REVIEW article

Front. Cardiovasc. Med., 24 April 2025

Sec. Atherosclerosis and Vascular Medicine

Volume 12 - 2025 | https://doi.org/10.3389/fcvm.2025.1568528

This article is part of the Research TopicImmunity, Atherosclerosis and Cardiovascular Disease: An Interdisciplinary Approach to Cardiometabolic HealthView all 18 articles

Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies


Edward B. ThorpEdward B. Thorp1Aparnaa AnanthakrishnanAparnaa Ananthakrishnan1Connor W. Lantz

Connor W. Lantz2*
  • 1Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
  • 2Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States

Chronic allograft vasculopathy (CAV) is a major cause of late graft failure in heart transplant recipients, characterized by progressive intimal thickening and diffuse narrowing of the coronary arteries. Unlike atherosclerosis, CAV exhibits a distinct cellular composition and lesion distribution, yet its pathogenesis remains incompletely understood. A major challenge in CAV research has been the limited application of advanced “-omics” technologies, which have revolutionized the study of other vascular diseases. Recent advancements in single-cell and spatial transcriptomics, proteomics, and metabolomics have begun to uncover the complex immune-endothelial-stromal interactions driving CAV progression. Notably, single-cell RNA sequencing has identified previously unrecognized immune cell populations and signaling pathways implicated in endothelial injury and vascular remodeling after heart transplantation. Despite these breakthroughs, studies applying these technologies to CAV remain sparse, limiting the translation of these insights into clinical practice. This review aims to bridge this gap by summarizing recent findings from single-cell and multi-omic approaches, highlighting key discoveries, and discussing their implications for understanding CAV pathogenesis.

Introduction

Atherosclerosis, characterized by the progressive buildup of plaques within arterial walls, is a leading cause of ischemic heart disease and heart failure. In severe cases where cardiac function is irreversibly compromised, heart transplantation remains the only viable option to fully restore cardiac function and improve patient survival. In the US alone, over 3,000 heart transplantations are performed annually (1). However, approximately 30% of cardiac allografts fail within the ten years of transplantation, with a significant proportion attributed to cardiac allograft vasculopathy (CAV) after 3 years following transplantation (24) CAV is a progressive, intimal hyperplastic lesion affecting both arteries and veins, leading to maladaptive vessel narrowing that clinically presents in a wide range of pathologies including myocardial infarction and sudden cardiac death.

CAV shares several similarities with atherosclerosis but also exhibits notable distinctions. Unlike atherosclerosis, which often presents as focal, eccentric lesions, CAV is a diffuse, concentric process that involves the entire coronary vascular tree (5). While atherosclerosis is a long-life remodeling process starting in childhood and manifesting in advanced adult age, CAV exhibits a rapid onset within months after transplantation. The rapid onset of CAV is triggered by immune-mediated processes initiated by alloimmune responses, while atherosclerosis is primarily driven by prolonged lipid accumulation and metabolic stress that leads to endothelial dysfunction. Even though the progression of disease pathology is distinct, both atherosclerosis and CAV are mediated by both the innate and adaptive immune system, particularly macrophages, DCs, T cells, and B cells (6).

Pathologically, CAV is predominantly immune-mediated and characterized by intimal hyperplasia composed of accumulation of vascular smooth muscle cells (VSMCs), an intact internal elastic lamina, a preserved tunica media, and mononuclear cell infiltrations (5, 7). Compared to atherosclerotic regions, CAV lesions frequently feature intraplaque hemorrhages which may accelerate lesion progression and contribute to lumen stenosis (8). These pathological differences also influence treatment strategies. While atherosclerosis is managed primarily through a combination of lipid-lowering therapies (e.g., statins, PCSK9 inhibitors) (9) and anti-inflammatory agents, CAV requires immunosuppressive strategies such as mTOR inhibitors (sirolimus and everolimus) (10, 11), calcineurin inhibitors (tacrolimus) (12), and proliferation inhibitors (mycophenolate) (13). Additionally, the diffuse nature of CAV makes revascularization techniques like stenting or bypass surgery less effective compared to their use in atherosclerosis (14). Given the “unnatural” process of heart transplantation, understanding the molecular and cellular mechanisms driving the pathogenesis of CAV may provide valuable insights into the interplay between the immune system and the vasculature that may also underlie other vascular diseases.

The rapid advances in “-omics” methodologies have revolutionized our ability to investigate biology across space and at numerous molecular levels, encompassing DNA, RNA, proteins, and metabolites (15, 16). These approaches are increasingly being applied to the field of allograft pathology and CAV, offering new avenues to investigate the driving factors for disease progression (1719) High-resolution multi-omics approaches provide unparalleled insights into the intricate tissue microenvironments that shape transplant outcomes, enabling a deeper understanding of the dynamic interactions between host and graft and the mechanisms driving alloimmune responses. These breakthroughs have the potential to redefine strategies for diagnosing, monitoring, and treating CAV, facilitating the development of precision medicine approaches in transplantation care (16). This review seeks to synthesize recent findings from a diverse array of bioinformatic approaches that have significantly advanced our understanding of immune-mediated processes underlying CAV (Table 1). Additionally, it highlights unresolved questions raised by these studies, identifying knowledge gaps and suggesting priority areas for future research to further elucidate CAV pathophysiology and improve prolonged clinical outcomes for patients with heart transplantation.

