A. A., and Pakdel, A. (2019). hippocampal up-regulation of apolipoprotein D in a rat model of maternal hypo- and hyperthyroidism: implication of oxidative stress. Neurochem. Res. 44, 2190–2201. doi: 10.1007/s11064-019-02859-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Sałkowska, A., Kara,ś, K., Karwaciak, I., Walczak-Drzewiecka, A., Krawczyk, M., Sobalska-Kwapis, M., et al. (2020). Identification of novel molecular markers of human Th17 cells. Cells 9:1611. doi: 10.3390/cells9071611

PubMed Abstract | CrossRef Full Text | Google Scholar

Salvatore, A., Cigliano, L., Bucci, E. M., Corpillo, D., Velasco, S., Carlucci, A., et al. (2007). Haptoglobin binding to apolipoprotein A-I prevents damage from hydroxyl radicals on its stimulatory activity of the enzyme lecithin-cholesterol acyl-transferase. Biochemistry 46, 11158–11168. doi: 10.1021/bi7006349

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez, D., Bajo-Grañeras, R., Del Caño-Espinel, M., Garcia-Centeno, R., Garcia-Mateo, N., Pascua-Maestro, R., et al. (2015). Aging without apolipoprotein D: molecular and cellular modifications in the hippocampus and cortex. Exp. Gerontol. 67, 19–47. doi: 10.1016/j.exger.2015.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Sanchez, D., Ganfornina, M. D., Gutiérrez, G., Gauthier-Jauneau, A.-C., Risler, J.-L., and Salier, J.-P. (2006). “Lipocalin genes and their evolutionary history,” in Molecular Biology Intelligence Unit: Lipocalins, 5–16. Available onnline at: https://uvadoc.uva.es/handle/10324/6219 (accessed June 29, 2021).

Google Scholar

Sánchez, D., Ganfornina, M. D., Gutiérrez, G., and Marín, A. (2003). Exon-intron structure and evolution of the lipocalin gene family. Mol. Biol. Evol. 20, 775–783. doi: 10.1093/molbev/msg079

PubMed Abstract | CrossRef Full Text | Google Scholar

Sánchez, D., Ganfornina, M. D., and Martínez, S. (2002). Expression pattern of the lipocalin apolipoprotein D during mouse embryogenesis. Mech. Dev. 110, 225–229. doi: 10.1016/S0925-4773(01)00578-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Sánchez, L. M., Díez-Itza, I., Vizoso, F., and López-Otín, C. (1992a). Cholesterol and apolipoprotein D in gross cystic disease of the breast. Clin. Chem. 38, 695–698.

PubMed Abstract | Google Scholar

Sánchez, L. M., Vizoso, F., Díez-Itza, I., and López-Otín, C. (1992b). Identification of the major protein components in breast secretions from women with benign and malignant breast diseases. Cancer Res. 52, 95–100.

PubMed Abstract | Google Scholar

Sandim, V., Pereira, D., de, A., Kalume, D. E., Oliveira-Carvalho, A. L., Ornellas, A. A., et al. (2016). Proteomic analysis reveals differentially secreted proteins in the urine from patients with clear cell renal cell carcinoma. Urol. Oncol. 34, 5.e11–e25. doi: 10.1016/j.urolonc.2015.07.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Santana, M. F. M., Lira, A. L. A., Pinto, R. S., Minanni, C. A., Silva, A. R. M., Sawada, M. I. B. A. C., et al. (2020). Enrichment of apolipoprotein A-IV and apolipoprotein D in the HDL proteome is associated with HDL functions in diabetic kidney disease without dialysis. Lipids Health Dis. 19:205. doi: 10.1186/s12944-020-01381-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Sarjeant, J. M., Lawrie, A., Kinnear, C., Yablonsky, S., Leung, W., Massaeli, H., et al. (2003). Apolipoprotein D inhibits platelet-derived growth factor-BB-induced vascular smooth muscle cell proliferated by preventing translocation of phosphorylated extracellular signal regulated kinase 1/2 to the nucleus. Arterioscler. Thromb. Vasc. Biol. 23, 2172–2177. doi: 10.1161/01.ATV.0000100404.05459.39

PubMed Abstract | CrossRef Full Text | Google Scholar

Sasaki, Y., Negishi, H., Koyama, R., Anbo, N., Ohori, K., Idogawa, M., et al. (2009). p53 family members regulate the expression of the apolipoprotein D gene. J. Biol. Chem. 284, 872–883. doi: 10.1074/jbc.M807185200

PubMed Abstract | CrossRef Full Text | Google Scholar

Satoh, F., Umemura, S., and Osamura, R. Y. (2000). Immunohistochemical analysis of GCDFP-15 and GCDFP-24 in mammary and non-mammary tissue. Breast Cancer 7, 49–55. doi: 10.1007/BF02967188

