Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Neurosci., 13 August 2025

Sec. Neuropharmacology

Volume 19 - 2025 | https://doi.org/10.3389/fnins.2025.1603292

This article is part of the Research TopicAdvancements in Translational Stroke Research: Spotlight on the Blood-Brain BarrierView all 3 articles

Exploring novel roles of lipid droplets and lipid metabolism in regulating inflammation and blood–brain barrier function in neurological diseases

Luo Fu,&#x;Luo Fu1,2Ting Luo,&#x;Ting Luo1,3Zhongnan HaoZhongnan Hao4Yongli Pan,Yongli Pan4,5Wenqiang XinWenqiang Xin6Lin Zhang,Lin Zhang4,7Zhuhong LaiZhuhong Lai8Haitao ZhangHaitao Zhang1Hua Liu
Hua Liu1*Wei Wei,
Wei Wei1,4*
  • 1Department of Neurology, The Affiliated Hospital of Southwest Jiaotong University and The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
  • 2Department of Gastrointestinal and Minimally Invasive Surgery, The Affiliated Hospital of Southwest Jiaotong University and The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
  • 3Department of Ophthalmology, The Affiliated Hospital of Southwest Jiaotong University and The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
  • 4Department of Neurology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany
  • 5Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
  • 6Jiangxi Key Laboratory of Neurological Diseases, Department of Neurosurgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
  • 7Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
  • 8Department of Cardiology, Mianyang Central Hospital, Mianyang, Sichuan, China

The blood–brain barrier (BBB) is a critical structure that maintains the brain’s homeostasis by regulating the transport of molecules and protecting it from harmful substances. However, in neurological diseases such as ischemic stroke, Alzheimer’s disease, Parkinson’s disease, and multiple sclerosis, the integrity and function of the BBB can be significantly compromised. In these conditions, BBB disruption leads to increased permeability, which facilitates neuroinflammation, exacerbates neuronal damage, and accelerates disease progression. Recent research has highlighted the potential of lipid-based carriers, including liposomes and lipid droplets (LDs), in modulating the BBB’s integrity and function in various neurological diseases. Liposomes, with their ability to cross the BBB via mechanisms such as receptor-mediated transcytosis and carrier-mediated transport, are emerging as promising vehicles for the targeted delivery of therapeutic agents to the brain. These properties allow liposomes to effectively reduce infarct size and promote neuroprotection in ischemic stroke, as well as deliver drugs in the treatment of neurodegenerative diseases. Furthermore, LDs—dynamic regulators of lipid metabolism and cellular energy—play an essential role in maintaining cellular homeostasis, particularly during periods of stress when BBB function is compromised. These LDs help sustain cellular energy needs and modulate inflammatory responses, which are key factors in maintaining BBB integrity. Surface modifications of liposomes can further enhance their targeting efficiency, enabling them to selectively bind to specific brain cell types, including neurons, astrocytes, and microglia. This customization improves the precision of therapeutic delivery and supports the development of more tailored treatments. However, challenges such as immune responses, rapid clearance, and complement activation-related toxicity continue to hinder the broader application of liposomes and LDs in clinical settings. This review will focus on the roles of liposomes and LDs in regulating BBB integrity across a range of neurological diseases, discussing their potential for targeted drug delivery, neuroprotection, and the modulation of neuroinflammation. Additionally, we will explore the strategies being developed to address the limitations that currently restrict their clinical use.

1 Introduction

The blood–brain barrier (BBB) is a dynamic and highly selective interface that separates the circulating blood from the brain parenchyma (Keaney and Campbell, 2015). Composed primarily of endothelial cells, tight junction proteins, astrocytes, and pericytes, the BBB plays a crucial role in maintaining central nervous system (CNS) homeostasis by regulating the transport of ions, nutrients, signaling molecules, and lipids (Correale and Villa, 2009; Singh and Vellapandian, 2023; Lochhead et al., 2020). Among the critical lipid species required for brain function is docosahexaenoic acid (DHA), an omega-3 fatty acid essential for neuronal development and synaptic function (Dyall, 2015; Sinclair, 2019). The transport of DHA across the BBB is largely mediated by the lipid transporter MFSD2A, which highlights the specialized lipid transport mechanisms necessary to support brain metabolism and function (Chan et al., 2018; Wood et al., 2021). Lipid metabolism is increasingly recognized as a fundamental component of both physiological BBB maintenance and pathological disruption (Pifferi et al., 2021). In this context, lipid droplets (LDs), intracellular organelles involved in lipid storage, signaling, and energy homeostasis, have emerged as key regulators of cellular stress responses, including inflammation and oxidative stress, particularly in neural and endothelial cells (Geltinger et al., 2020; Zhang et al., 2025). Under pathological conditions, the dynamics of LDs are closely associated with neuroinflammatory responses, mitochondrial dysfunction, and the integrity of the endothelial barrier (Parodi-Rullán et al., 2021; Chen et al., 2024; Zhong et al., 2025). For instance, during ischemic stroke, energy failure and oxidative stress compromise the structural and functional integrity of the BBB, resulting in increased permeability, vasogenic edema, and infiltration of peripheral immune cells and neurotoxic substances (Fang et al., 2024; Bernardo-Castro et al., 2020). This BBB breakdown exacerbates neuronal injury and promotes a harmful neuroinflammatory cascade (Gao et al., 2023). In parallel, the development of lipid-based therapeutic strategies—most notably liposomes and LD-modulating agents—has gained traction for their potential in restoring BBB integrity and enhancing drug delivery to the brain (Juhairiyah and de Lange, 2021; Mondal and Ghosh, 2023). Liposomes, due to their amphiphilic bilayer structure, can encapsulate both hydrophilic and hydrophobic drugs, protect them from enzymatic degradation, and facilitate targeted transport across the BBB (Sonju et al., 2021; Bruch et al., 2019). Meanwhile, LDs are increasingly studied not only for their involvement in cellular stress regulation but also as potential targets for modulating inflammation and metabolic dysregulation during cerebrovascular injury (Tan et al., 2024; Lan et al., 2023).

2 Lipid metabolism and neurological disorders

2.1 The brief course on lipid metabolism

Lipid metabolism encompasses the complex biochemical processes involved in the synthesis, transport, and degradation of lipids (Ridgway and McLeod, 2008). These processes are crucial for maintaining cellular homeostasis, energy production, and structural integrity of biological membranes (Fagone and Jackowski, 2009). The major pathways of lipid metabolism include fatty acid oxidation, lipid synthesis, and lipoprotein metabolism (Rinaldo et al., 2002). Dysregulation of lipid metabolism has been implicated in various pathological conditions, including metabolic syndrome, cardiovascular diseases, and neurological disorders (van Meer et al., 2008). In the nervous system, lipids play fundamental roles in neuronal function, including signal transduction, membrane fluidity, and myelination (Dietschy and Turley, 2004). The brain, despite comprising only about 2% of body weight, contains nearly 50% of the body’s total lipids, highlighting the significance of lipid metabolism in neural health and disease (Sonnino et al., 2015).

2.2 Biogenesis of LDs

LDs are dynamic organelles that store neutral lipids, primarily triglycerides and cholesteryl esters (Walther and Farese, 2012). They are formed through the coordinated actions of the endoplasmic reticulum (ER) and various lipid metabolism enzymes (Olzmann and Carvalho, 2019). LD biogenesis is initiated by the deposition of neutral lipids within the ER membrane bilayer, which subsequently buds outward to generate mature lipid droplets (Thiam and Beller, 2017) (Figure 1). LDs were once considered inert lipid reservoirs, but recent studies reveal their active role in lipid homeostasis, signaling, and cellular stress responses (Wang, 2016). In neurological contexts, LDs are increasingly recognized as key regulators of neuronal lipid metabolism, oxidative stress mitigation, and neuroinflammation (Farmer et al., 2020).

Figure 1
Diagram illustrating lipid droplet dynamics, with components like the endoplasmic reticulum, mitochondria, lysosome, and autophagosome. It depicts processes such as lipophagy, de novo lipid synthesis, and the role of reactive oxygen species (ROS). Legend identifies phospholipids, di-/tri-acylglycerol, cholesterol ester, and surface proteins.

Figure 1. Lipid droplet biogenesis. Lipid droplets (LDs) are spherical organelles with a unique structure consisting of a core of neutral lipids, mainly triglycerides (TG) and cholesteryl esters. Their core is surrounded by a monolayer of phospholipids and associated proteins. LDs are dynamic and functionally active organelles involved in various functions such as lipid metabolism, cell signaling and inflammation.

2.3 The vital role of lipid metabolism in neurological disorders

Lipid metabolism is intricately involved in maintaining neuronal function, membrane integrity, and energy homeostasis, and its dysregulation has been implicated in a wide range of neurological disorders (Tracey et al., 2018; Zhao et al., 2023) (Figure 2). In Alzheimer’s disease (AD), impaired lipid metabolism contributes to Aβ aggregation and tau pathology, with cholesterol modulating APP processing and ApoE4 impairing Aβ clearance (Rajmohan and Reddy, 2017). Lipid peroxidation products further amplify oxidative stress and neuroinflammation (Maulik et al., 2013). Similarly, in Parkinson’s disease (PD), especially peroxidized PUFAs and disrupted sphingolipids, promotes α-synuclein aggregation, mitochondrial dysfunction, and lysosomal impairment (Mesbahi, 2023). In multiple sclerosis (MS), disturbed lipid homeostasis affects myelin synthesis and oligodendrocyte function, while lipid mediators such as eicosanoids and sphingolipids modulate immune responses and neuroinflammation (López-Muguruza and Matute, 2023). In Huntington’s disease (HD), involved defective cholesterol and phospholipid metabolism due to mutant huntingtin-induced SREBP dysregulation, compromising synaptic function and neuronal viability (Leoni and Caccia, 2015). In amyotrophic lateral sclerosis (ALS), abnormal fatty acid utilization, hypermetabolism, and dyslipidemia contribute to motor neuron degeneration and inflammation (D'Amico et al., 2021). In ischemic stroke (IS), lipid metabolism plays dual roles in neuroprotection and neurotoxicity by regulating inflammation, oxidative stress, and BBB integrity through phospholipid and sphingolipid pathways (Sun et al., 2016; Zhao et al., 2022; Sandoval and Witt, 2008), whose disruption exacerbates vascular permeability, brain edema, and neuronal injury (Sandoval and Witt, 2008; Arbaizar-Rovirosa et al., 2023). Lipid peroxidation, triggered by oxidative stress following ischemia–reperfusion injury, generates reactive oxygen species (ROS) that further damage neuronal membranes and promote inflammation (Wu et al., 2020). Moreover, cholesterol metabolism influences post-stroke recovery, with evidence suggesting that high levels of oxidized cholesterol derivatives contribute to neuroinflammatory cascades (Ciancarelli et al., 2022; Kloska et al., 2020). Collectively, these findings highlight the fundamental role of lipid metabolism in neurological disorders, emphasizing its impact on neurodegeneration, neuroinflammation, cerebrovascular dysfunction, and bioenergetic deficits (Yin, 2023; Helgudóttir et al., 2024; Clemente-Suárez et al., 2024). Understanding these mechanisms may provide novel therapeutic targets aimed at modulating lipid metabolism to protect against neuronal dysfunction, enhance neuroprotection, and improve recovery following acute neurological insults.

Figure 2
Diagram showing the effects of lipids on neurological diseases and the blood-brain barrier. The diseases include stroke, Alzheimer's, Parkinson's, ALS, MS, and HD. Key lipids and pathways are listed under damage and protection for each condition. A graphic of the blood-brain barrier illustrates its components: neuron, endothelium, astrocyte, microglia, and a detailed view of astrocyte endfoot, pericyte, endothelial cell, tight junction, and basement membrane.

Figure 2. Schematic illustration of lipids and their respective roles in neurological diseases. In the central nervous system, lipid metabolism and lipid LDs play a crucial role in modulating neuroinflammation and preserving BBB integrity in neurological disorders. Disruptions in lipid homeostasis can impair the function of key cellular components, including neurons, microglia, astrocytes, and endothelial cells. Through the regulation of lipid metabolism, processes such as chronic inflammation, neuronal injury, and BBB dysfunction can be influenced, thereby impacting the progression and potential recovery of central nervous system disorders, including Stroke, Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS), Huntington’s disease (HD), and multiple sclerosis (MS). BBB, Blood–brain barrier; AS, Astrocytes; EC, Endothelial cell; CEs, Cholesterol esters; TC, Triglycerides; FA, Fatty Acid; LDs, Lipid droplets; LPA, Lysophosphatidic acid; ROS, Reactive oxygen species; ICH, Intracerebral hemorrhagic stroke; IS, Ischemic stroke; ACSL4, Acyl-CoA synthetase long-chain family member 4; S1P, Sphingosine-1-phosphate; S1PR3, Sphingosine-1-phosphate receptor 3; AD, Alzheimer’s disease; TREM2, Triggering Receptor Expressed on Myeloid cells 2; ApoE, Apolipoprotein E; LXR, Liver X Receptor; ABCA1, ATP-binding cassette transporter A1; PLA2, Phospholipase A2; PD, Parkinson’s disease; COX-2, Cyclooxygenase-2; PPAR-α, Peroxisome proliferator-activated receptor alpha; ACAT1, Acyl-CoA cholesterol acyltransferase 1; 4-HNE, 4-Hydroxynonenal; 10-NO2-OA, Nitroalkene 10-nitro-oleic acid; MS, Multiple sclerosis; HD, Huntington’s disease; ALS, Amyotrophic lateral sclerosis; HDL-C, high-density lipoprotein cholesterol.

2.4 Lipid metabolism in BBB penetration

Lipid metabolism plays a fundamental role in maintaining the structural and functional integrity of the BBB, a highly selective barrier that regulates molecular exchange between the bloodstream and the CNS (Pifferi et al., 2021). The BBB is composed of endothelial cells with tight junctions, pericytes, and astrocyte end-feet, all of which rely on tightly regulated lipid homeostasis to preserve barrier integrity and control permeability (Singh and Vellapandian, 2023). The lipid composition of the BBB is unique, being highly enriched in sphingolipids, phospholipids, and cholesterol, which are critical for maintaining membrane fluidity, vesicular trafficking, and signaling (Wang et al., 2020). Changes in lipid metabolism can directly influence BBB function, either enhancing its protective role or contributing to its dysfunction in neurological diseases. For instance, sphingolipids and ceramides are known regulators of BBB permeability, with their dysregulation leading to increased endothelial cell apoptosis, inflammation, and barrier disruption (Kuperberg and Wadgaonkar, 2017). Cholesterol metabolism also plays a crucial role, as cholesterol is a key component of endothelial membranes and lipid rafts, which mediate signal transduction and transport mechanisms at the BBB (Dai et al., 2021). However, excessive cholesterol accumulation or the presence of oxidized cholesterol derivatives can trigger inflammatory cascades, leading to endothelial dysfunction and increased BBB permeability, which has been implicated in diseases such as AD, MS, and stroke (Candore et al., 2010).

Lipid transporters and lipid-binding proteins further influence BBB penetration and function (Yamazaki et al., 2019). Apolipoproteins such as ApoE, which facilitates lipid transport in the brain via lipoprotein particles, significantly impact BBB dynamics. The ApoE4 variant, for example, has been associated with BBB breakdown and neurovascular dysfunction, increasing the risk of neurodegenerative diseases (Montagne et al., 2020). Additionally, ATP-binding cassette transporters, such as ABCA1 and ABCG1, regulate cholesterol and lipid efflux from endothelial cells, thereby modulating BBB permeability (Rohrer et al., 2009). Dysregulation of these transporters can impair lipid clearance, leading to increased neuroinflammation and neurovascular damage. Fatty acid metabolism also plays a critical role in BBB function, as endothelial cells utilize specific fatty acid oxidation pathways to maintain barrier homeostasis (Fock and Parnova, 2023). Disruptions in fatty acid oxidation have been linked to increased BBB permeability and neuroinflammatory responses, particularly in conditions like MS and ischemic stroke (Mitchell and Hatch, 2011).