Table 1
www.frontiersin.org

Table 1. Key computational biology studies to interrogate immune cell function during cardiac allograft vasculopathy and chronic rejection.

Immunity during cardiac allograft vasculopathy

One of the primary challenges in treating CAV is its delayed clinical presentation. Due to the lack of sensory nerves in the transplanted heart, CAV develops silently, without the typical warning signs of angina pectoris (20). As a result, it often manifests at an advanced stage presenting with symptoms of graft dysfunction, arrhythmias, or even sudden cardiac death. Although the clinical presentation of CAV typically emerge years after transplantation, early inflammatory events within the first year are critical to its initiation and pathogenesis (21). The upregulation of Human Leukocyte Antigen (HLA) class II molecules has been identified as a significant trigger of CAV in large cohorts of European and American heart transplant recipients (22). Additionally, acute cellular rejection, primarily mediated by T cells, has been recognized as an independent risk factor for CAV progression (22). While the precise mechanisms linking these inflammatory triggers to disease progression remain largely correlative, recent advances in large-scale biological approaches have greatly enhanced our understanding of CAV in preclinical models. In the following sections, we review the contributions of both the innate and adaptive immune systems to CAV progression. Future studies should aim to validate these findings in clinical cohorts and develop biomarkers or other diagnostic methods to improve early detection and intervention of CAV.

Monocytes and macrophages

Macrophages represent a heterogenous population within the innate immune system, performing diverse functions during homeostasis (23) and pathological conditions such as ischemia-reperfusion injury (24, 25), acute rejection (26, 27), and chronic rejection (28, 29) in cardiac transplantation. Macrophages play critical roles through the lifespan of an allograft, significantly influencing the onset and progression of CAV. Beyond their interactions with the adaptive immune system, macrophages actively shape the vasculature's microenvironment by secreting signals that activate fibroblasts and VSMCs driving fibrous intimal thickening of CAV (28, 30). Experimental depletion of myeloid cells after heterotopic heart transplantation suppresses the development of vasculopathy, underscoring their necessity in CAV pathogenesis (29, 31). Recent advances in single-cell and spatial transcriptomics technologies are beginning to unravel the intricate contributions of macrophages to CAV pathophysiology, shedding light on their origins, functional specialization, and localization within the heart.

Historically, macrophages were thought to originate exclusively from circulating monocytes, as described by the mononuclear phagocyte system. However, advanced genetic mouse models tracing the fates of myeloid precursor cells have demonstrated that tissue macrophages arise from distinct developmental origins, including macrophages residing within the heart (3235) During fetal development, progenitors from the yolk sac and fetal liver seed tissues, giving rise to local resident macrophage populations that perform specialized roles within tissue-specific niches (36, 37). These yolk sac-derived resident macrophages, despite adapting to diverse tissue environments, exhibit high transcriptional conservation and are characterized by the expression of TIM-4, LYVE-1, or FOLR2 (38). Within the heart, these embryonically derived macrophages are able to self-renew, persisting well into adulthood (39). Throughout aging, however, resident cardiac macrophages are continuously replaced by monocyte-derived macrophages, which can be identified by the expression of C-C chemokine receptor 2 (CCR2) (34, 40).

Insights from lineage-tracing studies and transplantation models have informed one another about macrophage origins and their immunogenic roles. Kory Lavine and colleagues reported that in sex-mismatched heart transplant recipients (female donors to male recipients), CCR2- macrophages lacked expression of the Y chromosome, demonstrating their donor tissue-residence origin (41). CCR2 + macrophages, on the other hand, represented a heterogeneous population containing cells both with and without Y chromosome expression, highlighting the significant contribution of peripheral monocyte recruitment to this macrophage subset in the human heart (41). Newer analyses of single cell transcriptomics now enable the differentiation of donor and recipient cells based on naturally occurring small-nucleotide variances (SNVs) (42, 43). In pediatric heart transplant recipients with CAV, almost 90% of myeloid cells within the allograft were of recipient origin by five days post-transplantation, with all cardiac immune cells transitioning to recipient origin by 15 months post-transplantation (19). This pattern mirrored observations in murine models of heterotopic heart transplantation (44). Shortly after transplantation, circulating monocytes from the recipient rapidly infiltrate the heart and polarize into proinflammatory macrophages which have been associated with CAV severity (Figure 1) (19, 45). The mechanisms by which this initial wave of inflammation, driven by this distinct population of hyperinflammatory macrophages, primes the vasculature for the chronic development of CAV remain incompletely understood. Additional epigenomic and transcriptomic studies are needed to elucidate the lasting effects of these macrophages on the vasculature and their role in the progression of CAV.