PubMed Abstract | CrossRef Full Text | Google Scholar

Scalf, C. S., Chariker, J. H., Rouchka, E. C., and Ashley, N. T. (2019). Transcriptomic analysis of immune response to bacterial lipopolysaccharide in zebra finch (Taeniopygia guttata). BMC Genomics 20:647. doi: 10.1186/s12864-019-6016-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Schaeren-Wiemers, N., Schaefer, C., Valenzuela, D. M., Yancopoulos, G. D., and Schwab, M. E. (1995). Identification of new oligodendrocyte- and myelin-specific genes by a differential screening approach. J. Neurochem. 65, 10–22. doi: 10.1046/j.1471-4159.1995.65010010.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Schäfer, N. F., Luhmann, U. F. O., Feil, S., and Berger, W. (2009). Differential gene expression in Ndph-knockout mice in retinal development. Invest. Ophthalmol. Vis. Sci. 50, 906–916. doi: 10.1167/iovs.08-1731

PubMed Abstract | CrossRef Full Text | Google Scholar

Schindler, P. A., Settineri, C. A., Collet, X., Fielding, C. J., and Burlingame, A. L. (1995). Site-specific detection and structural characterization of the glycosylation of human plasma proteins lecithin:cholesterol acyltransferase and apolipoprotein D using HPLC/electrospray mass spectrometry and sequential glycosidase digestion. Protein Sci. 4, 791–803. doi: 10.1002/pro.5560040419

PubMed Abstract | CrossRef Full Text | Google Scholar

Schlotter, F., de Freitas, R. C. C., Rogers, M. A., Blaser, M. C., Wu, P.-J., Higashi, H., et al. (2020). ApoC-III is a novel inducer of calcification in human aortic valves. J. Biol. Chem. 296:100193. doi: 10.1074/jbc.RA120.015700

PubMed Abstract | CrossRef Full Text | Google Scholar

Schröder, B., Elsässer, H.-P., Schmidt, B., and Hasilik, A. (2007). Characterisation of lipofuscin-like lysosomal inclusion bodies from human placenta. FEBS Lett. 581, 102–108. doi: 10.1016/j.febslet.2006.12.005

PubMed Abstract | CrossRef Full Text | Google Scholar

Séguin, D., Desforges, M., and Rassart, E. (1995). Molecular characterization and differential mRNA tissue distribution of mouse apolipoprotein D. Brain Res. Mol. Brain Res. 30, 242–250. doi: 10.1016/0169-328X(95)00008-G

PubMed Abstract | CrossRef Full Text | Google Scholar

Selim, A. A., El-Ayat, G., and Wells, C. A. (2001). Immunohistochemical localization of gross cystic disease fluid protein-15,−24 and−44 in ductal carcinoma in situ of the breast: relationship to the degree of differentiation. Histopathology 39, 198–202. doi: 10.1046/j.1365-2559.2001.01178.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Serra Díaz, C., Vizoso, F., Lamelas, M. L., Rodríguez, J. C., González, L. O., Baltasar, A., et al. (1999). Expression and clinical significance of apolipoprotein D in male breast cancer and gynaecomastia. Br. J. Surg. 86, 1190–1197. doi: 10.1046/j.1365-2168.1999.01157.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Serra, C., Vizoso, F., Lamelas, M. L., Rodríguez, J. C., González, L. O., Merino, A. M., et al. (2000). Comparative study of two androgen-induced markers (apolipoprotein D and pepsinogen C) in female and male breast carcinoma. Int. J. Surg. Investig. 2, 183–192.

PubMed Abstract | Google Scholar

Shibata, N., Nagata, T., Shinagawa, S., Ohnuma, T., Shimazaki, H., Komatsu, M., et al. (2013). Genetic association between APOA1 and APOD polymorphisms and Alzheimer's disease in a Japanese population. J. Neural Transm. 120, 1599–1603. doi: 10.1007/s00702-013-1036-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Simard, J., Dauvois, S., Haagensen, D. E., Lévesque, C., Mérand, Y., and Labrie, F. (1990). Regulation of progesterone-binding breast cyst protein GCDFP-24 secretion by estrogens and androgens in human breast cancer cells: a new marker of steroid action in breast cancer. Endocrinology 126, 3223–3231. doi: 10.1210/endo-126-6-3223

PubMed Abstract | CrossRef Full Text | Google Scholar

Simard, J., de Launoit, Y., Haagensen, D. E., and Labrie, F. (1992). Additive stimulatory action of glucocorticoids and androgens on basal and estrogen-repressed apolipoprotein-D messenger ribonucleic acid levels and secretion in human breast cancer cells. Endocrinology 130, 1115–1121.

PubMed Abstract | Google Scholar

Simard, J., Veilleux, R., de Launoit, Y., Haagensen, D. E., and Labrie, F. (1991). Stimulation of apolipoprotein D secretion by steroids coincides with inhibition of cell proliferation in human LNCaP prostate cancer cells. Cancer Res. 51, 4336–4341.