Beyond its endogenous functions, lipid metabolism is also crucial for drug delivery strategies targeting the CNS (Correia et al., 2022). The BBB presents a major challenge for therapeutic agents due to its restrictive permeability, but lipid-based approaches have been explored to enhance drug transport across the barrier (Wu et al., 2023). Although lipophilic drugs, lipid nanoparticles, and liposome-based carriers have been explored to enhance drug delivery across the BBB via lipid-mediated transport, the notion that liposomes easily cross the BBB is often misleading (Micheli et al., 2012). While PEGylation improves systemic circulation by reducing clearance, it does not inherently facilitate BBB penetration and may even hinder interaction with the endothelial surface. Effective transport across the BBB typically requires active targeting strategies, such as receptor-mediated transcytosis. This involves functionalizing liposomes with ligands or peptides that bind to specific receptors expressed on brain endothelial cells—such as the transferrin receptor (TfR), insulin receptor (IR), and low-density lipoprotein receptor (LDLR). These receptors naturally mediate the transport of essential molecules like iron, insulin, and cholesterol into the brain. By mimicking endogenous ligands, targeted liposomes can hijack these pathways to enhance BBB penetration. However, such strategies are still underutilized and often insufficiently optimized in preclinical models, limiting their translational impact. Nevertheless, understanding the interplay between lipid metabolism and BBB function not only provides insights into disease mechanisms but also opens avenues for developing novel therapeutics aimed at restoring BBB integrity and enhancing drug delivery for neurological disorders.

3 Lipid metabolism and inflammation

3.1 The brief course on inflammation

Inflammation plays a central role in neurological diseases, acting as both an acute defense mechanism and a contributor to chronic neurodegeneration when dysregulated (Leng and Edison, 2021; Marogianni et al., 2020; Tansey et al., 2022). In the CNS, inflammation is primarily mediated by microglia and astrocytes, which become activated in response to injury or pathological stimuli, releasing pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6, as well as ROS, leading to neuronal damage and BBB disruption (Sochocka et al., 2017). In AD, Aβ deposition persistently stimulates microglia, promoting chronic neuroinflammation that accelerates neuronal degeneration (Leng and Edison, 2021). In PD, aggregated α-synuclein triggers microglial activation, releasing toxic mediators that exacerbate dopaminergic neuron loss (Zhang et al., 2005). MS is characterized by T-cell-mediated autoimmune attacks on myelin, leading to demyelination, neurodegeneration, and lipid metabolism dysregulation, further impairing myelin synthesis and repair (Correale et al., 2019). In ischemic stroke, hypoxia-ischemia induces an inflammatory cascade where activated microglia and infiltrating peripheral immune cells release inflammatory mediators, amplifying neuronal injury, while lipid peroxidation and BBB breakdown further exacerbate inflammation (Zhang, 2014; Maida et al., 2020). Additionally, lipid metabolism plays a crucial role in regulating neuroinflammation, with arachidonic acid-derived prostaglandins and leukotrienes promoting inflammation, whereas ω-3 fatty acid metabolites such as resolvins and protecting facilitate inflammation resolution and homeostasis (Shang et al., 2019; Farooqui, 2012). The intricate crosstalk between inflammation and lipid metabolism is particularly evident in neurodegenerative diseases, cerebrovascular disorders, and neuroimmune conditions, highlighting potential therapeutic targets for anti-inflammatory and metabolic interventions.

3.2 A double-edged sword role of lipids and possible signal pathway

Lipid metabolism plays a paradoxical role in the CNS, acting as both a protector of homeostasis and a contributor to disease progression (Zhang et al., 2025; Walther and Farese, 2012). On one hand, lipids are essential for neuronal membrane integrity, synaptic function, and energy metabolism (Walther and Farese, 2012). Dysregulated lipid pathways can drive neuroinflammation, oxidative stress, and neurodegeneration (Zhang et al., 2025; Farmer et al., 2020). Several key lipid-related signaling pathways illustrate this dual role in neurological health and disease. The SREBP pathway, a central regulator of cholesterol and lipid homeostasis, is vital for maintaining neuronal function (Shimano and Sato, 2017). Under normal conditions, SREBP activation ensures adequate lipid supply for membrane synthesis and repair (Wang et al., 2025). However, in diseases such as AD and HD, impaired SREBP signaling disrupts lipid balance, leading to synaptic dysfunction and neuronal death (Wang et al., 2025; Hu et al., 2023; Li et al., 2020). Similarly, the PPAR (Peroxisome Proliferator-Activated Receptor) family, particularly PPAR-γ, plays a neuroprotective role by regulating fatty acid oxidation, anti-inflammatory responses, and mitochondrial function (Kapadia et al., 2008; Fidaleo et al., 2014; Escandon et al., 2021). Activation of PPAR-γ has been shown to mitigate neuroinflammation in PD and MS (Escandon et al., 2021; Chen et al., 2012). However, excessive PPAR-γ activation may also lead to lipid accumulation, contributing to metabolic stress and neuronal vulnerability (Xi et al., 2020). Sphingolipid metabolism exemplifies the complex interplay between lipid signaling, neuroinflammation, and BBB regulation (Arsenault et al., 2021). A central mediator in this pathway is sphingosine-1-phosphate (S1P), a bioactive lipid that binds to a family of G-protein–coupled receptors (S1PR1–S1PR5) expressed on endothelial cells, astrocytes, and immune cells. S1P signaling plays a dual role: it promotes BBB stability by enhancing endothelial cell junction integrity and simultaneously regulates immune cell trafficking across the BBB (Cartier and Hla, 2019; Wang et al., 2025). Dysregulation of this pathway contributes to pathological immune infiltration and barrier breakdown in neuroinflammatory disorders (Wang et al., 2025). Therapeutically, S1P receptor modulators such as fingolimod (FTY720) function by downregulating S1PR1 on lymphocytes, thereby sequestering them in lymph nodes and reducing CNS infiltration (Brinkmann, 2009; Pournajaf et al., 2022). In MS, this mechanism attenuates neuroinflammation and protects BBB integrity, making S1P signaling a critical target for both immune modulation and vascular stabilization (Bravo et al., 2022; Groves et al., 2013). However, elevated levels of ceramides and sphingolipids can induce apoptosis and neurotoxicity, exacerbating disease pathology in stroke and neurodegenerative disorders (Farooqui and Farooqui, 2024).

LDs, once considered passive fat reservoirs, have emerged as critical regulators of cellular homeostasis (Tracey et al., 2018). In glial cells, LDs sequester toxic lipid peroxidation products, protecting neurons from oxidative damage (Olzmann and Carvalho, 2019). Persistent accumulation of LDs under pathological conditions—such as in ALS, PD, and other neuroinflammatory disorders—has been increasingly recognized as a contributor to chronic inflammation and metabolic dysfunction (Farmer et al., 2020). LDs serve not only as energy reserves but also as platforms for the synthesis and storage of bioactive lipids, including pro-inflammatory mediators (Tan et al., 2024; Zhang et al., 2025). A key inflammatory pathway linked to LD metabolism is the cyclooxygenase-lipoxygenase (COX-LOX) axis, which metabolizes arachidonic acid into a diverse range of eicosanoids, such as prostaglandins, leukotrienes, and thromboxanes (Wang et al., 2021; Rudrapal et al., 2023). These lipid mediators orchestrate immune cell recruitment, glial activation, oxidative stress, and vascular permeability—processes that are central to the progression of neurodegenerative diseases (Rudrapal et al., 2023; Adibhatla and Hatcher, 2010). In PD, aberrant activation of the COX-2 isoform has been implicated in dopaminergic neuron loss, with elevated levels of PGE2 detected in affected brain regions (Kumar et al., 2020). While PGE2 and leukotrienes amplify inflammatory responses in acute CNS injury and chronic neurodegeneration, their counterbalancing counterparts, such as resolvins and protectins, derived from omega-3 fatty acids, promote the resolution of inflammation and neuroprotection (Artru et al., 2022). Similarly, LOX-derived leukotrienes have been associated with microglial activation and neurotoxicity (Aoki et al., 2023). However, evidence remains conflicted, as some lipid mediators—such as lipoxins and resolvins—produced downstream of LOX pathways exhibit anti-inflammatory and pro-resolving effects (Spite et al., 2014; Buckley et al., 2014). This dual nature highlights the complexity of targeting the COX-LOX axis therapeutically: while inhibition of COX-2 has shown neuroprotective effects in preclinical PD models, clinical outcomes with COX inhibitors have been inconsistent, possibly due to interference with beneficial lipid mediators or compensatory pathway activation (Mukhopadhyay et al., 2023; Agrawal, 2025). Adding to this complexity is the role of acyl-CoA cholesterol acyltransferase 1 (ACAT1), an enzyme that regulates intracellular cholesterol esterification and LD formation (Xu et al., 2019; Chang et al., 2001). In PD, ACAT1 has been found to be upregulated in activated microglia and is associated with cholesterol ester accumulation and sustained neuroinflammatory responses (Chang et al., 2001; Huynh et al., 2024). Genetic or pharmacological inhibition of ACAT1 has been shown to reduce LD burden, suppress pro-inflammatory cytokine release, and protect dopaminergic neurons in experimental models of PD and AD (Huynh et al., 2024; Sun et al., 2025; Valencia-Olvera et al., 2023). These findings suggest that targeting ACAT1 may not only restore lipid homeostasis but also indirectly modulate COX-LOX signaling by limiting the availability of lipid substrates stored in LDs.

These observations underscore the central role of lipid-mediated inflammation in neurodegeneration, while also revealing the intricate balance between protective and harmful lipid signaling. The imbalance between pro-inflammatory and pro-resolving lipid mediators is a hallmark of neuroinflammatory diseases (Ahluwalia et al., 2022). These findings highlight the intricate role of lipid metabolism in neurological disorders, where the same pathways that sustain brain homeostasis can, under pathological conditions, drive disease progression. Understanding these signaling networks may pave the way for targeted lipid-based therapies that balance neuroprotection and metabolic regulation in CNS disorders.

3.3 The crosstalk of impact of inflammation on LDs

Emerging evidence suggests that LDs function as key regulators of cellular responses to stress, particularly in the context of inflammation (Walther and Farese, 2012). The interplay between inflammation and LD metabolism is bidirectional: inflammatory stimuli can induce LD biogenesis and alter lipid composition, while LDs, in turn, modulate inflammatory signaling pathways by sequestering bioactive lipids and influencing immune cell function (Tan et al., 2024; Monson et al., 2021). Inflammation profoundly impacts LD formation and turnover through multiple mechanisms. Pro-inflammatory cytokines such as TNF-α, IL-1β, and IFN-γ promote LD accumulation in various cell types, including microglia, astrocytes, and endothelial cells (Rodríguez-Gómez et al., 2020; Fornari Laurindo et al., 2023). This process serves both protective and pathological roles—on one hand, LDs act as reservoirs to store potentially harmful lipids, preventing lipotoxicity and excessive oxidative stress (Geltinger et al., 2020; Pressly et al., 2022). On the other hand, LDs serve as platforms for the synthesis of eicosanoids, prostaglandins, and leukotrienes, which are lipid mediators that amplify the inflammatory response (Pereira-Dutra and Bozza, 2021). Additionally, chronic inflammation disrupts LD homeostasis, leading to excessive lipid accumulation, mitochondrial dysfunction, and sustained immune activation, all of which contribute to neuroinflammation and disease progression (Leyane et al., 2022).

The regulation of LD dynamics during inflammation is tightly controlled by lipid metabolism-associated transcription factors, transporters, and structural proteins (Florance and Ramasubbu, 2022). Several key regulators, including TREM2, SREBPs, ABCA1, Perilipins (PLIN2-5), ApoE, and LXRs, play crucial roles in balancing LD formation, lipid transport, and inflammatory signaling (Florance and Ramasubbu, 2022). Understanding how these factors mediate the crosstalk between inflammation and LDs provides important insights into potential therapeutic targets for neuroinflammatory and neurodegenerative diseases.

3.4 Key regulatory factors in the crosstalk between inflammation and LDs

3.4.1 TREM2

TREM2 is a lipid-sensing receptor expressed in microglia that plays a critical role in lipid uptake, LD biogenesis, and the resolution of inflammation (Hou et al., 2022). Under inflammatory conditions, TREM2 facilitates the clearance of apoptotic cells and lipid debris by promoting phagocytosis and lipid catabolism. Loss-of-function mutations in TREM2, such as those associated with neurodegenerative diseases, impair LD metabolism, leading to lipid overload, increased oxidative stress, and chronic microglial activation (Klionsky et al., 2021; Smolič et al., 2021). Furthermore, TREM2-deficient microglia fail to efficiently regulate inflammation, resulting in prolonged cytokine release and exacerbated neurodegeneration (Hou et al., 2022). The interaction between TREM2 and LDs highlights the importance of lipid metabolism in controlling inflammatory responses in the brain (Gouna, 2024).

3.4.2 SREBPs

SREBPs are master regulators of lipid biosynthesis, controlling the expression of genes involved in fatty acid and cholesterol synthesis (Eberlé et al., 2004). Inflammation activates SREBPs through cytokine signaling pathways, leading to increased lipid accumulation within LDs (Sahini and Borlak, 2014). While SREBP activation is essential for maintaining membrane integrity and cellular lipid reserves, excessive activation in response to chronic inflammation can result in lipid imbalances, mitochondrial dysfunction, and inflammatory stress (Jarc and Petan, 2019). In microglia and astrocytes, SREBP-driven lipid accumulation contributes to a pro-inflammatory state by fueling the production of prostaglandins and other eicosanoids (Zhang et al., 2025). Additionally, cross-talk between SREBPs and inflammatory pathways, such as NF-κB signaling, further amplifies immune activation, linking lipid dysregulation to neuroinflammatory disorders (Xu et al., 2024; Li et al., 2022).

3.4.3 ABCA1

ABCA1 is a key lipid transporter responsible for cholesterol and phospholipid efflux, playing a vital role in LD homeostasis and inflammatory regulation (Kotlyarov, 2021). Inflammatory stimuli downregulate ABCA1 expression, leading to impaired lipid clearance, increased LD accumulation, and exacerbated immune activation (Wang and Westerterp, 2020). In contrast, enhancing ABCA1 function promotes lipid efflux, reducing inflammatory lipid species and protecting against neuroinflammatory damage (Karasinska et al., 2013). ABCA1 also influences microglial polarization by modulating the balance between pro-inflammatory M1 and anti-inflammatory M2 phenotypes, demonstrating its role in lipid-mediated immune regulation (Reid, 2023; Wei et al., 2024).

3.4.4 PLINs

Perilipins (PLINs) are structural proteins that coat LDs and regulate lipid storage, hydrolysis, and signaling (Chandrasekaran et al., 2024). Among them, PLIN2 and PLIN3 are highly induced during inflammation, promoting LD expansion and stabilizing lipid storage under oxidative stress (Chandrasekaran et al., 2024; Griseti et al., 2024). PLIN5, in contrast, plays a role in mitochondrial-lipid interactions, facilitating lipid utilization to prevent excessive accumulation (Dabravolski et al., 2021). Dysregulation of PLIN proteins during chronic inflammation leads to lipid overload, increased ROS production, and heightened immune activation (Bombarda-Rocha et al., 2023). Studies suggest that targeting PLIN-regulated LD dynamics may help modulate inflammation in neurodegenerative conditions.

3.4.5 ApoE

ApoE is a critical lipid transport protein in the brain that influences LD metabolism and inflammation (Husain et al., 2021). ApoE facilitates cholesterol and phospholipid transport between cells, regulating lipid availability for LD biogenesis (Hauser et al., 2011). However, ApoE4, a genetic variant linked to neurodegenerative diseases, is associated with altered lipid metabolism, impaired LD function, and heightened inflammatory responses. ApoE deficiency or dysfunction exacerbates neuroinflammation by promoting lipid accumulation, reducing lipid clearance efficiency, and amplifying microglial activation (Montagne et al., 2020; Husain et al., 2021; Ayyubova, 2024). These findings highlight ApoE’s essential role in balancing lipid metabolism and immune regulation in the CNS.