Figure 1
www.frontiersin.org

Figure 1. Immune cell interactions driving cardiac allograft vasculopathy. The schematic illustrates the key cellular and molecular mechanisms involved in the progression of CAV. After ischemic-reperfusion injury during heart transplantation, endothelial cell activation and the development of de novo DSAs promotes the recruitment of monocytes, NK cells and T cells into the inflamed vessel. Monocyte-derived macrophages are stimulated through Notch signaling by the endothelium, polarizing them into an inflammatory phenotype. Upon Lyve-1 stimulation, macrophages secrete MMP-9, remodeling the basement membrane and promoting further immune cell extravasation. Additionally, macrophages produce growth factors such as PDGF which drive VSMC proliferation—a hallmark of CAV. DSAs also promote complement activation, leading to the formation of the membrane attack complex and causing endothelial cell damage. Released DAMPs generate new autoantibodies, further amplifying antibody-mediated rejection and allograft injury. Regulatory T cells may play a protective role within the vessel by suppressing NK cell and CD8+ T cell activation. These cellular interactions collectively contribute to the remodeling of the vascular intima, leading to luminal narrowing and eventual graft dysfunction.

Importantly, donor-derived CCR2 + macrophages have been implicated in graft survival. Loss of these macrophages was associated with prolonged allograft survival, likely due to reduced signaling through the MyD88 pathway (44). This pathway plays a pivotal role in enabling antigen-presenting cells to present antigens to T cells, and its inhibition significantly extends graft survival. In contrast, selective depletion of donor CCR2- macrophages prior to transplantation has been shown to acutely reduce allograft survival (44). Therefore, CCR2- macrophages likely play a protective role early post-transplantation and their ensuing loss may result from process linked to allograft rejection. In human transplant recipients, immune suppression therapy appears to impair the proliferative capacity of donor CCR2- macrophages, thus leading to their eventual replacement by recipient monocyte-derived macrophages.

While recipient macrophages infiltrating the allograft exhibit transcriptomic similarities to donor-resident macrophages, they do not fully replicate their functional properties. Specifically, recipient-derived macrophages demonstrate reduced phagocytic activity yet an increased expression of inflammation-resolving genes (19). Natural defects in macrophage phagocytosis have been correlated with poor post-transplant outcomes, although a direct relation has yet to be fully established (46). Efficient clearance of dead cells by macrophages prevents the accumulation of immunogenic self-antigens and promotes tissue-reparative and tolerogenic signaling (47). Leveraging this tolerogenic function, interventions such as the injection of apoptotic donor cells have been shown to induce immune tolerance and prolong heart allograft survival in murine models (4850) Notably, these recipient macrophages also exhibit enhanced fibrotic signaling, including elevated expression of PDGFB, a gene implicated in fibrotic remodeling of the heart (19, 51, 52). Bone marrow-derived myeloid cells upregulate PDGFB to also promote the proliferation of vascular smooth muscle cells, a process that is dependent on the efferocytosis receptor AXL (28). Further studies are needed to delineate the unique roles of donor- and recipient-derived CCR2- macrophages in apoptotic cell clearance, inflammation resolution, and fibrotic signaling during chronic rejection and CAV.

While these single cell approaches utilizing endomyocardial biopsies have tremendously increased our understanding of macrophage origin and function in the allografted heart, they may not fully capture the immune and vascular dynamics contributing to neointima formation. During homeostasis, two different interstitial macrophage populations exist across tissues including the heart, fat, dermis, and lung: one that preferentially surrounds the nerves and one that localizes with blood vessels (53). After cardiac transplantation, macrophages are present within the neointima and adventitia of CAV lesions and are associated with antibody-mediated rejection (5456) Leveraging the utility of spatial transcriptomics and proteomics, Elaine Reed and colleagues revealed that arterial lesions with low neointimal thickness exhibit higher inflammatory and cell death signatures, whereas lesions with high neointimal thickness display remodeling and fibrotic profiles aligning with neointima expansion (17). These findings support the observations that inflammation precedes the expansion of the neointima through SMC accumulation followed by subsequent vascular fibrosis (57). Furthermore, macrophages around blood vessels express high levels of the hyaluronan receptor LYVE-1, which interacts with hyaluronic acid on the surface of VSMCs to promote collagen degradation through pericellular MMP-9 activity (58). Interestingly, in CAV-affected lesions, macrophages secrete MMP-9 in response to endothelial cells activated by anti-HLA class I donor-specific antibodies (59). MMP-9-producing monocytes degradation of the collagen basement membrane allows for invasion of T cells in large vessels (60). These findings highlight the complex interplay between macrophages, vascular smooth muscle cells, and immune responses in driving the progression of CAV, underscoring the need for further research to fully elucidate the mechanisms contributing to neointima formation and vascular fibrosis after transplantation.