PubMed Abstract | Google Scholar

Singh, S. A., Andraski, A. B., Pieper, B., Goh, W., Mendivil, C. O., Sacks, F. M., et al. (2016). Multiple apolipoprotein kinetics measured in human HDL by high-resolution/accurate mass parallel reaction monitoring. J. Lipid Res. 57, 714–728. doi: 10.1194/jlr.D061432

PubMed Abstract | CrossRef Full Text | Google Scholar

Skerra, A. (2000). Lipocalins as a scaffold. Biochim. Biophys. Acta Prot. Struct. Mol. Enzymol. 1482, 337–350. doi: 10.1016/S0167-4838(00)00145-X

PubMed Abstract | CrossRef Full Text | Google Scholar

Smith, K. M., Lawn, R. M., and Wilcox, J. N. (1990). Cellular localization of apolipoprotein D and lecithin:cholesterol acyltransferase mRNA in rhesus monkey tissues by in situ hybridization. J. Lipid Res. 31, 995–1004. doi: 10.1016/S0022-2275(20)42739-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Søiland, H., Janssen, E. A. M., Kørner, H., Varhaug, J. E., Skaland, I., Gudlaugsson, E., et al. (2009a). Apolipoprotein D predicts adverse outcome in women >or=70 years with operable breast cancer. Breast Cancer Res. Treat. 113, 519–528. doi: 10.1007/s10549-008-9955-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Søiland, H., Skaland, I., Varhaug, J. E., Kørner, H., Janssen, E. A. M., Gudlaugsson, E., et al. (2009b). Co-expression of estrogen receptor alpha and Apolipoprotein D in node positive operable breast cancer–possible relevance for survival and effects of adjuvant tamoxifen in postmenopausal patients. Acta Oncol 48, 514–521. doi: 10.1080/02841860802620613

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, J. Y., Lee, J. K., Lee, N. W., Jung, H. H., Kim, S. H., and Lee, K. W. (2008). Microarray analysis of normal cervix, carcinoma in situ, and invasive cervical cancer: identification of candidate genes in pathogenesis of invasion in cervical cancer. Int. J. Gynecol. Cancer 18, 1051–1059. doi: 10.1111/j.1525-1438.2007.01164.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, Y. J. C., Halliday, G. M., Holton, J. L., Lashley, T., O'Sullivan, S. S., McCann, H., et al. (2009). Degeneration in different parkinsonian syndromes relates to astrocyte type and astrocyte protein expression. J. Neuropathol. Exp. Neurol. 68, 1073–1083. doi: 10.1097/NEN.0b013e3181b66f1b

PubMed Abstract | CrossRef Full Text | Google Scholar

Søreide, J. A., Kolnes, J., Skarstein, A., Aas, T., and Kvinnsland, S. (1994). Progesterone binding cyst protein in hormone receptor positive breast cancer; a predictive factor for effect of adjuvant tamoxifen treatment. Anticancer Res. 14, 2105–2108.

PubMed Abstract | Google Scholar

Søreide, J. A., Lea, O. A., Anda, O., Skarstein, A., Varhaug, J. E., and Kvinnsland, S. (1991a). Progesterone-binding cyst protein (PBCP) in operable breast cancer: correlations with prognostic factors and predictive value for effect of adjuvant tamoxifen treatment. Anticancer Res. 11, 601–605.

PubMed Abstract | Google Scholar

Søreide, J. A., Lea, O. A., and Kvinnsland, S. (1991b). Progesterone-binding cyst protein (PBCP = GCDFP-24) and steroid hormone receptors as markers of differentiation in breast cancer. Inverse relation of distribution in normal and malignant tissue of the same breast. Anticancer Res. 11, 1323–1326.

PubMed Abstract | Google Scholar

Soria, J., Acera, A., Merayo-LLoves, J., Durán, J. A., González, N., Rodriguez, S., et al. (2017). Tear proteome analysis in ocular surface diseases using label-free LC-MS/MS and multiplexed-microarray biomarker validation. Sci. Rep. 7:17478. doi: 10.1038/s41598-017-17536-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Spreyer, P., Schaal, H., Kuhn, G., Rothe, T., Unterbeck, A., Olek, K., et al. (1990). Regeneration-associated high level expression of apolipoprotein D mRNA in endoneurial fibroblasts of peripheral nerve. EMBO J. 9, 2479–2484. doi: 10.1002/j.1460-2075.1990.tb07426.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Sreckovic, I., Birner-Gruenberger, R., Obrist, B., Stojakovic, T., Scharnagl, H., Holzer, M., et al. (2013). Distinct composition of human fetal HDL attenuates its anti-oxidative capacity. Biochim. Biophys. Acta 1831, 737–746. doi: 10.1016/j.bbalip.2012.12.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Starodubtseva, N. L., Kononikhin, A. S., Bugrova, A. E., Chagovets, V., Indeykina, M., Krokhina, K. N., et al. (2016). Investigation of urine proteome of preterm newborns with respiratory pathologies. J. Proteomics 149, 31–37. doi: 10.1016/j.jprot.2016.06.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Steyrer, E., and Kostner, G. M. (1988). Activation of lecithin-cholesterol acyltransferase by apolipoprotein D: comparison of proteoliposomes containing apolipoprotein D, A-I or C-I. Biochim. Biophys. Acta 958, 484–491. doi: 10.1016/0005-2760(88)90235-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Sugimoto, K., Simard, J., Haagensen, D. E., and Labrie, F. (1994). Inverse relationships between cell proliferation and basal or androgen-stimulated apolipoprotein D secretion in LNCaP human prostate cancer cells. J. Steroid Biochem. Mol. Biol. 51, 167–174. doi: 10.1016/0960-0760(94)90090-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Sulkava, M., Raitoharju, E., Levula, M., Seppälä, I., Lyytikäinen, L.-P., Mennander, A., et al. (2017). Differentially expressed genes and canonical pathway expression in human atherosclerotic plaques - Tampere Vascular Study. Sci. Rep. 7:41483. doi: 10.1038/srep41483