3.4.6 LXRs

LXRs are nuclear receptors that regulate cholesterol homeostasis, lipid transport, and inflammatory gene expression (Wang and Tontonoz, 2018). LXRs promote ABCA1-mediated lipid efflux, reducing LD accumulation and mitigating inflammatory stress (Gu et al., 2019; Wu et al., 2019). Additionally, LXRs suppress pro-inflammatory cytokine production through direct transcriptional repression of NF-κB target genes, establishing a protective mechanism against excessive immune activation (A-González and Castrillo, 2011; Zhao et al., 2021). Pharmacological activation of LXRs has been proposed as a therapeutic strategy to enhance lipid metabolism and counteract inflammation in neurodegenerative diseases (Xu et al., 2013; Fitz et al., 2019). However, the precise role of LXRs in different CNS cell types remains an area of active investigation.

4 Lipid metabolism is involved in regulating inflammation in neurological disorders

4.1 Stroke

Stroke, including ischemic and hemorrhagic subtypes, triggers a complex cascade of inflammatory responses that significantly impact neuronal survival, brain repair, and long-term recovery (Reid, 2023). In stroke, particularly ischemic stroke, lipid metabolism exerts a dual role in neuroprotection and neurotoxicity by modulating inflammatory responses (Xu et al., 2021), immune cell activation, oxidative stress, and maintaining BBB integrity through phospholipid and sphingolipid pathways (Sun et al., 2016; Zhao et al., 2022; Sandoval and Witt, 2008), whose disruption exacerbates vascular permeability, brain edema, and neuronal injury (Sandoval and Witt, 2008; Arbaizar-Rovirosa et al., 2023). Dysregulated lipid metabolism in stroke can either exacerbate neuroinflammation and BBB disruption or facilitate resolution of inflammation and neuroprotection, making it a crucial factor in stroke pathophysiology (Zhang et al., 2025; Bernardo-Castro et al., 2020; Yang et al., 2019; Jiang et al., 2021; Janssen et al., 2021; Cui et al., 2021) (Table 1).

Table 1
www.frontiersin.org

Table 1. Preclinical studies assessing the effect of lipids on neuroinflammation or BBB function in stroke.

During stroke, lipid metabolism is profoundly altered due to changes in oxygen availability, cellular stress, and inflammatory signaling (Wei et al., 2024; Adibhatla and Hatcher, 2008). One of the key lipid pathways involved is arachidonic acid metabolism, which generates pro-inflammatory mediators such as prostaglandins and leukotrienes (Wang et al., 2021; Haley et al., 2020). Following ischemic stroke, activation of COX-2 and LOXs leads to excessive production of prostaglandins and leukotrienes, which amplify microglial activation, increase cytokine release, and promote further neuronal injury (Kloska et al., 2020; Ahmad et al., 2009). In contrast, metabolites derived from omega-3 PUFAs, such as resolvins, protectins, and maresins, counteract inflammation and promote resolution by inhibiting leukocyte infiltration and reducing oxidative damage. Cholesterol metabolism also plays a key role in stroke-induced inflammation (Ament et al., 2024). ABCA1 and LXRs regulate cholesterol efflux from microglia and astrocytes, reducing pro-inflammatory lipid accumulation (Lei et al., 2025). However, stroke-induced metabolic stress downregulates ABCA1/LXR signaling, leading to cholesterol buildup, LD formation, and chronic neuroinflammation. Enhancing LXR activation has been shown to mitigate neuroinflammation and improve functional recovery post-stroke by promoting anti-inflammatory lipid mediator production (Zeng et al., 2020). In hemorrhagic stroke, lipid peroxidation is a major driver of neuroinflammation (Wu et al., 2024; Karuppagounder et al., 2018). Excessive release of free iron from blood degradation products catalyzes lipid oxidation, leading to the accumulation of ROS and toxic lipid peroxidation byproducts such as 4-HNE and MDA (Karuppagounder et al., 2018). These oxidative lipid derivatives induce neuronal apoptosis, activate inflammatory pathways, and exacerbate secondary injury. Additionally, microglia and macrophages surrounding hematomas exhibit altered lipid metabolism, shifting toward a pro-inflammatory M1 phenotype, which sustains local inflammation and hampers recovery (Wei et al., 2024; Xin et al., 2023; Pan et al., 2024).

Stroke-induced inflammation severely compromises BBB integrity, leading to increased permeability, immune cell infiltration, and cerebral edema. Lipid metabolism significantly influences BBB function through various mechanisms (Pifferi et al., 2021). Phospholipids and sphingolipids, essential components of the BBB, undergo extensive remodeling during stroke. In ischemic stroke, activation of PLA2 leads to excessive phospholipid hydrolysis, generating pro-inflammatory lipid mediators that weaken tight junction proteins (Muralikrishna Adibhatla and Hatcher, 2006). Additionally, sphingolipid metabolism, particularly S1P signaling, regulates endothelial barrier function (Fan et al., 2022). While S1P promotes endothelial stability under physiological conditions, its dysregulation post-stroke contributes to BBB breakdown and leukocyte infiltration (Fan et al., 2022). S1P receptor modulators, such as fingolimod, have shown potential in reducing BBB disruption and neuroinflammation in preclinical stroke models (Nakagawa and Aruga, 2020). Furthermore, LDs have emerged as key players in BBB regulation. Endothelial cells accumulate LDs in response to ischemic stress, potentially serving as energy reservoirs to support cellular survival (Nitzsche et al., 2021). However, excessive LDs accumulation due to impaired lipid metabolism can lead to endothelial dysfunction and heightened BBB permeability. Inflammatory cytokines, particularly TNF-α and IL-1β, further disrupt lipid homeostasis in BBB cells, exacerbating lipid oxidation and structural damage.

4.2 Alzheimer’s disease

Alzheimer’s disease (AD) is characterized by progressive neurodegeneration, neuroinflammation, and the accumulation of Aβ and tau pathology. Lipid metabolism is increasingly recognized as a key player in AD pathogenesis, influencing inflammatory responses and BBB integrity (Table 2). In AD, lipid metabolism disturbances contribute to amyloid-beta (Aβ) aggregation and tau hyperphosphorylation, two key pathological hallmarks of the disease (Rajmohan and Reddy, 2017). Cholesterol plays a particularly crucial role in modulating amyloid precursor protein (APP) processing, with elevated cholesterol levels promoting the amyloidogenic pathway, leading to increased Aβ production (Maulik et al., 2013). Moreover, ApoE, the primary lipid transporter in the brain, significantly affects lipid homeostasis and Aβ clearance, with the ApoE4 allele being the strongest genetic risk factor for late-onset AD (Husain et al., 2021). Oxidized lipids and lipid peroxidation products further exacerbate neuroinflammation and oxidative stress, accelerating disease progression (Ayyubova, 2024). Neuroinflammation in AD is largely driven by activated microglia and astrocytes, which respond to Aβ plaques by releasing pro-inflammatory cytokines. Lipid metabolism profoundly affects microglial activation states and their ability to clear Aβ aggregates. Triggering Receptor Expressed on TREM2 is crucial for microglial function and lipid uptake. Loss-of-function mutations in TREM2 impair microglial lipid metabolism, reducing Aβ phagocytosis and exacerbating inflammatory responses (Nugent et al., 2020). Furthermore, dysfunctional cholesterol metabolism, mediated by ApoE, particularly the ApoE4 variant, disrupts lipid homeostasis, leading to excessive LDs accumulation in microglia and astrocytes (Qi et al., 2021). This lipid overload induces oxidative stress and sustains a pro-inflammatory microglial phenotype, further contributing to synaptic dysfunction and neuronal loss. Fatty acid metabolism is another key regulator of inflammation in AD. Dysregulated omega-6/omega-3 PUFA balance leads to an overproduction of pro-inflammatory lipid mediators, such as prostaglandins and leukotrienes, which perpetuate neuroinflammation (Devassy et al., 2016; Ma et al., 2020). In contrast, bioactive lipid metabolites derived from DHA, such as resolvins and protectins, have neuroprotective and anti-inflammatory properties (Siqueira et al., 2021). However, AD patients often exhibit reduced DHA levels in the brain, contributing to a pro-inflammatory milieu. Cholesterol metabolism also modulates inflammatory responses in AD. LXRs regulate cholesterol transporters like ABCA1, facilitate lipid efflux and maintain microglial homeostasis (Zhang et al., 2015). However, in AD, LXR signaling is often impaired, leading to cholesterol accumulation, LD formation, and sustained inflammatory activation. Enhancing LXR activity has been shown to promote anti-inflammatory responses and improve lipid metabolism, making it a promising therapeutic target.

Table 2
www.frontiersin.org

Table 2. Preclinical studies assessing the effect of lipids on BBB function in AD.

BBB dysfunction is an early pathological feature of AD, contributing to impaired Aβ clearance and increased neuroinflammation. Lipid metabolism significantly influences BBB integrity through various mechanisms, including tight junction regulation, endothelial lipid composition, and lipid-mediated signaling pathways. Phospholipid and sphingolipid metabolism play crucial roles in maintaining BBB stability. PLA2 activation in AD leads to excessive phospholipid degradation, weakening endothelial tight junctions and increasing vascular permeability (Doody et al., 2015; Kerman et al., 2022). Additionally, alterations in S1P signaling affect endothelial barrier function, facilitating immune cell infiltration and chronic inflammation. Cholesterol dysregulation also contributes to BBB breakdown in AD (McManus et al., 2017). The ApoE4 isoform is associated with reduced lipid transport efficiency, leading to cholesterol and phospholipid accumulation in BBB endothelial cells (Liu et al., 2022). This lipid imbalance disrupts endothelial function, enhances oxidative stress, and weakens BBB integrity. Furthermore, excessive lipid peroxidation, driven by ROS and toxic lipid aldehydes, exacerbates vascular damage and neuroinflammation (Moulton et al., 2021). LDs in BBB endothelial cells have recently been implicated in AD pathogenesis. Under inflammatory conditions, LD accumulation in endothelial cells can serve as an adaptive response to oxidative stress (Lee et al., 2017). However, excessive LD storage due to dysregulated lipid metabolism leads to endothelial dysfunction, impairing BBB repair mechanisms.

4.3 Parkinson’s disease

Parkinson’s disease (PD) is a neurodegenerative disorder characterized by dopaminergic neuronal loss, chronic neuroinflammation, and BBB dysfunction, with lipid metabolism and LDs playing critical roles in these pathological processes (Table 3). In PD, altered lipid metabolism has been linked to α-synuclein pathology, mitochondrial dysfunction, and neuroinflammation (Alecu and Bennett, 2019). Lipids such as polyunsaturated fatty acids (PUFAs) are prone to peroxidation, generating toxic lipid-derived radicals that contribute to dopaminergic neuron loss. Sphingolipid dysregulation, particularly changes in ceramide and glucosylceramide levels, has also been associated with lysosomal dysfunction and impaired autophagy, which are critical in PD pathogenesis (Mesbahi, 2023). Dysregulated lipid metabolism exacerbates neuroinflammation through the accumulation of lipid peroxidation products such as 4-HNE, which interact with α-synuclein to promote its aggregation and toxicity (Di Maio et al., 2023). Disruptions in cholesterol metabolism, particularly in the LXR/ABCA1 signaling pathway, impair lipid efflux, leading to LD accumulation in microglia and astrocytes, which sustains a pro-inflammatory state and amplifies the release of cytokines such as TNF-α, IL-1β, and IL-6, further contributing to neuronal damage (Marchi et al., 2019; Wouters et al., 2019). Although LDs initially serve as protective organelles against oxidative stress, their excessive accumulation within microglia is associated with persistent inflammation and impaired lipid clearance, exacerbating neuronal injury (Lee et al., 2017; Alarcon-Gil et al., 2022; Han et al., 2018). Similarly, BBB integrity is highly dependent on proper lipid metabolism, and its dysfunction in PD is linked to altered phospholipid and sphingolipid homeostasis (Wang et al., 2020). Overactivation of PLA2 weakens endothelial membranes and disrupts tight junctions, increasing BBB permeability and facilitating the entry of peripheral immune cells and inflammatory mediators into the brain, which in turn worsens neuroinflammation (Bate and Williams, 2015; Wu et al., 2021). Additionally, LDs within BBB endothelial cells may act as a temporary defense mechanism against oxidative damage, but prolonged lipid dysregulation contributes to endothelial dysfunction, impairing BBB repair mechanisms and further increasing barrier permeability (Wang et al., 2020). Collectively, these findings suggest that lipid metabolism and LD dynamics are central to PD pathogenesis, influencing both neuroinflammation and BBB function (Xie et al., 2020). Targeting lipid metabolic pathways, including cholesterol homeostasis, LD turnover, and phospholipid signaling, may offer novel therapeutic strategies to mitigate neuroinflammation and protect BBB integrity in PD (More et al., 2017; Takabe et al., 2016; Urano et al., 2022).

Table 3
www.frontiersin.org

Table 3. Preclinical studies assessing the effect of lipids on neuroinflammation or BBB function in PD.

4.4 Other neurological disorders

Emerging research indicates that lipid metabolism and LDs play significant roles in neuroinflammation and BBB integrity across various neurodegenerative diseases, including multiple sclerosis (MS), Huntington’s disease (HD), and amyotrophic lateral sclerosis (ALS) (Table 4). In MS, disturbances in lipid metabolism have been linked to disease progression (De Nuccio et al., 2011). Lipid metabolism is essential for myelin synthesis and maintenance, and disruptions in lipid homeostasis contribute to demyelination and neuroinflammation (López-Muguruza and Matute, 2023). Oligodendrocytes rely on lipid biosynthesis to generate myelin sheaths, and their dysfunction leads to impaired remyelination in MS lesions (López-Muguruza and Matute, 2023). Moreover, lipid mediators such as eicosanoids and sphingolipids play pivotal roles in immune cell activation and neuroinflammation, influencing disease progression (Podbielska et al., 2021). Notably, increased levels of cholesteryl esters have been observed in MS patients, suggesting a potential role in the disease’s pathology. These alterations in lipid composition may influence inflammatory processes and impact BBB integrity, thereby contributing to the neuroinflammatory environment characteristic of MS (Lorincz et al., 2024; Fellows et al., 2015). In HD, lipid abnormalities have been observed in both the CNS and peripheral tissues, with changes in cholesterol homeostasis affecting synaptic function and neuronal viability (Leoni and Caccia, 2015). The mutant huntingtin protein disrupts lipid metabolism by impairing SREBPs, leading to reduced cholesterol biosynthesis and altered phospholipid composition, which may exacerbate neuronal dysfunction and degeneration (González-Guevara et al., 2020). HD is also associated with significant lipid metabolic disturbances. Elevated cholesteryl ester levels have been detected in specific brain regions, such as the caudate and putamen, of HD patients (Phillips et al., 2020). This accumulation may affect neuronal function and viability, potentially exacerbating neuroinflammation and compromising BBB integrity (Joshi et al., 2019). The presence of increased cholesteryl esters underscores the importance of lipid metabolism in HD pathology. In ALS, lipid metabolism alterations have been implicated in motor neuron degeneration, energy metabolism deficits, and neuroinflammation (D'Amico et al., 2021). ALS patients often exhibit hypermetabolism and dyslipidemia, with increased lipid oxidation and altered fatty acid composition in motor neurons (D'Amico et al., 2021). Dysfunctional LDs in astrocytes and microglia also contribute to oxidative stress and inflammatory responses, aggravating disease pathology. While direct evidence linking LDs and lipid metabolism to neuroinflammation and BBB dysfunction in ALS is less extensive, metabolic alterations have been observed in ALS patients (Díaz et al., 2024; Mesev et al., 2017). These metabolic changes could influence lipid processing and storage, potentially affecting inflammatory responses and BBB function. Further research is necessary to elucidate the specific roles of lipid metabolism and LDs in ALS pathology.

Table 4
www.frontiersin.org

Table 4. Preclinical studies assessing the effect of lipids on neuroinflammation or BBB function in other neurological disorders.