Natural killer cells

Natural killer (NK) cells, lymphocytes of the innate immune system, have been implicated in the development and progression of transplant-associated arteriosclerosis (61). NK cells control pathogenic viral infections and eliminate malignant cells, but their role extends beyond cytotoxicity as they also secrete cytokines and chemokines, most notably IFN-γ (6264) Historically, NK cells were defined by their morphology and function of that of a large granular lymphocyte that can kill its target cells naturally, meaning NK cells were not restricted by MHC expression on the target cell (65, 66). These insights stemmed from the peculiar observations by Gustavo Cudkowicz and Michael Bennet that F1 hybrid mice reject the transplantation of parental bone marrow cells (67, 68) leading to the “missing self-hypothesis” which states that NK cells would eliminate target cells that lacked self MHC-I molecule expression (69). Further studies revealed that NK cells expressed killer immunoglobulin-like receptors (KIRs) that engage MHC-I molecules to generate an inhibitory signal to prevent NK-mediated cell killing (70). In the context of HLA class I mismatches in transplantation, NK cells are likely to perceive graft cells as “missing self”, triggering NK cell activation and subsequent endothelial cell death (71). Further supporting this hypothesis, it was demonstrated that in semi-allogenic cardiac transplants between parental donors and F1 hybrid recipients, that NK cells contribute to the development of CAV by recruiting T cells through secretion of IFN-γ (72). Remarkably, even in the absence of T and B cells, NK cells are sufficient to promote vasculopathy in mice (73). This phenomenon may stem from their role in viral immunity, as lymphocytic choriomeningitis virus can induce CAV through NK cell activity without involvement of adaptive immune cells (74). Moreover, NK cell inhibition in T-cell-depleted mice resulted in prolonged acceptance of cardiac allografts (75). Together, these findings reveal that NK cells are key drivers of chronic vascular injury, significantly contributing to the development and progression of allograft vasculopathy.

NK cells efficiently lyse target cells without prior stimulation, relying on a finely tuned balance of activating and inhibitory signals mediated by their receptors (66). Inhibition of one such activating receptor, NKG2D, has been shown to prolong the survival of allografted hearts (76, 77). However, contradictory findings suggest that NKG2D deletion can also accelerate heart allograft rejection (78). These contrasting outcomes highlight the duality of NK cell functions: while they are associated with chronic graft injury, they can also promote tolerance by targeting donor antigen-presenting cells (APCs) and preventing their migration to recipient secondary lymphoid tissue (79). However, this tolerogenic role of NK cells is likely diminished as donor APCs are rapidly replaced by recipient-derived cells (19). Beyond their direct cytotoxic effects, NK cells are integral to the immune response to donor-specific antibodies (DSAs). NK cell-mediated IFN-γ production and contact-dependent cytotoxic activity are rate-limiting effector pathways during antibody-induced chronic allograft vasculopathy (80). Through the Fc receptor CD16a, NK cells recognize DSAs, triggering antibody-dependent cytotoxicity (81). Notably, elevated CD16a expression is linked to a higher risk of CAV in human patients, driving both IFN-γ production and the release of cytotoxic molecules (82, 83).

Recent advances in single-cell sequencing have revealed a distinct subset of NK cells that are linked to dysfunctional allografts and persistent CAV (19). These NK cells exhibit elevated levels of IFN-γ, CRTAM, and Fas-ligand, linking them to chronic inflammatory and cytotoxic processes within the graft. Further analysis of cell communication from single-cell data suggests that proinflammatory macrophages regulate NK cell activation via IL-18 and CSF2 signaling (19, 84). This interaction underscores the adaptability of NK cells and their significant role in long-term graft dysfunction. In human decedent studies with pig heart xenografts, integrative multi-omics analysis showed a marked increase in NK cell activity as early as one day after transplantation (85). Secreted IFN-γ by NK cells promotes the production of CXCL9 and CXCL10 by cardiac fibroblasts, essential chemokines for NK cell recruitment, thereby creating a persistent feedforward loop within the allograft (86, 87). Furthermore, IFN-γ has long been recognized as a central effector in allograft arteriosclerosis due to its pleiotropic roles in cell proliferation, death, inflammation, and fibrosis (88). Together, these findings highlight a complex and self-sustaining network of NK cell activation and chemokine signaling within dysfunctional allografts, emphasizing the need for targeted therapeutic strategies to disrupt this feedforward loop and mitigate chronic graft inflammation and dysfunction.

B cells and antibodies

The innate immune system alone is not sufficient for causing chronic rejection of cardiac allografts (72, 89). The interplay between adaptive immune cells and antibody-mediated mechanisms is now recognized as a significant contributor to the progression of allograft vasculopathy within patients, particularly with the strong association of DSAs to chronic allograft rejection and CAV (90). Early studies demonstrated that T cells are necessary for CAV development as transfer of T cells in immunodeficient mice successfully recapitulated the characteristic vascular damage of CAV (91). In contrast, the role of B cells in CAV has been more uncertain, with findings varying based on mouse models and the immunosuppressive regimens used to induce CAV (9295) Despite these challenges in animal models, clinical evidence shows a clear association between the presence of B and plasma cells around coronary arteries in patients with CAV—a feature that distinguishes it from atherosclerosis (96, 97). Additionally, analysis of clonal expansion of B cells using next generation sequencing of single cell methods revealed that specific B cell clones undergo robust expansion within the allograft (98).

These B and plasma cells, along with T cells and macrophages, are frequently organized into tertiary lymphoid-like structures within the adventitia (97, 99). While initially hypothesized to be a local source of DSAs, these nodules of B cells were found to exhibit a polyreactive profile, with a majority of cells secreting natural antibodies that react to autoantigens (100). One such autoantigen is vimentin, an intermediate filament protein that is expressed within the cytosol of smooth muscle cells and fibroblasts, where antibodies against it have been predictive of vasculopathy in heart allografts (101, 102). Although the exact mechanism by which intracellular vimentin expression contributes to the production of anti-vimentin antibodies during CAV remains unclear, it has been shown that vimentin becomes a target of caspases during inflammation, leading to the exposure of antigenic vimentin in apoptotic cells (103). Further research is needed to elucidate the processing and presentation of vimentin, as well as other “self” targets, to B cells, driving the production of these autoantibodies.