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, D., Zhang, H., Guo, D., Sun, A., and Wang, H. (2013). Shotgun proteomic analysis of plasma from dairy cattle suffering from footrot: characterization of potential disease-associated factors. PLoS ONE 8:e55973. doi: 10.1371/journal.pone.0055973

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, H., Wang, D., Liu, D., Guo, Z., Shao, C., Sun, W., et al. (2019). Differential urinary proteins to diagnose coronary heart disease based on iTRAQ quantitative proteomics. Anal. Bioanal. Chem. 411, 2273–2282. doi: 10.1007/s00216-019-01668-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Sun, Q., Disher, M. J., Rustad, T., Telian, S. A., and Andrews, P. C. (1998). AP30, a differential protein marker for perilymph and cerebrospinal fluid in middle ear fluid, has been purified and identified as human apolipoprotein D. Biochim. Biophys. Acta 1384, 405–413. doi: 10.1016/S0167-4838(97)00198-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Suresh, S., Yan, Z., Patel, R. C., Patel, Y. C., and Patel, S. C. (1998). Cellular cholesterol storage in the Niemann-Pick disease type C mouse is associated with increased expression and defective processing of apolipoprotein D. J. Neurochem. 70, 242–251. doi: 10.1046/j.1471-4159.1998.70010242.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Tanase-Nakao, K., Mizuno, K., Hayashi, Y., Kojima, Y., Hara, M., Matsumoto, K., et al. (2019). Dihydrotestosterone induces minor transcriptional alterations in genital skin fibroblasts of children with and without androgen insensitivity. Endocr. J. 66, 387–393. doi: 10.1507/endocrj.EJ18-0494

PubMed Abstract | CrossRef Full Text | Google Scholar

Tao, Z., Song, W., Zhu, C., Xu, W., Liu, H., Zhang, S., et al. (2017). Comparative transcriptomic analysis of high and low egg-producing duck ovaries. Poult. Sci. 96, 4378–4388. doi: 10.3382/ps/pex229

PubMed Abstract | CrossRef Full Text | Google Scholar

Tapia, A., Vilos, C., Marín, J. C., Croxatto, H. B., and Devoto, L. (2011). Bioinformatic detection of E47, E2F1 and SREBP1 transcription factors as potential regulators of genes associated to acquisition of endometrial receptivity. Reprod. Biol. Endocrinol. 9:14. doi: 10.1186/1477-7827-9-14

PubMed Abstract | CrossRef Full Text | Google Scholar

Terrisse, L., Marcoux, K., Do Carmo, S., Brissette, L., Milne, R., and Rassart, E. (2001). Structure-function relationships of human apolipoprotein D an immunochemical analysis. Life Sci. 70, 629–638. doi: 10.1016/S0024-3205(01)01439-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Terrisse, L., Poirier, J., Bertrand, P., Merched, A., Visvikis, S., Siest, G., et al. (1998). Increased levels of apolipoprotein D in cerebrospinal fluid and hippocampus of Alzheimer's patients. J. Neurochem. 71, 1643–1650. doi: 10.1046/j.1471-4159.1998.71041643.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Terrisse, L., Séguin, D., Bertrand, P., Poirier, J., Milne, R., and Rassart, E. (1999). Modulation of apolipoprotein D and apolipoprotein E expression in rat hippocampus after entorhinal cortex lesion. Brain Res. Mol. Brain Res. 70, 26–35. doi: 10.1016/S0169-328X(99)00123-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Tew, S. R., Clegg, P. D., Brew, C. J., Redmond, C. M., and Hardingham, T. E. (2007). SOX9 transduction of a human chondrocytic cell line identifies novel genes regulated in primary human chondrocytes and in osteoarthritis. Arthritis Res. Ther. 9:R107. doi: 10.1186/ar2311

PubMed Abstract | CrossRef Full Text | Google Scholar

Thalmann, I., Kohut, R. I., Ryu, J., Comegys, T. H., Senarita, M., and Thalmann, R. (1994). Protein profile of human perilymph: in search of markers for the diagnosis of perilymph fistula and other inner ear disease. Otolaryngol. Head Neck Surg. 111, 273–280. doi: 10.1177/01945998941113P117