5 Applications in neurological disorders’ therapy

5.1 Regulating lipid metabolism to improve neurological disorders

Lipid metabolism, LDs, and liposomes have emerged as significant therapeutic targets in the treatment of neurological disorders (Gigliobianco et al., 2019; Mondal and Ghosh, 2023). Modulating lipid homeostasis and LD dynamics offers promising strategies for alleviating neuroinflammation, supporting neuronal survival, and restoring brain homeostasis in conditions such as AD, PD, and MS (Alecu and Bennett, 2019; Chiurchiù et al., 2022; Kang and Rivest, 2012). For instance, enhancing the activity of enzymes involved in lipid biosynthesis, such as LXRs and ABCA1, may promote cholesterol efflux, reduce lipid accumulation in glial cells, and thereby alleviate neuroinflammation (Kang and Rivest, 2012). Additionally, targeting lipid signaling pathways, such as the SREBPs and TREM2, may help control LD formation and mitigate chronic inflammatory responses in microglia and astrocytes, two critical cell types involved in neuroinflammation (Wang et al., 2025; Loving and Bruce, 2020). LDs, as intracellular lipid storage organelles, play a pivotal role in cellular energy balance and inflammation regulation. Excessive LD accumulation and lipid peroxidation are characteristic features of several neurodegenerative diseases, and regulating LD metabolism may help prevent oxidative stress and chronic neuroinflammation (Yin, 2023). Particularly in diseases like AD, microglial LD accumulation has been linked to prolonged inflammatory states and neuronal damage. Pharmacological interventions targeting LD-associated proteins, such as perilipins, may provide novel therapeutic opportunities by limiting inflammation and promoting tissue repair (Mi et al., 2023; Simpson and Oliver, 2020; Vigouroux et al., 2011).

5.2 Lipid particles for drug delivery across the blood–brain barrier

Liposomes, as lipid-based nanocarriers, are increasingly being explored as drug delivery systems for neurological disorders (Vieira and Gamarra, 2016). Their ability to encapsulate hydrophobic drugs, protect them from degradation, and facilitate targeted delivery across the BBB makes them ideal candidates for treating brain diseases (Table 5). Liposomes can be designed to encapsulate anti-inflammatory agents, neuroprotective compounds, or gene therapies, providing sustained and localized treatment in the brain (Wu et al., 2023; Cascione et al., 2020; Kahana et al., 2021; Nong et al., 2024; Al-Ahmady et al., 2019; Lee et al., 2014; Yang et al., 2018; Ediriweera et al., 2025). Notably, liposomal formulations of compounds such as curcumin and cannabinoids have demonstrated anti-inflammatory and neuroprotective potential in models of AD and PD (Alecu and Bennett, 2019; Pandian et al., 2022; Zhang et al., 2018). Additionally, liposomes can be engineered to target specific cell types within the brain, such as neurons, astrocytes, and microglia, enabling tailored therapeutic approaches (Kahana et al., 2021; Pandian et al., 2022; Moreira et al., 2024; Sela et al., 2023). Liposomes can also deliver lipid-lowering agents or molecules that modulate lipid metabolism to restore proper lipid homeostasis in the brain, potentially alleviating neuroinflammation and improving cognitive function (Torres et al., 2021; Campos-Peña et al., 2022).

Table 5
www.frontiersin.org

Table 5. Liposome-based nano drugs for CNS diseases and their status.

6 Conclusions and prospects

Lipid metabolism and LDs play central roles in regulating neuroinflammation and maintaining BBB integrity in neurological disorders. Dysregulation of lipid homeostasis contributes to chronic inflammation, neuronal damage, and BBB dysfunction, exacerbating diseases like stroke, AD, PD, and MS. Targeting lipid metabolic pathways, modulating LD dynamics, and utilizing lipid-based nanocarriers like liposomes offer promising therapeutic strategies to mitigate inflammation, restore lipid balance, and improve drug delivery to the brain. Future research should focus on developing specific interventions that regulate lipid metabolism and LD formation, offering new avenues for treating neurodegenerative diseases and enhancing therapeutic efficacy.

Author contributions

LF: Conceptualization, Data curation, Investigation, Resources, Validation, Visualization, Writing – original draft, Writing – review & editing. TL: Data curation, Investigation, Writing – original draft, Writing – review & editing. ZH: Data curation, Investigation, Validation, Writing – original draft, Writing – review & editing. YP: Data curation, Investigation, Writing – original draft, Writing – review & editing. WX: Conceptualization, Data curation, Writing – original draft, Writing – review & editing. LZ: Conceptualization, Data curation, Investigation, Writing – original draft, Writing – review & editing. ZL: Writing – original draft, Writing – review & editing. HZ: Data curation, Validation, Writing – original draft, Writing – review & editing. HL: Conceptualization, Funding acquisition, Investigation, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing. WW: Conceptualization, Data curation, Funding acquisition, Investigation, Methodology, Project administration, Resources, Software, Supervision, Validation, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that no financial support was received for the research and/or publication of this article.

Acknowledgments

We thank the support of the China Scholarship Council for YP, WW, and WX.

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.

Generative AI statement

The authors declare that no Gen AI was used in the creation of this manuscript.

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.

References

Adibhatla, R. M., and Hatcher, J. F. (2008). Altered lipid metabolism in brain injury and disorders. Subcell. Biochem. 49, 241–268. doi: 10.1007/978-1-4020-8831-5_9

PubMed Abstract | Crossref Full Text | Google Scholar

Adibhatla, R. M., and Hatcher, J. F. (2010). Lipid oxidation and peroxidation in CNS health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 12, 125–169. doi: 10.1089/ars.2009.2668

PubMed Abstract | Crossref Full Text | Google Scholar

A-González, N., and Castrillo, A. (2011). Liver X receptors as regulators of macrophage inflammatory and metabolic pathways. Biochim. Biophys. Acta 1812, 982–994. doi: 10.1016/j.bbadis.2010.12.015

PubMed Abstract | Crossref Full Text | Google Scholar

Agrawal, N. (2025). A comprehensive review on the advancements of dual COX-2/5-LOX inhibitors as anti-inflammatory drugs. Chem. Biol. Drug Des. 105:e70114. doi: 10.1111/cbdd.70114

PubMed Abstract | Crossref Full Text | Google Scholar

Ahluwalia, K., Ebright, B., Chow, K., Dave, P., Mead, A., Poblete, R., et al. (2022). Lipidomics in understanding pathophysiology and pharmacologic effects in inflammatory diseases: considerations for drug development. Meta 12:333. doi: 10.3390/metabo12040333

PubMed Abstract | Crossref Full Text | Google Scholar

Ahmad, M., Zhang, Y., Liu, H., Rose, M. E., and Graham, S. H. (2009). Prolonged opportunity for neuroprotection in experimental stroke with selective blockade of cyclooxygenase-2 activity. Brain Res. 1279, 168–173. doi: 10.1016/j.brainres.2009.05.020

PubMed Abstract | Crossref Full Text | Google Scholar

Al-Ahmady, Z. S., Jasim, D., Ahmad, S. S., Wong, R., Haley, M., Coutts, G., et al. (2019). Selective liposomal transport through blood brain barrier disruption in ischemic stroke reveals two distinct therapeutic opportunities. ACS Nano 13, 12470–12486. doi: 10.1021/acsnano.9b01808

PubMed Abstract | Crossref Full Text | Google Scholar

Alarcon-Gil, J., Sierra-Magro, A., Morales-Garcia, J. A., Sanz-SanCristobal, M., Alonso-Gil, S., Cortes-Canteli, M., et al. (2022). Neuroprotective and anti-inflammatory effects of linoleic acid in models of Parkinson's disease: the implication of lipid droplets and lipophagy. Cells 11:2297. doi: 10.3390/cells11152297

PubMed Abstract | Crossref Full Text | Google Scholar

Alecu, I., and Bennett, S. A. L. (2019). Dysregulated lipid metabolism and its role in α-synucleinopathy in Parkinson's disease. Front. Neurosci. 13:328. doi: 10.3389/fnins.2019.00328

PubMed Abstract | Crossref Full Text | Google Scholar

Ament, Z., Patki, A., Bhave, V. M., Kijpaisalratana, N., Jones, A. C., Couch, C. A., et al. (2024). Omega-3 fatty acids and risk of ischemic stroke in REGARDS. Transl. Stroke Res. 16, 747–756. doi: 10.1007/s12975-024-01256-7

PubMed Abstract | Crossref Full Text | Google Scholar

Aoki, Y., Dai, H., Furuta, F., Akamatsu, T., Oshima, T., Takahashi, N., et al. (2023). LOX-1 mediates inflammatory activation of microglial cells through the p38-MAPK/NF-κB pathways under hypoxic-ischemic conditions. Cell Commun. Signal 21:126. doi: 10.1186/s12964-023-01048-w

PubMed Abstract | Crossref Full Text | Google Scholar

Arbaizar-Rovirosa, M., Pedragosa, J., Lozano, J. J., Casal, C., Pol, A., Gallizioli, M., et al. (2023). Aged lipid-laden microglia display impaired responses to stroke. EMBO Mol. Med. 15:e17175. doi: 10.15252/emmm.202217175

PubMed Abstract | Crossref Full Text | Google Scholar

Arsenault, E. J., McGill, C. M., and Barth, B. M. (2021). Sphingolipids as regulators of neuro-inflammation and NADPH oxidase 2. NeuroMolecular Med. 23, 25–46. doi: 10.1007/s12017-021-08646-2

PubMed Abstract | Crossref Full Text | Google Scholar

Artru, F., McPhail, M. J. W., Triantafyllou, E., and Trovato, F. M. (2022). Lipids in liver failure syndromes: a focus on eicosanoids, specialized pro-resolving lipid mediators and lysophospholipids. Front. Immunol. 13:867261. doi: 10.3389/fimmu.2022.867261

PubMed Abstract | Crossref Full Text | Google Scholar

Ayyubova, G. (2024). APOE4 is a risk factor and potential therapeutic target for Alzheimer's disease. CNS Neurol. Disord. Drug Targets 23, 342–352. doi: 10.2174/1871527322666230303114425

PubMed Abstract | Crossref Full Text | Google Scholar

Bate, C., and Williams, A. (2015). α-Synuclein-induced synapse damage in cultured neurons is mediated by cholesterol-sensitive activation of cytoplasmic phospholipase A2. Biomolecules 5, 178–193. doi: 10.3390/biom5010178

PubMed Abstract | Crossref Full Text | Google Scholar

Bernardo-Castro, S., Sousa, J. A., Brás, A., Cecília, C., Rodrigues, B., Almendra, L., et al. (2020). Pathophysiology of blood-brain barrier permeability throughout the different stages of ischemic stroke and its implication on hemorrhagic transformation and recovery. Front. Neurol. 11:594672. doi: 10.3389/fneur.2020.594672

PubMed Abstract | Crossref Full Text | Google Scholar

Bombarda-Rocha, V., Silva, D., Badr-Eddine, A., Nogueira, P., Gonçalves, J., and Fresco, P. (2023). Challenges in pharmacological intervention in perilipins (PLINs) to modulate lipid droplet dynamics in obesity and Cancer. Cancers (Basel) 15:4013. doi: 10.3390/cancers15154013

PubMed Abstract | Crossref Full Text | Google Scholar

Bravo, G., Cedeño, R. R., Casadevall, M. P., and Ramió-Torrentà, L. (2022). Sphingosine-1-phosphate (S1P) and S1P signaling pathway modulators, from current insights to future perspectives. Cells 11:2058. doi: 10.3390/cells11132058

PubMed Abstract | Crossref Full Text | Google Scholar

Brinkmann, V. (2009). FTY720 (fingolimod) in multiple sclerosis: therapeutic effects in the immune and the central nervous system. Br. J. Pharmacol. 158, 1173–1182. doi: 10.1111/j.1476-5381.2009.00451.x

PubMed Abstract | Crossref Full Text | Google Scholar

Bruch, G. E., Fernandes, L. F., Bassi, B. L. T., Alves, M. T. R., Pereira, I. O., Frézard, F., et al. (2019). Liposomes for drug delivery in stroke. Brain Res. Bull. 152, 246–256. doi: 10.1016/j.brainresbull.2019.07.015

PubMed Abstract | Crossref Full Text | Google Scholar

Buckley, C. D., Gilroy, D. W., and Serhan, C. N. (2014). Proresolving lipid mediators and mechanisms in the resolution of acute inflammation. Immunity 40, 315–327. doi: 10.1016/j.immuni.2014.02.009

PubMed Abstract | Crossref Full Text | Google Scholar

Campos-Peña, V., Pichardo-Rojas, P., Sánchez-Barbosa, T., Ortíz-Islas, E., Rodríguez-Pérez, C. E., Montes, P., et al. (2022). Amyloid β, lipid metabolism, basal cholinergic system, and therapeutics in Alzheimer's disease. Int. J. Mol. Sci. 23:12092. doi: 10.3390/ijms232012092

PubMed Abstract | Crossref Full Text | Google Scholar

Candore, G., Bulati, M., Caruso, C., Castiglia, L., Colonna-Romano, G., di Bona, D., et al. (2010). Inflammation, cytokines, immune response, apolipoprotein E, cholesterol, and oxidative stress in Alzheimer disease: therapeutic implications. Rejuvenation Res. 13, 301–313. doi: 10.1089/rej.2009.0993

PubMed Abstract | Crossref Full Text | Google Scholar

Cartier, A., and Hla, T. (2019). Sphingosine 1-phosphate: lipid signaling in pathology and therapy. Science 366:eaar5551. doi: 10.1126/science.aar5551

PubMed Abstract | Crossref Full Text | Google Scholar

Cascione, M., de Matteis, V., Leporatti, S., and Rinaldi, R. (2020). The new frontiers in neurodegenerative diseases treatment: liposomal-based strategies. Front. Bioeng. Biotechnol. 8:566767. doi: 10.3389/fbioe.2020.566767

PubMed Abstract | Crossref Full Text | Google Scholar

Chan, J. P., Wong, B. H., Chin, C. F., Galam, D. L. A., Foo, J. C., Wong, L. C., et al. (2018). The lysolipid transporter Mfsd2a regulates lipogenesis in the developing brain. PLoS Biol. 16:e2006443. doi: 10.1371/journal.pbio.2006443

PubMed Abstract | Crossref Full Text | Google Scholar

Chandrasekaran, P., Weiskirchen, S., and Weiskirchen, R. (2024). Perilipins: a family of five fat-droplet storing proteins that play a significant role in fat homeostasis. J. Cell. Biochem. 125:e30579. doi: 10.1002/jcb.30579

PubMed Abstract | Crossref Full Text | Google Scholar

Chang, T. Y., Chang, C. C. Y., Lin, S., Yu, C., Li, B. L., and Miyazaki, A. (2001). Roles of acyl-coenzyme A:cholesterol acyltransferase-1 and -2. Curr. Opin. Lipidol. 12, 289–296. doi: 10.1097/00041433-200106000-00008

Crossref Full Text | Google Scholar

Chen, T., Dai, Y., Hu, C., Lin, Z., Wang, S., Yang, J., et al. (2024). Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 21:60. doi: 10.1186/s12987-024-00557-1

PubMed Abstract | Crossref Full Text | Google Scholar

Chen, Y. C., Wu, J. S., Tsai, H. D., Huang, C. Y., Chen, J. J., Sun, G. Y., et al. (2012). Peroxisome proliferator-activated receptor gamma (PPAR-γ) and neurodegenerative disorders. Mol. Neurobiol. 46, 114–124. doi: 10.1007/s12035-012-8259-8

PubMed Abstract | Crossref Full Text | Google Scholar

Chiurchiù, V., Tiberi, M., Matteocci, A., Fazio, F., Siffeti, H., Saracini, S., et al. (2022). Lipidomics of bioactive lipids in Alzheimer's and Parkinson's diseases: where are we? Int. J. Mol. Sci. 23:6235. doi: 10.3390/ijms23116235

PubMed Abstract | Crossref Full Text | Google Scholar

Ciancarelli, I., Morone, G., Iosa, M., Cerasa, A., Calabrò, R. S., Iolascon, G., et al. (2022). Influence of oxidative stress and inflammation on nutritional status and neural plasticity: new perspectives on post-stroke neurorehabilitative outcome. Nutrients 15:108. doi: 10.3390/nu15010108

PubMed Abstract | Crossref Full Text | Google Scholar

Clemente-Suárez, V. J., Rubio-Zarapuz, A., Belinchón-deMiguel, P., Beltrán-Velasco, A. I., Martín-Rodríguez, A., and Tornero-Aguilera, J. F. (2024). Impact of physical activity on cellular metabolism across both neurodegenerative and general neurological conditions: a narrative review. Cells 13:1940. doi: 10.3390/cells13231940