The generation of de novo DSAs by B cells plays a critical role in the development and progression of CAV through antibody-mediated rejection mechanisms (Figure 1) (104). B cells residing within the grafted endothelium are actively generating DSAs against donor MHC-I molecules (105). These antibodies have long been linked to CAV, where the extent of HLA mismatches is associated with allograft rejection (106108) Indeed, the transfer of DSAs targeting MHC-I has been shown to initiate endothelial inflammation followed by the development of CAV, independent of complement fixation that often accompanies acute antibody-mediated rejection (109, 110). In an intriguing murine model of CAV, CCR5-deficient and CD8-deficient mice, transiently treated with anti-CD4 therapy, developed an exaggerated antibody response after cardiac transplantation, leading to the generation of DSAs and subsequent vasculopathy (56). Notably, variability in DSA titers within this model closely correlated with the severity of vasculopathy, reflecting similar observations in human heart transplantation patients (56, 111). Thus, the dual role of B cells in producing both donor-specific and self-reactive antibodies significantly contributes to endothelial injury, amplifies immune responses, and drives the development of chronic allograft vasculopathy.

Antibodies play a multifaceted role in activating immune responses during allograft rejection. One of the most well defined roles of antibodies is their ability to activate the classical pathway of the complement cascade, synergizing their direct effects on immune cells during vasculopathy (96). Upon binding to antibodies, C1 activates the complement cascade by cleaving C4 and C2, forming the C4b2a complex, also known as C3 convertase. This enzyme cleaves C3 into two fragments, C3a and C3b. C3b is a chief component of the complement system as it can readily coat pathogens to promote their clearance, combine with other components of the complement system to form the membrane attack complex (MAC), and initiate a self-perpetuating amplification loop to produce more C3b (96). Additionally, many of the cleavage products of the complement system serve as chemoattractant signals to recruit neutrophils, monocytes, NK cells, B cells, and T cells to the vessel.

Antibodies also exert direct effects on endothelial and smooth muscle cells within the coronary arteries. DSAs for HLA class I antigens modulate endothelial cell function by stimulating the release of von Willebrand factor and P-selectin, leading to the aggregation of platelets and recruitment of circulating monocytes (112). Engagement of MHC class I molecules by antibodies increased expression of fibroblast growth factor receptor enhancing the proliferative responses of vascular smooth muscle cells (113). Furthermore, clinical biopsies of cardiac allografts undergoing antibody-mediated rejection and mouse models with high titers of DSA revealed upregulation of the Notch ligand Dll4, specifically with the lesions of large arteries (56, 114). Interestingly, endothelial Dll4 induces inflammatory polarization of macrophages leading to their production of the inflammatory cytokine IL-6 (114). Lastly, many immune cells including macrophages and NK cells contain receptors that recognize the Fc region of antibodies (Figure 1). Engagement of the FcγRIII(CD16a) receptor on NK cells initiates antibody-dependent cell-mediated cytotoxicity on the endothelium (82). Endothelial cell death may further diversify the antigenic load of the allograft (83), possibly leading to the production of non-DSA autoantigens such as vimentin.

In summary, antibodies orchestrate a complex and multifaceted immune response during allograft rejection by activating the complement cascade, recruiting and modulating immune cells, directly influencing vascular cells, and promoting inflammatory and cytotoxic pathways. These diverse mechanisms underscore the central role of antibodies in driving vasculopathy and immune-mediated injury, ultimately contributing to allograft dysfunction and failure.

T cells

T cells are key players in cellular-mediated rejection, which is often associated with acute rejection, but their contributions to chronic rejection and allograft vasculopathy remain less clearly understood. T cells do contribute to chronic rejection as mice lacking an adaptive immune system fail to reject cardiac allografts until activated T cells are transferred exogenously (89). In humans, both CD4 + and CD8+ T cells infiltrated the intima and adventitia of large coronary arteries associated with allograft vasculopathy (115). In the absence of CD4+ T cells, primed CD8+ T cells were sufficient to develop robust CAV (91). Deletion of effector molecules within these CD8+ T cells revealed that these cells perform a distinct IFN-γ-dependent mechanism to promote vasculopathy along with direct cytolysis. During acute rejection of cardiac allografts, TCR sequencing reveals a robust expansion of cytotoxic CD8+ T cell clones (86). However, in patients with an HLA-mismatched graft and who developed vasculopathy, the T cell repertoire did not differ significantly between the circulating blood and grafted tissue indicating a lack of clonal expansion within the allograft (45, 116). In contrast, the repertoire of BCR sequences with the graft minimally overlapped with the circulating B cells providing further evidence that chronic rejection may be driven more by B cell expansion (98).