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., Danielson, P. E., Nelson, P. A., Pribyl, T. M., Hilbush, B. S., Hasel, K. W., et al. (2001a). Clozapine increases apolipoprotein D expression in rodent brain: towards a mechanism for neuroleptic pharmacotherapy. J. Neurochem. 76, 789–796. doi: 10.1046/j.1471-4159.2001.00027.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., Dean, B., Pavey, G., and Sutcliffe, J. G. (2001b). Increased CNS levels of apolipoprotein D in schizophrenic and bipolar subjects: implications for the pathophysiology of psychiatric disorders. Proc. Natl. Acad. Sci. U.S.A. 98, 4066–4071. doi: 10.1073/pnas.071056198

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., Dean, B., Scarr, E., Copolov, D., and Sutcliffe, J. G. (2003a). Differences in neuroanatomical sites of apoD elevation discriminate between schizophrenia and bipolar disorder. Mol. Psychiatry 8, 167–175. doi: 10.1038/sj.mp.4001223

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., George, R. C., Danielson, P. E., Nelson, P. A., Warren, A. J., Lo, D., et al. (2003b). Antipsychotic drug treatment alters expression of mRNAs encoding lipid metabolism-related proteins. Mol. Psychiatry 8, 983–993, 950. doi: 10.1038/sj.mp.4001425

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., George, R. C., and Sutcliffe, J. G. (2003c). Apolipoprotein D modulates arachidonic acid signaling in cultured cells: implications for psychiatric disorders. Prostaglandins Leukot. Essent. Fatty Acids 69, 421–427. doi: 10.1016/j.plefa.2003.08.014

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., Laws, S. M., Sutcliffe, J. G., Harper, C., Dean, B., McClean, C., et al. (2003d). Apolipoprotein D levels are elevated in prefrontal cortex of subjects with Alzheimer's disease: no relation to apolipoprotein E expression or genotype. Biol. Psychiatry 54, 136–141. doi: 10.1016/s0006-3223(02)01976-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., Sautkulis, L. N., Criado, J. R., Games, D., and Sutcliffe, J. G. (2001c). Apolipoprotein D mRNA expression is elevated in PDAPP transgenic mice. J. Neurochem. 79, 1059–1064. doi: 10.1046/j.1471-4159.2001.00654.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Thomas, E. A., and Yao, J. K. (2007). Clozapine specifically alters the arachidonic acid pathway in mice lacking apolipoprotein D. Schizophr. Res. 89, 147–153. doi: 10.1016/j.schres.2006.08.011

PubMed Abstract | CrossRef Full Text | Google Scholar

Tomarev, S. I., Wistow, G., Raymond, V., Dubois, S., and Malyukova, I. (2003). Gene expression profile of the human trabecular meshwork: NEIBank sequence tag analysis. Invest. Ophthalmol. Vis. Sci. 44, 2588–2596. doi: 10.1167/iovs.02-1099

PubMed Abstract | CrossRef Full Text | Google Scholar

Trieu, V. N., and Uckun, F. M. (2000). Apolipoprotein E and apolipoprotein D expression in a murine model of singlet oxygen-induced cerebral stroke. Biochem. Biophys. Res. Commun. 268, 835–841. doi: 10.1006/bbrc.2000.2205

PubMed Abstract | CrossRef Full Text | Google Scholar

Tristán-Noguero, A., Borràs, E., Molero-Luis, M., Wassenberg, T., Peters, T., Verbeek, M. M., et al. (2021). Novel protein biomarkers of monoamine metabolism defects correlate with disease severity. Mov. Disord. 36, 690–703. doi: 10.1002/mds.28362

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsukamoto, K., Mani, D. R., Shi, J., Zhang, S., Haagensen, D. E., Otsuka, F., et al. (2013). Identification of apolipoprotein D as a cardioprotective gene using a mouse model of lethal atherosclerotic coronary artery disease. Proc. Natl. Acad. Sci. U.S.A. 110, 17023–17028. doi: 10.1073/pnas.1315986110

PubMed Abstract | CrossRef Full Text | Google Scholar

Utermann, G., Menzel, H.-J., Adler, G., Dieker, P., and Weber, W. (1980). Substitution in vitro of Lecithin–cholesterol acyltransferase. Eur. J. Biochem. 107, 225–241. doi: 10.1111/j.1432-1033.1980.tb04643.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Utsunomiya, T., Ogawa, K., Yoshinaga, K., Ohta, M., Yamashita, K., Mimori, K., et al. (2005). Clinicopathologic and prognostic values of apolipoprotein D alterations in hepatocellular carcinoma. Int. J. Cancer 116, 105–109. doi: 10.1002/ijc.20986

PubMed Abstract | CrossRef Full Text | Google Scholar

van den Boom, J., Wolter, M., Blaschke, B., Knobbe, C. B., and Reifenberger, G. (2006). Identification of novel genes associated with astrocytoma progression using suppression subtractive hybridization and real-time reverse transcription-polymerase chain reaction. Int. J. Cancer 119, 2330–2338. doi: 10.1002/ijc.22108