PubMed Abstract | Crossref Full Text | Google Scholar

Correale, J., Marrodan, M., and Ysrraelit, M. C. (2019). Mechanisms of neurodegeneration and axonal dysfunction in progressive multiple sclerosis. Biomedicine 7:14. doi: 10.3390/biomedicines7010014

PubMed Abstract | Crossref Full Text | Google Scholar

Correale, J., and Villa, A. (2009). Cellular elements of the blood-brain barrier. Neurochem. Res. 34, 2067–2077. doi: 10.1007/s11064-009-0081-y

PubMed Abstract | Crossref Full Text | Google Scholar

Correia, A. C., Monteiro, A. R., Silva, R., Moreira, J. N., Sousa Lobo, J. M., and Silva, A. C. (2022). Lipid nanoparticles strategies to modify pharmacokinetics of central nervous system targeting drugs: crossing or circumventing the blood-brain barrier (BBB) to manage neurological disorders. Adv. Drug Deliv. Rev. 189:114485. doi: 10.1016/j.addr.2022.114485

PubMed Abstract | Crossref Full Text | Google Scholar

Cui, Y., Zhang, Y., Zhao, X., Shao, L., Liu, G., Sun, C., et al. (2021). ACSL4 exacerbates ischemic stroke by promoting ferroptosis-induced brain injury and neuroinflammation. Brain Behav. Immun. 93, 312–321. doi: 10.1016/j.bbi.2021.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

Dabravolski, S. A., Bezsonov, E. E., Baig, M. S., Popkova, T. V., and Orekhov, A. N. (2021). Mitochondrial lipid homeostasis at the crossroads of liver and heart diseases. Int. J. Mol. Sci. 22:6949. doi: 10.3390/ijms22136949

PubMed Abstract | Crossref Full Text | Google Scholar

Dai, L., Zou, L., Meng, L., Qiang, G., Yan, M., and Zhang, Z. (2021). Cholesterol metabolism in neurodegenerative diseases: molecular mechanisms and therapeutic targets. Mol. Neurobiol. 58, 2183–2201. doi: 10.1007/s12035-020-02232-6

PubMed Abstract | Crossref Full Text | Google Scholar

D'Amico, E., Grosso, G., Nieves, J. W., Zanghì, A., Factor-Litvak, P., and Mitsumoto, H. (2021). Metabolic abnormalities, dietary risk factors and nutritional management in amyotrophic lateral sclerosis. Nutrients 13:2273. doi: 10.3390/nu13072273

PubMed Abstract | Crossref Full Text | Google Scholar

De Nuccio, C., Bernardo, A., De Simone, R., Mancuso, E., Magnaghi, V., Visentin, S., et al. (2011). Peroxisome proliferator-activated receptor γ agonists accelerate oligodendrocyte maturation and influence mitochondrial functions and oscillatory Ca(2+) waves. J. Neuropathol. Exp. Neurol. 70, 900–912. doi: 10.1097/NEN.0b013e3182309ab1

PubMed Abstract | Crossref Full Text | Google Scholar

Devassy, J. G., Leng, S., Gabbs, M., Monirujjaman, M., and Aukema, H. M. (2016). Omega-3 polyunsaturated fatty acids and oxylipins in neuroinflammation and management of Alzheimer disease. Adv. Nutr. 7, 905–916. doi: 10.3945/an.116.012187

PubMed Abstract | Crossref Full Text | Google Scholar

Di Maio, R., Keeney, M. T., Cechova, V., Mortimer, A., Sekandari, A., Rowart, P., et al. (2023). Neuroprotective actions of a fatty acid nitroalkene in Parkinson's disease. NPJ Parkinsons Dis. 9:55. doi: 10.1038/s41531-023-00502-3

PubMed Abstract | Crossref Full Text | Google Scholar

Díaz, M., Fabelo, N., Martín, M. V., Santos, G., and Ferrer, I. (2024). Evidence for alterations in lipid profiles and biophysical properties of lipid rafts from spinal cord in sporadic amyotrophic lateral sclerosis. J. Mol. Med. (Berl) 102, 391–402. doi: 10.1007/s00109-024-02419-7

PubMed Abstract | Crossref Full Text | Google Scholar

Dietschy, J. M., and Turley, S. D. (2004). Thematic review series: brain lipids. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J. Lipid Res. 45, 1375–1397. doi: 10.1194/jlr.R400004-JLR200

PubMed Abstract | Crossref Full Text | Google Scholar

Doody, R. S., Demirovic, J., Ballantyne, C. M., Chan, W., Barber, R., Powell, S., et al. (2015). Lipoprotein-associated phospholipase A2, homocysteine, and Alzheimer's disease. Alzheimers Dement. (Amst) 1, 464–471. doi: 10.1016/j.dadm.2015.08.001

PubMed Abstract | Crossref Full Text | Google Scholar

Dyall, S. C. (2015). Long-chain omega-3 fatty acids and the brain: a review of the independent and shared effects of EPA, DPA and DHA. Front. Aging Neurosci. 7:52. doi: 10.3389/fnagi.2015.00052

Crossref Full Text | Google Scholar

Eberlé, D., Hegarty, B., Bossard, P., Ferré, P., and Foufelle, F. (2004). SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86, 839–848. doi: 10.1016/j.biochi.2004.09.018

PubMed Abstract | Crossref Full Text | Google Scholar

Ediriweera, G. R., Sivaram, A. J., Cowin, G., Brown, M. L., McAlary, L., Lum, J. S., et al. (2025). Lipid nanoparticles and transcranial focused ultrasound enhance the delivery of SOD1 antisense oligonucleotides to the murine brain for ALS therapy. J. Control. Release 378, 221–235. doi: 10.1016/j.jconrel.2024.11.074

PubMed Abstract | Crossref Full Text | Google Scholar

Escandon, P., Vasini, B., Whelchel, A. E., Nicholas, S. E., Matlock, H. G., Ma, J. X., et al. (2021). The role of peroxisome proliferator-activated receptors in healthy and diseased eyes. Exp. Eye Res. 208:108617. doi: 10.1016/j.exer.2021.108617

PubMed Abstract | Crossref Full Text | Google Scholar

Fagone, P., and Jackowski, S. (2009). Membrane phospholipid synthesis and endoplasmic reticulum function. J. Lipid Res. 50, S311–S316. doi: 10.1194/jlr.R800049-JLR200

PubMed Abstract | Crossref Full Text | Google Scholar

Fan, X., Chen, H., Xu, C., Wang, Y., Yin, P., Li, M., et al. (2022). S1PR3, as a core protein related to ischemic stroke, is involved in the regulation of blood-brain barrier damage. Front. Pharmacol. 13:834948. doi: 10.3389/fphar.2022.834948

PubMed Abstract | Crossref Full Text | Google Scholar

Fang, M., Yu, Q., Ou, J., Lou, J., Zhu, J., and Lin, Z. (2024). The neuroprotective mechanisms of PPAR-γ: inhibition of microglia-mediated neuroinflammation and oxidative stress in a neonatal mouse model of hypoxic-ischemic white matter injury. CNS Neurosci. Ther. 30:e70081. doi: 10.1111/cns.70081

PubMed Abstract | Crossref Full Text | Google Scholar

Farmer, B. C., Walsh, A. E., Kluemper, J. C., and Johnson, L. A. (2020). Lipid droplets in neurodegenerative disorders. Front. Neurosci. 14:742. doi: 10.3389/fnins.2020.00742

PubMed Abstract | Crossref Full Text | Google Scholar

Farooqui, A. A. (2012). N-3 fatty acid-derived lipid mediators in the brain: new weapons against oxidative stress and inflammation. Curr. Med. Chem. 19, 532–543. doi: 10.2174/092986712798918851

PubMed Abstract | Crossref Full Text | Google Scholar

Farooqui, A. A., and Farooqui, T. (2024). Phospholipids, sphingolipids, and cholesterol-derived lipid mediators and their role in neurological disorders. Int. J. Mol. Sci. 25:10672. doi: 10.3390/ijms251910672

PubMed Abstract | Crossref Full Text | Google Scholar

Fellows, K., Uher, T., Browne, R. W., Weinstock-Guttman, B., Horakova, D., Posova, H., et al. (2015). Protective associations of HDL with blood-brain barrier injury in multiple sclerosis patients. J. Lipid Res. 56, 2010–2018. doi: 10.1194/jlr.M060970

PubMed Abstract | Crossref Full Text | Google Scholar

Fidaleo, M., Fanelli, F., Ceru, M., and Moreno, S. (2014). Neuroprotective properties of peroxisome proliferator-activated receptor alpha (PPARα) and its lipid ligands. Curr. Med. Chem. 21, 2803–2821. doi: 10.2174/0929867321666140303143455

PubMed Abstract | Crossref Full Text | Google Scholar

Fitz, N. F., Nam, K. N., Koldamova, R., and Lefterov, I. (2019). Therapeutic targeting of nuclear receptors, liver X and retinoid X receptors, for Alzheimer's disease. Br. J. Pharmacol. 176, 3599–3610. doi: 10.1111/bph.14668

PubMed Abstract | Crossref Full Text | Google Scholar

Florance, I., and Ramasubbu, S. (2022). Current understanding on the role of lipids in macrophages and associated diseases. Int. J. Mol. Sci. 24:589. doi: 10.3390/ijms24010589

PubMed Abstract | Crossref Full Text | Google Scholar

Fock, E., and Parnova, R. (2023). Mechanisms of blood-brain barrier protection by microbiota-derived short-chain fatty acids. Cells 12:657. doi: 10.3390/cells12040657

PubMed Abstract | Crossref Full Text | Google Scholar

Fornari Laurindo, L., Aparecido Dias, J., Cressoni Araújo, A., Torres Pomini, K., Machado Galhardi, C., Rucco Penteado Detregiachi, C., et al. (2023). Immunological dimensions of neuroinflammation and microglial activation: exploring innovative immunomodulatory approaches to mitigate neuroinflammatory progression. Front. Immunol. 14:1305933. doi: 10.3389/fimmu.2023.1305933

Crossref Full Text | Google Scholar

Gao, H. M., Chen, H., Cui, G. Y., and Hu, J. X. (2023). Damage mechanism and therapy progress of the blood-brain barrier after ischemic stroke. Cell Biosci. 13:196. doi: 10.1186/s13578-023-01126-z

PubMed Abstract | Crossref Full Text | Google Scholar

Geltinger, F., Schartel, L., Wiederstein, M., Tevini, J., Aigner, E., Felder, T. K., et al. (2020). Friend or foe: lipid droplets as organelles for protein and lipid storage in cellular stress response, aging and disease. Molecules 25:5053. doi: 10.3390/molecules25215053

PubMed Abstract | Crossref Full Text | Google Scholar

Gigliobianco, M. R., di Martino, P., Deng, S., Casadidio, C., and Censi, R. (2019). New advanced strategies for the treatment of lysosomal diseases affecting the central nervous system. Curr. Pharm. Des. 25, 1933–1950. doi: 10.2174/1381612825666190708213159

PubMed Abstract | Crossref Full Text | Google Scholar

González-Guevara, E., Cárdenas, G., Pérez-Severiano, F., and Martínez-Lazcano, J. C. (2020). Dysregulated brain cholesterol metabolism is linked to neuroinflammation in Huntington's disease. Mov. Disord. 35, 1113–1127. doi: 10.1002/mds.28089

PubMed Abstract | Crossref Full Text | Google Scholar

Gouna, G. (2024). Lipid metabolism in remyelination: does a hidden dialogue exist between the central nervous system and the periphery? LMU Munich.

Google Scholar

Griseti, E., Bello, A. A., Bieth, E., Sabbagh, B., Iacovoni, J. S., Bigay, J., et al. (2024). Molecular mechanisms of perilipin protein function in lipid droplet metabolism. FEBS Lett. 598, 1170–1198. doi: 10.1002/1873-3468.14792

PubMed Abstract | Crossref Full Text | Google Scholar

Groves, A., Kihara, Y., and Chun, J. (2013). Fingolimod: direct CNS effects of sphingosine 1-phosphate (S1P) receptor modulation and implications in multiple sclerosis therapy. J. Neurol. Sci. 328, 9–18. doi: 10.1016/j.jns.2013.02.011

PubMed Abstract | Crossref Full Text | Google Scholar

Gu, H. F., Li, N., Xu, Z. Q., Hu, L., Li, H., Zhang, R. J., et al. (2019). Chronic unpredictable mild stress promotes atherosclerosis via HMGB1/TLR4-mediated downregulation of PPARγ/LXRα/ABCA1 in ApoE(−/−) mice. Front. Physiol. 10:165. doi: 10.3389/fphys.2019.00165

PubMed Abstract | Crossref Full Text | Google Scholar

Haley, M. J., White, C. S., Roberts, D., O’Toole, K., Cunningham, C. J., Rivers-Auty, J., et al. (2020). Stroke induces prolonged changes in lipid metabolism, the liver and body composition in mice. Transl. Stroke Res. 11, 837–850. doi: 10.1007/s12975-019-00763-2

PubMed Abstract | Crossref Full Text | Google Scholar

Han, X., Zhu, J., Zhang, X., Song, Q., Ding, J., Lu, M., et al. (2018). Plin4-dependent lipid droplets hamper neuronal mitophagy in the MPTP/p-induced mouse model of Parkinson's disease. Front. Neurosci. 12:397. doi: 10.3389/fnins.2018.00397

PubMed Abstract | Crossref Full Text | Google Scholar

Hauser, P. S., Narayanaswami, V., and Ryan, R. O. (2011). Apolipoprotein E: from lipid transport to neurobiology. Prog. Lipid Res. 50, 62–74. doi: 10.1016/j.plipres.2010.09.001

PubMed Abstract | Crossref Full Text | Google Scholar

Helgudóttir, S. S., Mørkholt, A. S., Lichota, J., Bruun-Nyzell, P., Andersen, M. C., Kristensen, N. M. J., et al. (2024). Rethinking neurodegenerative diseases: neurometabolic concept linking lipid oxidation to diseases in the central nervous system. Neural Regen. Res. 19, 1437–1445. doi: 10.4103/1673-5374.387965

PubMed Abstract | Crossref Full Text | Google Scholar

Hou, J., Chen, Y., Grajales-Reyes, G., and Colonna, M. (2022). TREM2 dependent and independent functions of microglia in Alzheimer's disease. Mol. Neurodegener. 17:84. doi: 10.1186/s13024-022-00588-y

PubMed Abstract | Crossref Full Text | Google Scholar

Hu, Z., Yuan, Y., Tong, Z., Liao, M., Yuan, S., Wu, W., et al. (2023). Aerobic exercise facilitates the nuclear translocation of SREBP2 by activating AKT/SEC24D to contribute cholesterol homeostasis for improving cognition in APP/PS1 mice. Int. J. Mol. Sci. 24:12847. doi: 10.3390/ijms241612847

PubMed Abstract | Crossref Full Text | Google Scholar

Husain, M. A., Laurent, B., and Plourde, M. (2021). APOE and Alzheimer's disease: from lipid transport to physiopathology and therapeutics. Front. Neurosci. 15:630502. doi: 10.3389/fnins.2021.630502

PubMed Abstract | Crossref Full Text | Google Scholar

Huynh, T. N., Fikse, E. N., de la Torre, A. L., Havrda, M. C., Chang, C. C. Y., and Chang, T. Y. (2024). Inhibiting the cholesterol storage enzyme ACAT1/SOAT1 in aging apolipoprotein E4 mice alters their brains' inflammatory profiles. Int. J. Mol. Sci. 25:13690. doi: 10.3390/ijms252413690

PubMed Abstract | Crossref Full Text | Google Scholar

Janssen, L., Ai, X., Zheng, X., Wei, W., Caglayan, A. B., Kilic, E., et al. (2021). Inhibition of fatty acid synthesis aggravates brain injury, reduces blood-brain barrier integrity and impairs neurological recovery in a murine stroke model. Front. Cell. Neurosci. 15:733973. doi: 10.3389/fncel.2021.733973

PubMed Abstract | Crossref Full Text | Google Scholar

Jarc, E., and Petan, T. (2019). Lipid droplets and the management of cellular stress. Yale J. Biol. Med. 92, 435–452.