While T cells may now be considered “bystanders” in the immune processes driving chronic vasculopathy, they contribute significantly to the inflammatory environment within the vasculature, even in the absence of antigen specificity. Infiltrating T cells are key producers of IFN-γ and TGF-β, two critical mediators of inflammation and fibrosis that drive the pathophysiology of CAV (116, 117). Additionally, endothelial cells upregulate nitric oxide signaling in bystander CD8+ T cells via iNOS expression, a process linked to vascular dysfunction (118, 119). In contrast, regulatory T cells (Tregs) are a specialized subset of T cells that play a critical role in maintaining immune homeostasis by suppressing excessive immune responses and promoting tolerance to self and non-self-antigens. Although Tregs are not detected in large numbers within coronary arteries with CAV lesions (117), their expansion within cardiac allografts has been shown to attenuate CAV progression and delay chronic rejection (120122) Conversely, depletion of Tregs leads to uncontrolled activation of NK cells, which accelerates CAV progression (123, 124). Interestingly, strategies that block memory T cell activation or deplete gamma delta T cells have been shown to promote Treg expansion (124, 125). Together, these findings illustrate that distinct T cell populations play active and opposing roles in shaping the inflammatory and fibrotic environment that drives the progression of CAV (Figure 1).

Translating bioinformatics into clinical practice

Applications

The rapid advancements in single-cell sequencing, spatial transcriptomics, proteomics, metabolomics, and other bioinformatic techniques have revolutionized our understanding of the molecular and cellular mechanisms underlying the development of CAV in heart transplant patients (Table 1). These technologies have not only deepened our knowledge of disease progression in humans but also enhanced our ability to refine animal models, improving the gap between preclinical studies and human pathology. Importantly, as these techniques continue to evolve, they bring us closer to their application in clinical practice. Ultimately, these powerful tools will transform patient care by enabling more accurate prediction, early diagnosis, and personalized management of rejection and vasculopathy.

Gene expression profiling has long been recognized as a powerful tool for non-invasive prediction of cardiac vasculopathy, offering an alternative to the more invasive endomyocardial biopsy. By analyzing gene expression patterns in transplant patients' blood across multiple centers, researchers identified a set of genes optimized for detecting acute allograft rejection (126). This discovery led to the development of AlloMap, a clinical tool that non-invasively predicts acute cellular rejection by profiling the expression of these specific genes. While AlloMap has demonstrated a strong negative predictive value for ruling out rejection, its positive predictive value is limited, particularly in predicting CAV, as it was specifically designed for acute cellular rejection surveillance (127). More recent advancements in gene expression analysis have identified rejection-associated transcripts that include many key molecular mediators of immunity discussed in the preceding sections of this review. These rejection-associated transcripts have shown improved utility in diagnosing antibody-mediated rejection from endomyocardial biopsies, offering a more precise tool for detecting rejection and CAV (128, 129). Beyond traditional transcriptomics, emerging technologies like spatial transcriptomics hold immense promise for advancing clinical care (130). This cutting-edge platform allows for the spatial visualization of gene expression within specific structures, such as coronary arteries, enabling direct assessment of pathological lesions from biopsies. Though still in its infancy, particularly in heart transplantation, spatial transcriptomics has the potential to revolutionize the diagnosis and treatment of CAV by providing unparalleled insight into the molecular and spatial landscape of the disease. As these technologies mature, they promise to redefine the standard of care for transplant patients, paving the way for more precise, personalized, and effective interventions.

A unique and innovative application of sequencing technologies in transplantation is the measurement of donor-derived cell-free DNA (dd-cfDNA) in the blood of recipients. This biomarker reflects tissue damage, as cell-free DNA is released from damaged donor cells into the recipient's bloodstream. Initially utilized to predict acute rejection after heart transplantation, elevated dd-cfDNA levels have been strongly associated with acute rejection episodes (131). In the context of chronic rejection, higher dd-cfDNA levels in patients with CAV have been linked to the presence of de novo DSAs and the development of vasculopathy (132, 133). Despite these promising associations, the relationship between dd-cfDNA levels and the severity of CAV remains unclear and warrants further investigation. Overall, measuring dd-cfDNA has become a critical tool in the clinical management of solid organ transplantation, offering a non-invasive method to monitor graft health and predict rejection.

The widespread and deep profiling of other biomolecules, including proteins and metabolites, offer significant potential for improving the clinical management of heart transplant patients. Proteomics has been employed to identify novel biomarkers associated with an increased risk of post-graft dysfunction (134). Additionally, urinary proteomic signatures are a potential non-invasive tool for the surveillance of CAV progression (135). Similarly, metabolomics is gaining traction in transplantation research. For instance, studies have shown that ex vivo perfusion can normalize the metabolomes of hearts procured from deceased cardiac donors and those from brain-dead donors, potentially improving organ viability and function prior to transplantation (136). While the development of these tools may be lagging behind the translation of transcriptomics into clinical practice, these advancements underscore the promise of proteomics, metabolomics, and other “-omics” in accelerating the discovery of biomarkers that will enhance the diagnostic and prognostic strategies after heart transplantation.