PubMed Abstract | CrossRef Full Text | Google Scholar

Vardi, A., Pri-Or, A., Wigoda, N., Grishchuk, Y., and Futerman, A. H. (2021). Proteomics analysis of a human brain sample from a mucolipidosis type IV patient reveals pathophysiological pathways. Orphanet. J. Rare Dis. 16:39. doi: 10.1186/s13023-021-01679-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Vázquez, J., González, L., Merino, A., and Vizoso, F. (2000). Expression and clinical significance of apolipoprotein D in epithelial ovarian carcinomas. Gynecol. Oncol. 76, 340–347. doi: 10.1006/gyno.1999.5678

PubMed Abstract | CrossRef Full Text | Google Scholar

Vieira, A. V., Lindstedt, K., Schneider, W. J., and Vieira, P. M. (1995). Identification of a circulatory and oocytic avian apolipoprotein D. Mol. Reprod. Dev. 42, 443–446. doi: 10.1002/mrd.1080420411

PubMed Abstract | CrossRef Full Text | Google Scholar

Vijayaraghavan, S., Hitman, G. A., and Kopelman, P. G. (1994). Apolipoprotein-D polymorphism: a genetic marker for obesity and hyperinsulinemia. J. Clin. Endocrinol. Metab. 79, 568–570.

PubMed Abstract | Google Scholar

Vizoso, F., Díez-Itza, I., Sánchez, L. M., Tuya, A. F., Ruibal, A., and López-Otín, C. (1994). Relationship between serum prolactin levels and protein composition of breast secretions in nonlactating women. J. Clin. Endocrinol. Metab. 79, 525–529.

PubMed Abstract | Google Scholar

Vizoso, F., Sánchez, L. M., Díez-Itza, I., Luz Lamelas, M., and López-Otín, C. (1992). Factors affecting protein composition of breast secretions from nonlactating women. Breast Cancer Res. Treat. 23, 251–258. doi: 10.1007/BF01833522

PubMed Abstract | CrossRef Full Text | Google Scholar

Vizoso, F. J., Rodriguez, M., Altadill, A., González-Diéguez, M. L., Linares, A., González, L. O., et al. (2007). Liver expression of steroid hormones and Apolipoprotein D receptors in hepatocellular carcinoma. World J. Gastroenterol. 13, 3221–3227. doi: 10.3748/wjg.v13.i23.3221

PubMed Abstract | CrossRef Full Text | Google Scholar

Vogt, M., and Skerra, A. (2001). Bacterially produced apolipoprotein D binds progesterone and arachidonic acid, but not bilirubin or E-3M2H. J. Mol. Recognit. 14, 79–86. doi: 10.1002/1099-1352(200101/02)14:1<79::AIDJMR521>3.0.CO;2-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Wade, N. M., Tollenaere, A., Hall, M. R., and Degnan, B. M. (2009). Evolution of a novel carotenoid-binding protein responsible for crustacean shell color. Mol. Biol. Evol. 26, 1851–1864. doi: 10.1093/molbev/msp092

PubMed Abstract | CrossRef Full Text | Google Scholar

Waldner, A., Dassati, S., Redl, B., Smania, N., and Gandolfi, M. (2018). Apolipoprotein D concentration in human plasma during aging and in Parkinson's disease: a cross-sectional study. Parkinsons Dis 2018:3751516. doi: 10.1155/2018/3751516

PubMed Abstract | CrossRef Full Text | Google Scholar

Walsh, N., Dale, J., McGraw, K. J., Pointer, M. A., and Mundy, N. I. (2012). Candidate genes for carotenoid coloration in vertebrates and their expression profiles in the carotenoid-containing plumage and bill of a wild bird. Proc. Biol. Sci. 279, 58–66. doi: 10.1098/rspb.2011.0765

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, J., Scholtens, D., Holko, M., Ivancic, D., Lee, O., Hu, H., et al. (2013). Lipid metabolism genes in contralateral unaffected breast and estrogen receptor status of breast cancer. Cancer Prev. Res. 6, 321–330. doi: 10.1158/1940-6207.CAPR-12-0304

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, S.-C. M., Myers, S., Dooms, C., Capon, R., and Muscat, G. E. O. (2010). An ERRbeta/gamma agonist modulates GRalpha expression, and glucocorticoid responsive gene expression in skeletal muscle cells. Mol. Cell. Endocrinol. 315, 146–152. doi: 10.1016/j.mce.2009.07.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Warden, C. H., Diep, A., Taylor, B. A., and Lusis, A. J. (1992). Localization of the gene for apolipoprotein D on mouse chromosome 16. Genomics 12, 851–852. doi: 10.1016/0888-7543(92)90325-M

PubMed Abstract | CrossRef Full Text | Google Scholar

Watanabe, Y., Hirao, Y., Kasuga, K., Tokutake, T., Kitamura, K., Niida, S., et al. (2020). Urinary apolipoprotein C3 is a potential biomarker for Alzheimer's disease. Dement. Geriatr. Cogn. Dis. Extra 10, 94–104. doi: 10.1159/000509561