PubMed Abstract | Google Scholar

Jiang, W., Gong, L., Liu, F., Ren, Y., and Mu, J. (2021). Alteration of gut microbiome and correlated lipid metabolism in post-stroke depression. Front. Cell. Infect. Microbiol. 11:663967. doi: 10.3389/fcimb.2021.663967

PubMed Abstract | Crossref Full Text | Google Scholar

Joshi, A. U., Minhas, P. S., Liddelow, S. A., Haileselassie, B., Andreasson, K. I., Dorn, G. W. II, et al. (2019). Fragmented mitochondria released from microglia trigger A1 astrocytic response and propagate inflammatory neurodegeneration. Nat. Neurosci. 22, 1635–1648. doi: 10.1038/s41593-019-0486-0

PubMed Abstract | Crossref Full Text | Google Scholar

Juhairiyah, F., and de Lange, E. C. M. (2021). Understanding drug delivery to the brain using liposome-based strategies: studies that provide mechanistic insights are essential. AAPS J. 23:114. doi: 10.1208/s12248-021-00648-z

PubMed Abstract | Crossref Full Text | Google Scholar

Kahana, M., Weizman, A., Gabay, M., Loboda, Y., Segal-Gavish, H., Gavish, A., et al. (2021). Liposome-based targeting of dopamine to the brain: a novel approach for the treatment of Parkinson's disease. Mol. Psychiatry 26, 2626–2632. doi: 10.1038/s41380-020-0742-4

PubMed Abstract | Crossref Full Text | Google Scholar

Kang, J., and Rivest, S. (2012). Lipid metabolism and neuroinflammation in Alzheimer's disease: a role for liver X receptors. Endocr. Rev. 33, 715–746. doi: 10.1210/er.2011-1049

PubMed Abstract | Crossref Full Text | Google Scholar

Kapadia, R., Yi, J. H., and Vemuganti, R. (2008). Mechanisms of anti-inflammatory and neuroprotective actions of PPAR-gamma agonists. Front. Biosci. 13, 1813–1826. doi: 10.2741/2802

PubMed Abstract | Crossref Full Text | Google Scholar

Karasinska, J. M., de Haan, W., Franciosi, S., Ruddle, P., Fan, J., Kruit, J. K., et al. (2013). ABCA1 influences neuroinflammation and neuronal death. Neurobiol. Dis. 54, 445–455. doi: 10.1016/j.nbd.2013.01.018

PubMed Abstract | Crossref Full Text | Google Scholar

Karuppagounder, S. S., Alin, L., Chen, Y., Brand, D., Bourassa, M. W., Dietrich, K., et al. (2018). N-acetylcysteine targets 5 lipoxygenase-derived, toxic lipids and can synergize with prostaglandin E(2) to inhibit ferroptosis and improve outcomes following hemorrhagic stroke in mice. Ann. Neurol. 84, 854–872. doi: 10.1002/ana.25356

PubMed Abstract | Crossref Full Text | Google Scholar

Keaney, J., and Campbell, M. (2015). The dynamic blood–brain barrier. FEBS J. 282, 4067–4079. doi: 10.1111/febs.13412

PubMed Abstract | Crossref Full Text | Google Scholar

Kerman, B. E., Duro, M. V. V., Maria, N. S., Liu, X., Wang, S., van Valkenburgh, J. S., et al. (2022). Inhibition of calcium-dependent phospholipase A2 improves blood-brain barrier integrity in ApoE4 carrying mice. Alzheimers Dement. 18:e067937. doi: 10.1002/alz.067937

Crossref Full Text | Google Scholar

Klionsky, D. J., Petroni, G., Amaravadi, R. K., Baehrecke, E. H., Ballabio, A., Boya, P., et al. (2021). Autophagy in major human diseases. EMBO J. 40:e108863. doi: 10.15252/embj.2021108863

PubMed Abstract | Crossref Full Text | Google Scholar

Kloska, A., Malinowska, M., Gabig-Cimińska, M., and Jakóbkiewicz-Banecka, J. (2020). Lipids and lipid mediators associated with the risk and pathology of ischemic stroke. Int. J. Mol. Sci. 21:3618. doi: 10.3390/ijms21103618

PubMed Abstract | Crossref Full Text | Google Scholar

Kotlyarov, S. (2021). Participation of ABCA1 transporter in pathogenesis of chronic obstructive pulmonary disease. Int. J. Mol. Sci. 22:3334. doi: 10.3390/ijms22073334

PubMed Abstract | Crossref Full Text | Google Scholar

Kumar, A., Behl, T., Jamwal, S., Kaur, I., Sood, A., and Kumar, P. (2020). Exploring the molecular approach of COX and LOX in Alzheimer’s and Parkinson’s disorder. Mol. Biol. Rep. 47, 9895–9912. doi: 10.1007/s11033-020-06033-x

PubMed Abstract | Crossref Full Text | Google Scholar

Kuperberg, S. J., and Wadgaonkar, R. (2017). Sepsis-associated encephalopathy: the blood-brain barrier and the sphingolipid rheostat. Front. Immunol. 8:597. doi: 10.3389/fimmu.2017.00597

PubMed Abstract | Crossref Full Text | Google Scholar

Lan, Z. Q., Ge, Z. Y., Lv, S. K., Zhao, B., and Li, C. X. (2023). The regulatory role of lipophagy in central nervous system diseases. Cell Death Discov. 9:229. doi: 10.1038/s41420-023-01504-z

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, L. L., Aung, H. H., Wilson, D. W., Anderson, S. E., Rutledge, J. C., and Rutkowsky, J. M. (2017). Triglyceride-rich lipoprotein lipolysis products increase blood-brain barrier transfer coefficient and induce astrocyte lipid droplets and cell stress. Am. J. Physiol. Cell Physiol. 312, C500–c516. doi: 10.1152/ajpcell.00120.2016

PubMed Abstract | Crossref Full Text | Google Scholar

Lee, D. H., Rötger, C., Appeldoorn, C. C. M., Reijerkerk, A., Gladdines, W., Gaillard, P. J., et al. (2014). Glutathione PEGylated liposomal methylprednisolone (2B3-201) attenuates CNS inflammation and degeneration in murine myelin oligodendrocyte glycoprotein induced experimental autoimmune encephalomyelitis. J. Neuroimmunol. 274, 96–101. doi: 10.1016/j.jneuroim.2014.06.025

PubMed Abstract | Crossref Full Text | Google Scholar

Lei, X., Ran, X., Wang, J., Li, L., Wu, N., Ru, X., et al. (2025). CKN reduces TLR4-mediated inflammation and cerebral I/R injury by activating the LXRα/ABCA1 pathway in microglia. Life Sci. 370:123571. doi: 10.1016/j.lfs.2025.123571

PubMed Abstract | Crossref Full Text | Google Scholar

Leng, F., and Edison, P. (2021). Neuroinflammation and microglial activation in Alzheimer disease: where do we go from here? Nat. Rev. Neurol. 17, 157–172. doi: 10.1038/s41582-020-00435-y

PubMed Abstract | Crossref Full Text | Google Scholar

Leoni, V., and Caccia, C. (2015). The impairment of cholesterol metabolism in Huntington disease. Biochim. Biophys. Acta 1851, 1095–1105. doi: 10.1016/j.bbalip.2014.12.018

PubMed Abstract | Crossref Full Text | Google Scholar

Leyane, T. S., Jere, S. W., and Houreld, N. N. (2022). Oxidative stress in ageing and chronic degenerative pathologies: molecular mechanisms involved in counteracting oxidative stress and chronic inflammation. Int. J. Mol. Sci. 23:7273. doi: 10.3390/ijms23137273

PubMed Abstract | Crossref Full Text | Google Scholar

Li, W., Jin, K., Luo, J., Xu, W., Wu, Y., Zhou, J., et al. (2022). NF-κB and its crosstalk with endoplasmic reticulum stress in atherosclerosis. Front. Cardiovasc. Med. 9:988266. doi: 10.3389/fcvm.2022.988266

PubMed Abstract | Crossref Full Text | Google Scholar

Li, D., Li, S., Xue, A. Z., Smith Callahan, L. A., and Liu, Y. (2020). Expression of SREBP2 and cholesterol metabolism related genes in TCGA glioma cohorts. Medicine (Baltimore) 99:e18815. doi: 10.1097/MD.0000000000018815

PubMed Abstract | Crossref Full Text | Google Scholar

Liu, X., Liu, Z., Wang, X., and Zhao, Y. (2021). Therapeutic effect of multifunctional nano-liposomes on Alzheimer’s disease. Mater. Express 11, 1331–1338. doi: 10.1166/mex.2021.1993

Crossref Full Text | Google Scholar

Liu, C. C., Zhao, J., Fu, Y., Inoue, Y., Ren, Y., Chen, Y., et al. (2022). Peripheral apoE4 enhances Alzheimer's pathology and impairs cognition by compromising cerebrovascular function. Nat. Neurosci. 25, 1020–1033. doi: 10.1038/s41593-022-01127-0

PubMed Abstract | Crossref Full Text | Google Scholar

Lochhead, J. J., Yang, J., Ronaldson, P. T., and Davis, T. P. (2020). Structure, function, and regulation of the blood-brain barrier tight junction in central nervous system disorders. Front. Physiol. 11:914. doi: 10.3389/fphys.2020.00914

PubMed Abstract | Crossref Full Text | Google Scholar

López-Muguruza, E., and Matute, C. (2023). Alterations of oligodendrocyte and myelin energy metabolism in multiple sclerosis. Int. J. Mol. Sci. 24:12912. doi: 10.3390/ijms241612912

PubMed Abstract | Crossref Full Text | Google Scholar

Lorincz, B., Vrablik, M., Murali, R., Havrdova, E. K., Horakova, D., Krasensky, J., et al. (2024). Lipid and brain volumetric measures in multiple sclerosis patients: findings from a large observational study. Acta Neurol. Belg. 124, 1981–1988. doi: 10.1007/s13760-024-02676-w

PubMed Abstract | Crossref Full Text | Google Scholar

Loving, B. A., and Bruce, K. D. (2020). Lipid and lipoprotein metabolism in microglia. Front. Physiol. 11:393. doi: 10.3389/fphys.2020.00393

PubMed Abstract | Crossref Full Text | Google Scholar

Ma, Q. L., Zhu, C., Morselli, M., Su, T., Pelligrini, M., Lu, Z., et al. (2020). The novel Omega-6 fatty acid docosapentaenoic acid positively modulates brain innate immune response for resolving neuroinflammation at early and late stages of humanized APOE-based Alzheimer's disease models. Front. Immunol. 11:558036. doi: 10.3389/fimmu.2020.558036

PubMed Abstract | Crossref Full Text | Google Scholar

Maida, C. D., Norrito, R. L., Daidone, M., Tuttolomondo, A., and Pinto, A. (2020). Neuroinflammatory mechanisms in ischemic stroke: focus on cardioembolic stroke, background, and therapeutic approaches. Int. J. Mol. Sci. 21:6454. doi: 10.3390/ijms21186454

PubMed Abstract | Crossref Full Text | Google Scholar

Marchi, C., Adorni, M. P., Caffarra, P., Ronda, N., Spallazzi, M., Barocco, F., et al. (2019). ABCA1- and ABCG1-mediated cholesterol efflux capacity of cerebrospinal fluid is impaired in Alzheimer's disease. J. Lipid Res. 60, 1449–1456. doi: 10.1194/jlr.P091033

PubMed Abstract | Crossref Full Text | Google Scholar

Marogianni, C., Sokratous, M., Dardiotis, E., Hadjigeorgiou, G. M., Bogdanos, D., and Xiromerisiou, G. (2020). Neurodegeneration and inflammation-an interesting interplay in Parkinson's disease. Int. J. Mol. Sci. 21:8421. doi: 10.3390/ijms21228421

PubMed Abstract | Crossref Full Text | Google Scholar

Maulik, M., Westaway, D., Jhamandas, J. H., and Kar, S. (2013). Role of cholesterol in APP metabolism and its significance in Alzheimer's disease pathogenesis. Mol. Neurobiol. 47, 37–63. doi: 10.1007/s12035-012-8337-y

PubMed Abstract | Crossref Full Text | Google Scholar

McManus, R. M., Finucane, O. M., Wilk, M. M., Mills, K. H. G., and Lynch, M. A. (2017). FTY720 attenuates infection-induced enhancement of aβ accumulation in APP/PS1 mice by modulating astrocytic activation. J. Neuroimmune Pharmacol. 12, 670–681. doi: 10.1007/s11481-017-9753-6

PubMed Abstract | Crossref Full Text | Google Scholar

Mesbahi, H. (2023). Impact of microbiota on neurodegeneration in tauopathies using Caenorhabditis elegans as a model organism. Available online at: https://hdl.handle.net/11375/29312

Google Scholar

Mesev, E. V., Miller, D. S., and Cannon, R. E. (2017). Ceramide 1-phosphate increases P-glycoprotein transport activity at the blood-brain barrier via prostaglandin E2 signaling. Mol. Pharmacol. 91, 373–382. doi: 10.1124/mol.116.107169

PubMed Abstract | Crossref Full Text | Google Scholar

Mi, Y., Qi, G., Vitali, F., Shang, Y., Raikes, A. C., Wang, T., et al. (2023). Loss of fatty acid degradation by astrocytic mitochondria triggers neuroinflammation and neurodegeneration. Nat. Metab. 5, 445–465. doi: 10.1038/s42255-023-00756-4

PubMed Abstract | Crossref Full Text | Google Scholar

Micheli, M. R., Bova, R., Magini, A., Polidoro, M., and Emiliani, C. (2012). Lipid-based nanocarriers for CNS-targeted drug delivery. Recent Pat. CNS Drug Discov. 7, 71–86. doi: 10.2174/157488912798842241

PubMed Abstract | Crossref Full Text | Google Scholar

Mitchell, R. W., and Hatch, G. M. (2011). Fatty acid transport into the brain: of fatty acid fables and lipid tails. Prostaglandins Leukot. Essent. Fatty Acids 85, 293–302. doi: 10.1016/j.plefa.2011.04.007

PubMed Abstract | Crossref Full Text | Google Scholar

Mondal, S., and Ghosh, S. (2023). Liposome-mediated anti-viral drug delivery across blood-brain barrier: can lipid droplet target be game changers? Cell. Mol. Neurobiol. 44:9. doi: 10.1007/s10571-023-01443-4

PubMed Abstract | Crossref Full Text | Google Scholar

Monson, E. A., Trenerry, A. M., Laws, J. L., Mackenzie, J. M., and Helbig, K. J. (2021). Lipid droplets and lipid mediators in viral infection and immunity. FEMS Microbiol. Rev. 45:fuaa066. doi: 10.1093/femsre/fuaa066

PubMed Abstract | Crossref Full Text | Google Scholar

Montagne, A., Nation, D. A., Sagare, A. P., Barisano, G., Sweeney, M. D., Chakhoyan, A., et al. (2020). APOE4 leads to blood-brain barrier dysfunction predicting cognitive decline. Nature 581, 71–76. doi: 10.1038/s41586-020-2247-3

PubMed Abstract | Crossref Full Text | Google Scholar

More, V. R., Campos, C. R., Evans, R. A., Oliver, K. D., Chan, G. N. Y., Miller, D. S., et al. (2017). PPAR-α, a lipid-sensing transcription factor, regulates blood-brain barrier efflux transporter expression. J. Cereb. Blood Flow Metab. 37, 1199–1212. doi: 10.1177/0271678X16650216

PubMed Abstract | Crossref Full Text | Google Scholar

Moreira, R., Nóbrega, C., de Almeida, L. P., and Mendonça, L. (2024). Brain-targeted drug delivery - nanovesicles directed to specific brain cells by brain-targeting ligands. J. Nanobiotechnol. 22:260. doi: 10.1186/s12951-024-02511-7

PubMed Abstract | Crossref Full Text | Google Scholar

Moulton, M. J., Barish, S., Ralhan, I., Chang, J., Goodman, L. D., Harland, J. G., et al. (2021). Neuronal ROS-induced glial lipid droplet formation is altered by loss of Alzheimer's disease-associated genes. Proc. Natl. Acad. Sci. USA 118:e2112095118. doi: 10.1073/pnas.2112095118