Challenges and future directions

The promise and application of bioinformatics at both the bench and bedside are advancing rapidly, revolutionizing our understanding of chronic allograft vasculopathy. However, it is essential to recognize their current limitations and how these constraints may affect their clinical and research applications. High throughput approaches require the processing of a large quantity of data, which requires advanced and rigorous analyses to determine the underlying biological information. Proteomic and metabolomic tools that rely on mass spectrometry for biomolecule quantification face significant challenges in standardization and sensitivity, which hinder their translation into clinical diagnostic tests (137). For instance, the metabolome is highly sensitive to environmental and technical factors, making it difficult to achieve consistent results across clinical laboratories. Additionally, variation among samples processed through the same mass spectrometry pipeline can exceed biological differences, complicating the identification of true metabolic changes (138, 139). Overcoming these obstacles remains a significant challenge for the future application of these mass spectrometry-based technologies in both the clinical and research setting.

Single-cell RNA sequencing, while a powerful tool for delineating cell-specific gene expression, is subject to inherent biases such as dropout events, low RNA capture efficiency, and batch effects, all of which can confound downstream analyses (140142). For instance, the high cycle numbers required for polymerase chain reaction amplification can reduce the detection and analysis of low-abundance RNA transcripts, potentially obscuring biologically significant signals. However, rapidly evolving spatial transcriptomic and single cell sequencing methodologies are enhancing the sensitivity and resolution of these technologies, enabling the capture of greater transcriptomic complexity within individual cells (143). Despite these advancements, one of the most significant challenges in large-scale biological data analysis is the presence of unwanted variation, specifically “batch effects”, which represent variation originating from technical differences across samples that are unrelated to the biological variables being studied. Excitingly, new computational tools continue to expand the bioinformatician's toolbox, with methods such as Harmony (144) and Seurat (145) offering powerful approaches to mitigate batch effects while preserving true biological variance, ultimately improving the robustness and interpretability of single-cell analyses (146).

The most successful emerging application of large-scale biological data within the field of transplantation is the ability to measure dd-cfDNA using a blood test, however there are significant limitations of this approach. Initially sought to be a specific marker of allograft rejection, dd-cfDNA is more indicative of allograft tissue injury and cannot necessarily distinguish between acute or chronic rejection. Furthermore, economic analyses suggest that the cost of biomarkers, such as dd-cfDNA, may actually be less cost effective than the typical screening with protocol biopsy (147). As bioinformatics and “-omics” technologies continue to evolve, their integration into clinical transplantation has the potential to revolutionize diagnostics and patient management. However, overcoming technical challenges such as standardization, sensitivity, and cost-effectiveness remains crucial to ensuring their successful translation from research to routine clinical practice.

Conclusion

The application of computational approaches, such as single-cell sequencing, spatial transcriptomics, proteomics, and metabolomics, has significantly enhanced our understanding of chronic allograft vasculopathy and its complex pathophysiology. These tools have provided unparalleled insights into the molecular mechanisms driving CAV, including the intricate immune-endothelial interactions and novel cellular populations contributing to endothelial dysfunction and intimal thickening. Understanding the immune landscape of CAV, particularly the roles of macrophages, NK cells, T cells, and antibodies, will be critical for developing targeted therapies to enhance graft survival and mitigate chronic rejection. Additionally, they have led to the identification of new biomarkers that could improve early diagnosis, prognosis, and monitoring for vasculopathy, a typically challenging diagnosis requiring invasive imaging. Despite the promising advances, the clinical application of these technologies is still in its early stages. However, the potential to transform the diagnosis, treatment, and prevention of CAV is substantial. As these technologies become more refined, accessible, and affordable, they could revolutionize how we manage transplant patients. As these technologies grow more complex in both methodology and analysis, developing standardized platforms will be crucial to streamline workflows and enhance data interpretation in both research and clinical settings. By bridging the gap between molecular research and clinical practice, these innovations hold the potential to reshape CAV management, offering hope for more effective strategies that address chronic allograft rejection and improve the long-term survival of transplant recipients.

Author contributions

ET: Conceptualization, Funding acquisition, Writing – original draft, Writing – review & editing. AA: Visualization, Writing – original draft, Writing – review & editing. CL: Conceptualization, Funding acquisition, Visualization, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. Publication of this research was supported by the Sidney and Bess Eisenberg Memorial Fund. Additionally, the work was supported by funding through the American Heart Association and National Institutes of Health Grants to EBT (R01HL122309, R01HL159964, R35HL177401, and the AHA Project Award 24SFRNPCN1289611).

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 author(s) declare that no Generative 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

1. Dharmavaram N, Hess T, Jaeger H, Smith J, Hermsen J, Murray D, et al. National trends in heart donor usage rates: are we efficiently transplanting more hearts? J Am Heart Assoc. (2021) 10:e019655. doi: 10.1161/JAHA.120.019655

PubMed Abstract | Crossref Full Text | Google Scholar

2. Khush KK, Cherikh WS, Chambers DC, Harhay MO, Hayes D, Hsich E, et al. The international thoracic organ transplant registry of the international society for heart and lung transplantation: thirty-sixth adult heart transplantation report — 2019; focus theme: donor and recipient size match. J Heart Lung Transplant. (2019) 38:1056–66. doi: 10.1016/j.healun.2019.08.004