PubMed Abstract | CrossRef Full Text | Google Scholar

Wei, Y.-J., Huang, Y.-X., Zhang, X.-L., Li, J., Huang, J., Zhang, H., et al. (2008). Apolipoprotein D as a novel marker in human end-stage heart failure: a preliminary study. Biomarkers 13, 535–548. doi: 10.1080/13547500802030363

PubMed Abstract | CrossRef Full Text | Google Scholar

Weinberg, R. B. (1994). Identification of functional domains in the plasma apolipoproteins by analysis of inter-species sequence variability. J. Lipid Res. 35, 2212–2222. doi: 10.1016/S0022-2275(20)39927-2

PubMed Abstract | CrossRef Full Text | Google Scholar

West, R. B., Harvell, J., Linn, S. C., Liu, C. L., Prapong, W., Hernandez-Boussard, T., et al. (2004). Apo D in soft tissue tumors: a novel marker for dermatofibrosarcoma protuberans. Am. J. Surg. Pathol. 28, 1063–1069. doi: 10.1097/01.pas.0000126857.86186.4c

PubMed Abstract | CrossRef Full Text | Google Scholar

Wiklund, O., Fager, G., olofsson, S. O., Wilhelmsson, C., and Bondjers, G. (1980). Serum apolipoprotein levels in relation to acute myocardial infarction and its risk factors–determination of apolipoprotein D. Atherosclerosis 37, 631–636. doi: 10.1016/0021-9150(80)90070-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Wu, M., Li, Q., and Wang, H. (2021). Identification of novel biomarkers associated with the prognosis and potential pathogenesis of breast cancer via integrated bioinformatics analysis. Technol. Cancer Res. Treat. 20, 1–16. doi: 10.1177/1533033821992081

PubMed Abstract | CrossRef Full Text | Google Scholar

Xu, L., Jia, F., Luo, C., Yu, Q., Dai, R., and Li, X. (2018). Unravelling proteome changes of chicken egg whites under carbon dioxide modified atmosphere packaging. Food Chem. 239, 657–663. doi: 10.1016/j.foodchem.2017.06.128

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamashita, K., Upadhyay, S., Osada, M., Hoque, M. O., Xiao, Y., Mori, M., et al. (2002). Pharmacologic unmasking of epigenetically silenced tumor suppressor genes in esophageal squamous cell carcinoma. Cancer Cell 2, 485–495. doi: 10.1016/S1535-6108(02)00215-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Yan, L., Park, J. Y., Dillinger, J.-G., De Lorenzo, M. S., Yuan, C., Lai, L., et al. (2012). Common mechanisms for calorie restriction and adenylyl cyclase type 5 knockout models of longevity. Aging Cell 11, 1110–1120. doi: 10.1111/acel.12013

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C.-L., Kurczab, T., Down, G., Kealey, T., and Langlands, K. (2005). Gene expression profiling of the ageing rat vibrissa follicle. Br. J. Dermatol. 153, 22–28. doi: 10.1111/j.1365-2133.2005.06550.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C. Y., Gu, Z. W., Blanco-Vaca, F., Gaskell, S. J., Yang, M., Massey, J. B., et al. (1994). Structure of human apolipoprotein D: locations of the intermolecular and intramolecular disulfide links. Biochemistry 33, 12451–12455. doi: 10.1021/bi00207a011

PubMed Abstract | CrossRef Full Text | Google Scholar

Yang, C. Y., Kim, T. W., Weng, S. A., Lee, B. R., Yang, M. L., and Gotto, A. M. (1990). Isolation and characterization of sulfhydryl and disulfide peptides of human apolipoprotein B-100. PNAS 87, 5523–5527. doi: 10.1073/pnas.87.14.5523

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, J. K., Thomas, E. A., Reddy, R. D., and Keshavan, M. S. (2005). Association of plasma apolipoproteins D with RBC membrane arachidonic acid levels in schizophrenia. Schizophr. Res. 72, 259–266. doi: 10.1016/j.schres.2004.05.007

PubMed Abstract | CrossRef Full Text | Google Scholar

Yao, Y., and Vieira, A. (2002). Comparative 17beta-estradiol response and lipoprotein interactions of an avian apolipoprotein. Gen. Comp. Endocrinol. 127, 89–93. doi: 10.1016/S0016-6480(02)00032-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Yin, J., Spillman, E., Cheng, E. S., Short, J., Chen, Y., Lei, J., et al. (2021). Brain-specific lipoprotein receptors interact with astrocyte derived apolipoprotein and mediate neuron-glia lipid shuttling. Nat. Commun. 12:2408. doi: 10.1038/s41467-021-22751-7

PubMed Abstract | CrossRef Full Text | Google Scholar

Yokoo, H., Oishi, T., Isoda, K., Nakazato, Y., and Toyokuni, S. (2007). Oxidative stress is related to the formation of Antoni B patterns and eosinophilic hyaline droplets in schwannomas. Neuropathology 27, 237–244. doi: 10.1111/j.1440-1789.2007.00772.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Yoshida, K., Cleaveland, E. S., Nagle, J. W., French, S., Yaswen, L., Ohshima, T., et al. (1996). Molecular cloning of the mouse apolipoprotein D gene and its upregulated expression in Niemann-Pick disease type C mouse model. DNA Cell Biol. 15, 873–882. doi: 10.1089/dna.1996.15.873