PubMed Abstract | Crossref Full Text | Google Scholar

Mourtas, S., Lazar, A. N., Markoutsa, E., Duyckaerts, C., and Antimisiaris, S. G. (2014). Multifunctional nanoliposomes with curcumin-lipid derivative and brain targeting functionality with potential applications for Alzheimer disease. Eur. J. Med. Chem. 80, 175–183. doi: 10.1016/j.ejmech.2014.04.050

PubMed Abstract | Crossref Full Text | Google Scholar

Mukhopadhyay, N., Shukla, A., Makhal, P. N., and Kaki, V. R. (2023). Natural product-driven dual COX-LOX inhibitors: overview of recent studies on the development of novel anti-inflammatory agents. Heliyon 9:e14569. doi: 10.1016/j.heliyon.2023.e14569

PubMed Abstract | Crossref Full Text | Google Scholar

Muralikrishna Adibhatla, R., and Hatcher, J. F. (2006). Phospholipase A2, reactive oxygen species, and lipid peroxidation in cerebral ischemia. Free Radic. Biol. Med. 40, 376–387. doi: 10.1016/j.freeradbiomed.2005.08.044

PubMed Abstract | Crossref Full Text | Google Scholar

Nakagawa, S., and Aruga, J. (2020). Sphingosine 1-phosphate signaling is involved in impaired blood-brain barrier function in ischemia-reperfusion injury. Mol. Neurobiol. 57, 1594–1606. doi: 10.1007/s12035-019-01844-x

PubMed Abstract | Crossref Full Text | Google Scholar

Nitzsche, A., Poittevin, M., Benarab, A., Bonnin, P., Faraco, G., Uchida, H., et al. (2021). Endothelial S1P(1) signaling counteracts infarct expansion in ischemic stroke. Circ. Res. 128, 363–382. doi: 10.1161/CIRCRESAHA.120.316711

PubMed Abstract | Crossref Full Text | Google Scholar

Nong, J., Glassman, P. M., Shuvaev, V. V., Reyes-Esteves, S., Descamps, H. C., Kiseleva, R. Y., et al. (2024). Targeting lipid nanoparticles to the blood-brain barrier to ameliorate acute ischemic stroke. Mol. Ther. 32, 1344–1358. doi: 10.1016/j.ymthe.2024.03.004

PubMed Abstract | Crossref Full Text | Google Scholar

Nugent, A. A., Lin, K., van Lengerich, B., Lianoglou, S., Przybyla, L., Davis, S. S., et al. (2020). TREM2 regulates microglial cholesterol metabolism upon chronic phagocytic challenge. Neuron 105, 837–854.e9. doi: 10.1016/j.neuron.2019.12.007

PubMed Abstract | Crossref Full Text | Google Scholar

Olzmann, J. A., and Carvalho, P. (2019). Dynamics and functions of lipid droplets. Nat. Rev. Mol. Cell Biol. 20, 137–155. doi: 10.1038/s41580-018-0085-z

PubMed Abstract | Crossref Full Text | Google Scholar

Pan, Y., Xin, W., Wei, W., Tatenhorst, L., Graf, I., Popa-Wagner, A., et al. (2024). Knockdown of NEAT1 prevents post-stroke lipid droplet agglomeration in microglia by regulating autophagy. Cell. Mol. Life Sci. 81:30. doi: 10.1007/s00018-023-05045-7

PubMed Abstract | Crossref Full Text | Google Scholar

Pandian, S. R. K., Vijayakumar, K. K., Murugesan, S., and Kunjiappan, S. (2022). Liposomes: an emerging carrier for targeting Alzheimer's and Parkinson's diseases. Heliyon 8:e09575. doi: 10.1016/j.heliyon.2022.e09575

PubMed Abstract | Crossref Full Text | Google Scholar

Parodi-Rullán, R. M., Javadov, S., and Fossati, S. (2021). Dissecting the crosstalk between endothelial mitochondrial damage, vascular inflammation, and neurodegeneration in cerebral amyloid angiopathy and Alzheimer’s disease. Cells 10:2903. doi: 10.3390/cells10112903

PubMed Abstract | Crossref Full Text | Google Scholar

Pereira-Dutra, F. S., and Bozza, P. T. (2021). Lipid droplets diversity and functions in inflammation and immune response. Expert Rev. Proteomics 18, 809–825. doi: 10.1080/14789450.2021.1995356

PubMed Abstract | Crossref Full Text | Google Scholar

Phillips, G. R., Hancock, S. E., Brown, S. H. J., Jenner, A. M., Kreilaus, F., Newell, K. A., et al. (2020). Cholesteryl ester levels are elevated in the caudate and putamen of Huntington's disease patients. Sci. Rep. 10:20314. doi: 10.1038/s41598-020-76973-8

PubMed Abstract | Crossref Full Text | Google Scholar

Pifferi, F., Laurent, B., and Plourde, M. (2021). Lipid transport and metabolism at the blood-brain Interface: implications in health and disease. Front. Physiol. 12:645646. doi: 10.3389/fphys.2021.645646

PubMed Abstract | Crossref Full Text | Google Scholar

Podbielska, M., O'Keeffe, J., and Pokryszko-Dragan, A. (2021). New insights into multiple sclerosis mechanisms: lipids on the track to control inflammation and neurodegeneration. Int. J. Mol. Sci. 22:7319. doi: 10.3390/ijms22147319

PubMed Abstract | Crossref Full Text | Google Scholar

Pournajaf, S., Dargahi, L., Javan, M., and Pourgholami, M. H. (2022). Molecular pharmacology and novel potential therapeutic applications of Fingolimod. Front. Pharmacol. 13:807639. doi: 10.3389/fphar.2022.807639

PubMed Abstract | Crossref Full Text | Google Scholar

Pressly, J. D., Gurumani, M. Z., Varona Santos, J. T., Fornoni, A., Merscher, S., and al-Ali, H. (2022). Adaptive and maladaptive roles of lipid droplets in health and disease. Am. J. Physiol. Cell Physiol. 322, C468–c481. doi: 10.1152/ajpcell.00239.2021

PubMed Abstract | Crossref Full Text | Google Scholar

Qi, G., Mi, Y., Shi, X., Gu, H., Brinton, R. D., and Yin, F. (2021). ApoE4 impairs neuron-astrocyte coupling of fatty acid metabolism. Cell Rep. 34:108572. doi: 10.1016/j.celrep.2020.108572

PubMed Abstract | Crossref Full Text | Google Scholar

Rajmohan, R., and Reddy, P. H. (2017). Amyloid-beta and phosphorylated tau accumulations cause abnormalities at synapses of Alzheimer's disease neurons. J Alzheimer's Dis 57, 975–999. doi: 10.3233/JAD-160612

PubMed Abstract | Crossref Full Text | Google Scholar

Reid, M. M. (2023). Lipid mediated neuroprotective signaling in experimental ischemic stroke. Louisiana State University Health Sciences Center ProQuest Dissertations & Theses.

Google Scholar

Ridgway, N., and McLeod, R. (2008). Biochemistry of lipids, lipoproteins and membranes : Elsevier.

Google Scholar

Rinaldo, P., Matern, D., and Bennett, M. J. (2002). Fatty acid oxidation disorders. Annu. Rev. Physiol. 64, 477–502. doi: 10.1146/annurev.physiol.64.082201.154705

PubMed Abstract | Crossref Full Text | Google Scholar

Rodríguez-Gómez, J. A., Kavanagh, E., Engskog-Vlachos, P., Engskog, M. K. R., Herrera, A. J., Espinosa-Oliva, A. M., et al. (2020). Microglia: agents of the CNS pro-inflammatory response. Cells 9:1717. doi: 10.3390/cells9071717

PubMed Abstract | Crossref Full Text | Google Scholar

Rohrer, L., Ohnsorg, P. M., Lehner, M., Landolt, F., Rinninger, F., and von Eckardstein, A. (2009). High-density lipoprotein transport through aortic endothelial cells involves scavenger receptor BI and ATP-binding cassette transporter G1. Circ. Res. 104, 1142–1150. doi: 10.1161/CIRCRESAHA.108.190587

PubMed Abstract | Crossref Full Text | Google Scholar

Rudrapal, M., Eltayeb, W. A., Rakshit, G., el-Arabey, A. A., Khan, J., Aldosari, S. M., et al. (2023). Dual synergistic inhibition of COX and LOX by potential chemicals from Indian daily spices investigated through detailed computational studies. Sci. Rep. 13:8656. doi: 10.1038/s41598-023-35161-0

PubMed Abstract | Crossref Full Text | Google Scholar

Sahini, N., and Borlak, J. (2014). Recent insights into the molecular pathophysiology of lipid droplet formation in hepatocytes. Prog. Lipid Res. 54, 86–112. doi: 10.1016/j.plipres.2014.02.002

PubMed Abstract | Crossref Full Text | Google Scholar

Sandoval, K. E., and Witt, K. A. (2008). Blood-brain barrier tight junction permeability and ischemic stroke. Neurobiol. Dis. 32, 200–219. doi: 10.1016/j.nbd.2008.08.005

PubMed Abstract | Crossref Full Text | Google Scholar

Sela, M., Poley, M., Mora-Raimundo, P., Kagan, S., Avital, A., Kaduri, M., et al. (2023). Brain-targeted liposomes loaded with monoclonal antibodies reduce alpha-synuclein aggregation and improve behavioral symptoms in Parkinson's disease. Adv. Mater. 35:e2304654. doi: 10.1002/adma.202304654

PubMed Abstract | Crossref Full Text | Google Scholar

Shang, P., Zhang, Y., Ma, D., Hao, Y., Wang, X., Xin, M., et al. (2019). Inflammation resolution and specialized pro-resolving lipid mediators in CNS diseases. Expert Opin. Ther. Targets 23, 967–986. doi: 10.1080/14728222.2019.1691525

PubMed Abstract | Crossref Full Text | Google Scholar

Shimano, H., and Sato, R. (2017). SREBP-regulated lipid metabolism: convergent physiology - divergent pathophysiology. Nat. Rev. Endocrinol. 13, 710–730. doi: 10.1038/nrendo.2017.91

PubMed Abstract | Crossref Full Text | Google Scholar

Simpson, D. S. A., and Oliver, P. L. (2020). ROS generation in microglia: understanding oxidative stress and inflammation in neurodegenerative disease. Antioxidants (Basel) 9:743. doi: 10.3390/antiox9080743

PubMed Abstract | Crossref Full Text | Google Scholar

Sinclair, A. J. (2019). Docosahexaenoic acid and the brain-what is its role? Asia Pac. J. Clin. Nutr. 28, 675–688. doi: 10.6133/apjcn.201912_28(4).0002

PubMed Abstract | Crossref Full Text | Google Scholar

Singh, S. A., and Vellapandian, C. (2023). Structure of the blood brain barrier and its role in the transporters for the movement of substrates across the barriers. Curr. Drug Metab. 24, 250–269. doi: 10.2174/1389200224666230608110349

PubMed Abstract | Crossref Full Text | Google Scholar

Siqueira, L. D., Celes, A. P. M., Santos, H. D., and Ferreira, S. T. (2021). A specialized nutritional formulation prevents hippocampal glial activation and memory impairment induced by amyloid-β oligomers in mice. J Alzheimer's Dis 83, 1113–1124. doi: 10.3233/JAD-210139

PubMed Abstract | Crossref Full Text | Google Scholar

Smolič, T., Zorec, R., and Vardjan, N. (2021). Pathophysiology of lipid droplets in neuroglia. Antioxidants (Basel) 11:22. doi: 10.3390/antiox11010022

PubMed Abstract | Crossref Full Text | Google Scholar

Sochocka, M., Diniz, B. S., and Leszek, J. (2017). Inflammatory response in the CNS: friend or foe? Mol. Neurobiol. 54, 8071–8089. doi: 10.1007/s12035-016-0297-1

PubMed Abstract | Crossref Full Text | Google Scholar

Sonju, J. J., Dahal, A., Singh, S. S., and Jois, S. D. (2021). Peptide-functionalized liposomes as therapeutic and diagnostic tools for cancer treatment. J. Control. Release 329, 624–644. doi: 10.1016/j.jconrel.2020.09.055

PubMed Abstract | Crossref Full Text | Google Scholar

Sonnino, S., Aureli, M., Mauri, L., Ciampa, M. G., and Prinetti, A. (2015). Membrane lipid domains in the nervous system. Front. Biosci. 20, 280–302. doi: 10.2741/4309

PubMed Abstract | Crossref Full Text | Google Scholar

Spite, M., Clària, J., and Serhan, C. N. (2014). Resolvins, specialized proresolving lipid mediators, and their potential roles in metabolic diseases. Cell Metab. 19, 21–36. doi: 10.1016/j.cmet.2013.10.006

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, N., Keep, R. F., Hua, Y., and Xi, G. (2016). Critical role of the sphingolipid pathway in stroke: a review of current utility and potential therapeutic targets. Transl. Stroke Res. 7, 420–438. doi: 10.1007/s12975-016-0477-3

PubMed Abstract | Crossref Full Text | Google Scholar

Sun, Y., Wei, K., Liao, X., Wang, J., Gao, L., and Pang, B. (2025). Lipid metabolism in microglia: emerging mechanisms and therapeutic opportunities for neurodegenerative diseases (review). Int. J. Mol. Med. 56, 1–19. doi: 10.3892/ijmm.2025.5580

PubMed Abstract | Crossref Full Text | Google Scholar

Takabe, W., Urano, Y., Vo, D. K. H., Shibuya, K., Tanno, M., Kitagishi, H., et al. (2016). Esterification of 24S-OHC induces formation of atypical lipid droplet-like structures, leading to neuronal cell death. J. Lipid Res. 57, 2005–2014. doi: 10.1194/jlr.M068775

PubMed Abstract | Crossref Full Text | Google Scholar

Tan, Y. J., Jin, Y., Zhou, J., and Yang, Y. F. (2024). Lipid droplets in pathogen infection and host immunity. Acta Pharmacol. Sin. 45, 449–464. doi: 10.1038/s41401-023-01189-1

PubMed Abstract | Crossref Full Text | Google Scholar

Tansey, M. G., Wallings, R. L., Houser, M. C., Herrick, M. K., Keating, C. E., and Joers, V. (2022). Inflammation and immune dysfunction in Parkinson disease. Nat. Rev. Immunol. 22, 657–673. doi: 10.1038/s41577-022-00684-6

PubMed Abstract | Crossref Full Text | Google Scholar

Thiam, A. R., and Beller, M. (2017). The why, when and how of lipid droplet diversity. J. Cell Sci. 130, 315–324. doi: 10.1242/jcs.192021

PubMed Abstract | Crossref Full Text | Google Scholar

Torres, M., Parets, S., Fernández-Díaz, J., Beteta-Göbel, R., Rodríguez-Lorca, R., Román, R., et al. (2021). Lipids in pathophysiology and development of the membrane lipid therapy: new bioactive lipids. Membranes (Basel) 11:919. doi: 10.3390/membranes11120919

PubMed Abstract | Crossref Full Text | Google Scholar

Tracey, T. J., Steyn, F. J., Wolvetang, E. J., and Ngo, S. T. (2018). Neuronal lipid metabolism: multiple pathways driving functional outcomes in health and disease. Front. Mol. Neurosci. 11:10. doi: 10.3389/fnmol.2018.00010

PubMed Abstract | Crossref Full Text | Google Scholar

Urano, Y., Osaki, S., Chiba, R., and Noguchi, N. (2022). Integrated stress response is involved in the 24(S)-hydroxycholesterol-induced unconventional cell death mechanism. Cell Death Discov. 8:406. doi: 10.1038/s41420-022-01197-w

PubMed Abstract | Crossref Full Text | Google Scholar

Valencia-Olvera, A. C., Balu, D., Faulk, N., Amiridis, A., Wang, Y., Pham, C., et al. (2023). Inhibition of ACAT as a therapeutic target for Alzheimer's disease is independent of ApoE4 lipidation. Neurotherapeutics 20, 1120–1137. doi: 10.1007/s13311-023-01375-3

PubMed Abstract | Crossref Full Text | Google Scholar

van Meer, G., Voelker, D. R., and Feigenson, G. W. (2008). Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112–124. doi: 10.1038/nrm2330