PubMed Abstract | CrossRef Full Text | Google Scholar

Yu, R.-H., Zhang, X.-Y., Xu, W., Li, Z.-K., and Zhu, X.-D. (2020). Apolipoprotein D alleviates glucocorticoid-induced osteogenesis suppression in bone marrow mesenchymal stem cells via the PI3K/Akt pathway. J. Orthop. Surg. Res. 15:307. doi: 10.1186/s13018-020-01824-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Zeng, C., Spielman, A. I., Vowels, B. R., Leyden, J. J., Biemann, K., and Preti, G. (1996). A human axillary odorant is carried by apolipoprotein D. Proc. Natl. Acad. Sci. U.S.A. 93, 6626–6630. doi: 10.1073/pnas.93.13.6626

PubMed Abstract | CrossRef Full Text | Google Scholar

Zenkel, M., Pöschl, E., von der Mark, K., Hofmann-Rummelt, C., Naumann, G. O. H., Kruse, F. E., et al. (2005). Differential gene expression in pseudoexfoliation syndrome. Invest. Ophthalmol. Vis. Sci. 46, 3742–3752. doi: 10.1167/iovs.05-0249

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, J., He, Q., Liu, Q. Y., Guo, W., Deng, X. M., Zhang, W. W., et al. (2007). Differential gene expression profile in pig adipose tissue treated with/without clenbuterol. BMC Genomics 8:433. doi: 10.1186/1471-2164-8-433

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, S. X., Bentel, J. M., Ricciardelli, C., Horsfall, D. J., Haagensen, D. E., Marshall, V. R., et al. (1998). Immunolocalization of apolipoprotein D, androgen receptor and prostate specific antigen in early stage prostate cancers. J. Urol. 159, 548–554. doi: 10.1016/S0022-5347(01)63981-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Li, D., Duan, S., Duan, Y., Chen, Q., Li, X., et al. (2006). Analysis of the association between Apolipoprotein D and schizophrenia. Neuropsychobiology 54, 40–44. doi: 10.1159/000095740

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, Y., Chen, K., Sloan, S. A., Bennett, M. L., Scholze, A. R., O'Keeffe, S., et al. (2014). An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34:11929. doi: 10.1523/JNEUROSCI.1860-14.2014

PubMed Abstract | CrossRef Full Text | Google Scholar

Zheng, W., Mast, N., Saadane, A., and Pikuleva, I. A. (2015). Pathways of cholesterol homeostasis in mouse retina responsive to dietary and pharmacologic treatments. J. Lipid Res. 56, 81–97. doi: 10.1194/jlr.M053439

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, J., Ng, S., Adesanya-Famuiya, O., Anderson, K., and Bondy, C. A. (2000). Testosterone inhibits estrogen-induced mammary epithelial proliferation and suppresses estrogen receptor expression. FASEB J. 14, 1725–1730. doi: 10.1096/fj.99-0863com

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, X., Wang, J., Lu, Y., Chen, C., Hu, Y., Liu, P., et al. (2020). Anti-depressive effects of Kai-Xin-San on lipid metabolism in depressed patients and CUMS rats using metabolomic analysis. J. Ethnopharmacol. 252:112615. doi: 10.1016/j.jep.2020.112615

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, Z., Zhang, H., Luo, G., Xu, N., and Pan, Z. (2015). Association between the ABCC11 gene polymorphism and the expression of apolipoprotein D by the apocrine glands in axillary osmidrosis. Mol. Med. Rep. 11, 4463–4467. doi: 10.3892/mmr.2015.3274

PubMed Abstract | CrossRef Full Text | Google Scholar

Zou, S., Zhang, J., for Alzheimer's Disease Neuroimaging Initiative, and Chen, W. (2019). Subtypes based on six apolipoproteins in non-demented elderly are associated with cognitive decline and subsequent tau accumulation in cerebrospinal fluid. J. Alzheimers Dis. 72, 413–423. doi: 10.3233/JAD-190314

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: protein physiology, lipid peroxidation, membrane management, oxidative stress, lipoprotein particles, extracellular vesicles, lysosome, ApoD

Citation: Sanchez D and Ganfornina MD (2021) The Lipocalin Apolipoprotein D Functional Portrait: A Systematic Review. Front. Physiol. 12:738991. doi: 10.3389/fphys.2021.738991

Received: 09 July 2021; Accepted: 30 August 2021;
Published: 07 October 2021.

Edited by:

Elizabeth S. Fernandes, Pelé Pequeno Príncipe Research Institute, Brazil

Reviewed by:

Roland K. Strong, Fred Hutchinson Cancer Research Center, United States
Sten Braesch-Andersen, ImmunoIVD AB, Sweden

Copyright © 2021 Sanchez and Ganfornina. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Maria D. Ganfornina, opabinia@ibgm.uva.es

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.