PubMed Abstract | Crossref Full Text | Google Scholar

Vieira, D. B., and Gamarra, L. F. (2016). Getting into the brain: liposome-based strategies for effective drug delivery across the blood–brain barrier. Int. J. Nanomedicine 11, 5381–5414. doi: 10.2147/IJN.S117210

PubMed Abstract | Crossref Full Text | Google Scholar

Vigouroux, C., Caron-Debarle, M., le Dour, C., Magré, J., and Capeau, J. (2011). Molecular mechanisms of human lipodystrophies: from adipocyte lipid droplet to oxidative stress and lipotoxicity. Int. J. Biochem. Cell Biol. 43, 862–876. doi: 10.1016/j.biocel.2011.03.002

PubMed Abstract | Crossref Full Text | Google Scholar

Walther, T. C., and Farese, R. V. Jr. (2012). Lipid droplets and cellular lipid metabolism. Annu. Rev. Biochem. 81, 687–714. doi: 10.1146/annurev-biochem-061009-102430

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, C. W. (2016). Lipid droplets, lipophagy, and beyond. Biochim. Biophys. Acta 1861, 793–805. doi: 10.1016/j.bbalip.2015.12.010

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, L., Kong, Q., Leng, X., Leung, H., and Li, Y. (2025). The sphingosine-1-phosphate signaling pathway (sphingosine-1-phosphate and its receptor, sphingosine kinase) and epilepsy. Epilepsia Open 10, 55–73. doi: 10.1002/epi4.13112

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, N. Q., Sun, P. X., Shen, Q. Q., and Deng, M. Y. (2025). Cholesterol metabolism in CNS diseases: the potential of SREBP2 and LXR as therapeutic targets. Mol. Neurobiol. 62, 6283–6307. doi: 10.1007/s12035-024-04672-w

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, B., and Tontonoz, P. (2018). Liver X receptors in lipid signalling and membrane homeostasis. Nat. Rev. Endocrinol. 14, 452–463. doi: 10.1038/s41574-018-0037-x

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, G., Wang, Y., Liu, N., and Liu, M. (2020). The role of exosome lipids in central nervous system diseases. Rev. Neurosci. 31, 743–756. doi: 10.1515/revneuro-2020-0013

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, N., and Westerterp, M. (2020). ABC transporters, cholesterol efflux, and implications for cardiovascular diseases. Adv. Exp. Med. Biol. 1276, 67–83. doi: 10.1007/978-981-15-6082-8_6

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, B., Wu, L., Chen, J., Dong, L., Chen, C., Wen, Z., et al. (2021). Metabolism pathways of arachidonic acids: mechanisms and potential therapeutic targets. Signal Transduct. Target. Ther. 6:94. doi: 10.1038/s41392-020-00443-w

PubMed Abstract | Crossref Full Text | Google Scholar

Wang, Z., Zheng, Y., Wang, F., Zhong, J., Zhao, T., Xie, Q., et al. (2020). Mfsd2a and Spns2 are essential for sphingosine-1-phosphate transport in the formation and maintenance of the blood-brain barrier. Sci. Adv. 6:eaay8627. doi: 10.1126/sciadv.aay8627

PubMed Abstract | Crossref Full Text | Google Scholar

Wei, W., Lattau, S. S. J., Xin, W., Pan, Y., Tatenhorst, L., Zhang, L., et al. (2024). Dynamic brain lipid profiles modulate microglial lipid droplet accumulation and inflammation under ischemic conditions in mice. Adv. Sci. (Weinh) 11:e2306863. doi: 10.1002/advs.202306863

PubMed Abstract | Crossref Full Text | Google Scholar

Wei, W., Zhang, L., Xin, W., Pan, Y., Tatenhorst, L., Hao, Z., et al. (2024). TREM2 regulates microglial lipid droplet formation and represses post-ischemic brain injury. Biomed. Pharmacother. 170:115962. doi: 10.1016/j.biopha.2023.115962

PubMed Abstract | Crossref Full Text | Google Scholar

Wood, C. A. P., Zhang, J., Aydin, D., Xu, Y., Andreone, B. J., Langen, U. H., et al. (2021). Structure and mechanism of blood-brain-barrier lipid transporter MFSD2A. Nature 596, 444–448. doi: 10.1038/s41586-021-03782-y

PubMed Abstract | Crossref Full Text | Google Scholar

Wouters, E., de Wit, N. M., Vanmol, J., van der Pol, S. M. A., van het Hof, B., Sommer, D., et al. (2019). Liver X receptor alpha is important in maintaining blood-brain barrier function. Front. Immunol. 10:1811. doi: 10.3389/fimmu.2019.01811

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, D., Chen, Q., Chen, X., Han, F., Chen, Z., and Wang, Y. (2023). The blood-brain barrier: structure, regulation, and drug delivery. Signal Transduct. Target. Ther. 8:217. doi: 10.1038/s41392-023-01481-w

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, W., Luo, Z., Shen, D., Lan, T., Xiao, Z., Liu, M., et al. (2024). IL-10 protects against OPC ferroptosis by regulating lipid reactive oxygen species levels post stroke. Redox Biol. 69:102982. doi: 10.1016/j.redox.2023.102982

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, Y. R., Shi, X. Y., Ma, C. Y., Zhang, Y., Xu, R. X., and Li, J. J. (2019). Liraglutide improves lipid metabolism by enhancing cholesterol efflux associated with ABCA1 and ERK1/2 pathway. Cardiovasc. Diabetol. 18:146. doi: 10.1186/s12933-019-0954-6

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, Z., Wu, S., Liang, T., and Wang, L. (2021). Lipoprotein-associated phospholipase A2 is a risk factor for patients with Parkinson's disease. Front. Neurosci. 15:633022. doi: 10.3389/fnins.2021.633022

PubMed Abstract | Crossref Full Text | Google Scholar

Wu, L., Xiong, X., Wu, X., Ye, Y., Jian, Z., Zhi, Z., et al. (2020). Targeting oxidative stress and inflammation to prevent ischemia-reperfusion injury. Front. Mol. Neurosci. 13:28. doi: 10.3389/fnmol.2020.00028

PubMed Abstract | Crossref Full Text | Google Scholar

Xi, Y., Zhang, Y., Zhu, S., Luo, Y., Xu, P., and Huang, Z. (2020). PPAR-mediated toxicology and applied pharmacology. Cells 9:2. doi: 10.3390/cells9020352

PubMed Abstract | Crossref Full Text | Google Scholar

Xie, Y., Yan, L., Zeng, H., Chen, W., Lu, J. H., Wan, J. B., et al. (2020). Fish oil protects the blood-brain barrier integrity in a mouse model of Alzheimer's disease. Chin. Med. 15:29. doi: 10.1186/s13020-020-00314-0

PubMed Abstract | Crossref Full Text | Google Scholar

Xin, W., Pan, Y., Wei, W., Gerner, S. T., Huber, S., Juenemann, M., et al. (2023). TGF-β1 decreases microglia-mediated neuroinflammation and lipid droplet accumulation in an in vitro stroke model. Int. J. Mol. Sci. 24:17329. doi: 10.3390/ijms242417329

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, Y., du, X., Turner, N., Brown, A. J., and Yang, H. (2019). Enhanced acyl-CoA:cholesterol acyltransferase activity increases cholesterol levels on the lipid droplet surface and impairs adipocyte function. J. Biol. Chem. 294, 19306–19321. doi: 10.1074/jbc.RA119.011160

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, X., Jin, W., Chang, R., and Ding, X. (2024). Research progress of SREBP and its role in the pathogenesis of autoimmune rheumatic diseases. Front. Immunol. 15:1398921. doi: 10.3389/fimmu.2024.1398921

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, P., Li, D., Tang, X., Bao, X., Huang, J., Tang, Y., et al. (2013). LXR agonists: new potential therapeutic drug for neurodegenerative diseases. Mol. Neurobiol. 48, 715–728. doi: 10.1007/s12035-013-8461-3

PubMed Abstract | Crossref Full Text | Google Scholar

Xu, H., Wang, E., Chen, F., Xiao, J., and Wang, M. (2021). Neuroprotective phytochemicals in experimental ischemic stroke: mechanisms and potential clinical applications. Oxidative Med. Cell. Longev. 2021:6687386. doi: 10.1155/2021/6687386

PubMed Abstract | Crossref Full Text | Google Scholar

Yamazaki, Y., Zhao, N., Caulfield, T. R., Liu, C. C., and Bu, G. (2019). Apolipoprotein E and Alzheimer disease: pathobiology and targeting strategies. Nat. Rev. Neurol. 15, 501–518. doi: 10.1038/s41582-019-0228-7

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, T., Ferrill, L., Gallant, L., McGillicuddy, S., Fernandes, T., Schields, N., et al. (2018). Verapamil and riluzole cocktail liposomes overcome pharmacoresistance by inhibiting P-glycoprotein in brain endothelial and astrocyte cells: A potent approach to treat amyotrophic lateral sclerosis. Eur. J. Pharm. Sci. 120, 30–39. doi: 10.1016/j.ejps.2018.04.026

PubMed Abstract | Crossref Full Text | Google Scholar

Yang, C., Hawkins, K. E., Doré, S., and Candelario-Jalil, E. (2019). Neuroinflammatory mechanisms of blood-brain barrier damage in ischemic stroke. Am. J. Physiol. Cell Physiol. 316, C135–c153. doi: 10.1152/ajpcell.00136.2018

PubMed Abstract | Crossref Full Text | Google Scholar

Yin, F. (2023). Lipid metabolism and Alzheimer's disease: clinical evidence, mechanistic link and therapeutic promise. FEBS J. 290, 1420–1453. doi: 10.1111/febs.16344

PubMed Abstract | Crossref Full Text | Google Scholar

Zeng, X., Luo, Z., Wu, J., Zhou, J., Shan, Y., Zhu, Y., et al. (2020). Cerebral ischemia-reperfusion injury: lysophosphatidic acid mediates inflammation by decreasing the expression of liver X receptor. J. Mol. Neurosci. 70, 1376–1384. doi: 10.1007/s12031-020-01554-3

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, Z. (2014). The dual role of hypoxia-inducible factor-1 in ischemic stroke: neuroprotection and blood-brain barrier disruption : University of Kansas. Available online at: https://core.ac.uk/download/pdf/213407101.pdf

Google Scholar

Zhang, W., Wang, T., Pei, Z., Miller, D. S., Wu, X., Block, M. L., et al. (2005). Aggregated alpha-synuclein activates microglia: a process leading to disease progression in Parkinson's disease. FASEB J. 19, 533–542. doi: 10.1096/fj.04-2751com

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, N., Yan, F., Liang, X., Wu, M., Shen, Y., Chen, M., et al. (2018). Localized delivery of curcumin into brain with polysorbate 80-modified cerasomes by ultrasound-targeted microbubble destruction for improved Parkinson's disease therapy. Theranostics 8, 2264–2277. doi: 10.7150/thno.23734

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, D. D., Yu, H. L., Ma, W. W., Liu, Q. R., Han, J., Wang, H., et al. (2015). 27-hydroxycholesterol contributes to disruptive effects on learning and memory by modulating cholesterol metabolism in the rat brain. Neuroscience 300, 163–173. doi: 10.1016/j.neuroscience.2015.05.022

PubMed Abstract | Crossref Full Text | Google Scholar

Zhang, L., Zhou, Y., Yang, Z., Jiang, L., Yan, X., Zhu, W., et al. (2025). Lipid droplets in central nervous system and functional profiles of brain cells containing lipid droplets in various diseases. J. Neuroinflammation 22:7. doi: 10.1186/s12974-025-03334-5

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, Y., Gan, L., Ren, L., Lin, Y., Ma, C., and Lin, X. (2022). Factors influencing the blood-brain barrier permeability. Brain Res. 1788:147937. doi: 10.1016/j.brainres.2022.147937

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, L., Lei, W., Deng, C., Wu, Z., Sun, M., Jin, Z., et al. (2021). The roles of liver X receptor α in inflammation and inflammation-associated diseases. J. Cell. Physiol. 236, 4807–4828. doi: 10.1002/jcp.30204

PubMed Abstract | Crossref Full Text | Google Scholar

Zhao, X., Zhang, S., Sanders, A. R., and Duan, J. (2023). Brain lipids and lipid droplet dysregulation in Alzheimer's disease and neuropsychiatric disorders. Complex Psychiatry 9, 154–171. doi: 10.1159/000535131

PubMed Abstract | Crossref Full Text | Google Scholar

Zhong, J., Peng, Y., Zhang, L., Xiao, B., and Zhang, M. (2025). Astrocyte lipid droplet dynamics orchestrate neurological disorders and therapeutic horizons. Small Sci. :2500152. doi: 10.1002/smsc.202500152

PubMed Abstract | Crossref Full Text | Google Scholar

Glossary

Aβ - Amyloid-beta

ABCA1 - ATP-binding cassette transporter A1

ABCG1 - ATP-binding cassette transporter G1

ACAT1 - Acyl-CoA cholesterol acyltransferase 1

ACSL4 - Acyl-CoA synthetase long-chain family member 4

AD - Alzheimer’s Disease

ALS - Amyotrophic Lateral Sclerosis

ApoE - Apolipoprotein E

APP - Amyloid Precursor Protein

AS - Astrocytes

BBB - Blood–Brain Barrier

CEs - Cholesteryl Esters

CNS - Central Nervous System

COX - Cyclooxygenase

COX-2 - Cyclooxygenase-2

DHA - Docosahexaenoic Acid

EC - Endothelial Cell

ER - Endoplasmic Reticulum

FA - Fatty Acid

HDL-C - High-Density Lipoprotein Cholesterol

HD - Huntington’s Disease

ICH - Intracerebral Hemorrhagic Stroke

IFN-γ - Interferon-gamma

IL-1β - Interleukin-1 beta

IL-6 - Interleukin-6

IR - Insulin Receptor

IS - Ischemic Stroke

LDs - Lipid Droplets

LDLR - Low-Density Lipoprotein Receptor

LOX - Lipoxygenase

LPA - Lysophosphatidic Acid

LXRs - Liver X Receptors

MDA - Malondialdehyde

MD - Medical Doctor

MFSD2A - Major Facilitator Superfamily Domain-containing 2A

MS - Multiple Sclerosis

NF-κB - Nuclear Factor Kappa-Light-Chain-Enhancer of Activated B Cells

PD - Parkinson’s Disease

PEGylation - Polyethylene Glycol Modification

PLA2 - Phospholipase A2

PLINs - Perilipins

PPAR-γ - Peroxisome Proliferator-Activated Receptor Gamma

PUFA - Polyunsaturated Fatty Acid

ROS - Reactive Oxygen Species

S1P - Sphingosine-1-phosphate

S1PR - Sphingosine-1-phosphate Receptor

SREBPs - Sterol Regulatory Element-Binding Proteins

TC - Triglycerides

TfR - Transferrin Receptor

TG - Triglycerides

TNF-α - Tumor Necrosis Factor-alpha

TREM2 - Triggering Receptor Expressed on Myeloid cells 2

4-HNE - 4-Hydroxynonenal

10-NO2-OA - 10-Nitro-oleic Acid

Keywords: lipid droplets, liposomes, blood–brain barrier, neurological disorders, neuroprotection

Citation: Fu L, Luo T, Hao Z, Pan Y, Xin W, Zhang L, Lai Z, Zhang H, Liu H and Wei W (2025) Exploring novel roles of lipid droplets and lipid metabolism in regulating inflammation and blood–brain barrier function in neurological diseases. Front. Neurosci. 19:1603292. doi: 10.3389/fnins.2025.1603292

Received: 31 March 2025; Accepted: 01 August 2025;
Published: 13 August 2025.

Edited by:

Hansen Chen, Stanford University, United States

Reviewed by:

Abel Eraso Pichot, INSERM U1215 Neurocentre Magendie, France
Jia Nong, University of Pennsylvania, United States

Copyright © 2025 Fu, Luo, Hao, Pan, Xin, Zhang, Lai, Zhang, Liu and Wei. 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: Wei Wei, eHlxMDEzODYyQGdtYWlsLmNvbQ==; Hua Liu, aHhsaXVtZWRpZG9jdG9yQDE2My5jb20=

These authors have contributed equally to this work and share first authorship

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.