PubMed Abstract | Crossref Full Text | Google Scholar

3. Ross M, Kouretas P, Gamberg P, Miller J, Burge M, Reitz B, et al. Ten- and 20-year survivors of pediatric orthotopic heart transplantation. J Heart Lung Transplant. (2006) 25:261–70. doi: 10.1016/j.healun.2005.09.011

PubMed Abstract | Crossref Full Text | Google Scholar

4. Pober JS, Chih S, Kobashigawa J, Madsen JC, Tellides G. Cardiac allograft vasculopathy: current review and future research directions. Cardiovasc Res. (2021) 117:2624–38. doi: 10.1093/cvr/cvab259

PubMed Abstract | Crossref Full Text | Google Scholar

5. Angelini A, Castellani C, Fedrigo M, de Boer OJ, Meijer-Jorna LB, Li X, et al. Coronary cardiac allograft vasculopathy versus native atherosclerosis: difficulties in classification. Virchows Arch. (2014) 464:627–35. doi: 10.1007/s00428-014-1586-6

PubMed Abstract | Crossref Full Text | Google Scholar

6. Roy P, Orecchioni M, Ley K. How the immune system shapes atherosclerosis: roles of innate and adaptive immunity. Nat Rev Immunol. (2022) 22:251–65. doi: 10.1038/s41577-021-00584-1

PubMed Abstract | Crossref Full Text | Google Scholar

7. Salomon RN, Hughes CC, Schoen FJ, Payne DD, Pober JS, Libby P. Human coronary transplantation-associated arteriosclerosis. Evidence for a chronic immune reaction to activated graft endothelial cells. Am J Pathol. (1991) 138:791–8.2012171

PubMed Abstract | Google Scholar

8. Castellani C, Angelini A, de Boer OJ, van der Loos CM, Fedrigo M, Frigo AC, et al. Intraplaque hemorrhage in cardiac allograft vasculopathy. Am J Transplant. (2014) 14:184–92. doi: 10.1111/ajt.12517

PubMed Abstract | Crossref Full Text | Google Scholar

9. Michos ED, McEvoy JW, Blumenthal RS. Lipid management for the prevention of atherosclerotic cardiovascular disease. N Engl J Med. (2019) 381:1557–67. doi: 10.1056/NEJMra1806939

PubMed Abstract | Crossref Full Text | Google Scholar

10. Raichlin E, Bae J-H, Khalpey Z, Edwards BS, Kremers WK, Clavell AL, et al. Conversion to sirolimus as primary immunosuppression attenuates the progression of allograft vasculopathy after cardiac transplantation. Circulation. (2007) 116:2726–33. doi: 10.1161/CIRCULATIONAHA.107.692996

PubMed Abstract | Crossref Full Text | Google Scholar

11. Eisen HJ, Tuzcu EM, Dorent R, Kobashigawa J, Mancini D, Kaeppler HAV-v, et al. Everolimus for the prevention of allograft rejection and vasculopathy in cardiac-transplant recipients. N Engl J Med. (2003) 349:847–58. doi: 10.1056/NEJMoa022171

PubMed Abstract | Crossref Full Text | Google Scholar

12. Guethoff S, Meiser BM, Groetzner J, Eifert S, Grinninger C, Ueberfuhr P, et al. Ten-Year results of a randomized trial comparing tacrolimus versus cyclosporine A in combination with mycophenolate mofetil after heart transplantation. Transplantation. (2013) 95. doi: 10.1097/TP.0b013e318277e378

PubMed Abstract | Crossref Full Text | Google Scholar

13. Kaczmarek I, Ertl B, Schmauss D, Sadoni S, Knez A, Daebritz S, et al. Preventing cardiac allograft vasculopathy: long-term beneficial effects of mycophenolate mofetil. J Heart Lung Transplant. (2006) 25:550–6. doi: 10.1016/j.healun.2006.01.003

PubMed Abstract | Crossref Full Text | Google Scholar

14. Weis M, Weis M. Transplant vasculopathy versus native atherosclerosis: similarities and differences. Transplantation. (2024) 108:1342–9. doi: 10.1097/tp.0000000000004853

PubMed Abstract | Crossref Full Text | Google Scholar

15. Long X, Yuan X, Du J. Single-cell and spatial transcriptomics: advances in heart development and disease applications. Comput Struct Biotechnol J. (2023) 21:2717–31. doi: 10.1016/j.csbj.2023.04.007

PubMed Abstract | Crossref Full Text | Google Scholar

16. Truby LK, Maamari D, Saha A, Farr M, Abdulrahim J, Billia F, et al. Towards allograft longevity: leveraging omics technologies to improve heart transplant outcomes. Curr Heart Fail Rep. (2023) 20:493–503. doi: 10.1007/s11897-023-00631-z

PubMed Abstract | Crossref Full Text | Google Scholar

17. Nevarez-Mejia J, Pickering H, Sosa RA, Valenzuela NM, Fishbein GA, Baldwin WM, et al. Spatial multiomics of arterial regions from cardiac allograft vasculopathy rejected grafts reveal novel insights into the pathogenesis of chronic antibody-mediated rejection. Am J Transplant. (2024) 24:1146–60. doi: 10.1016/j.ajt.2024.01.004

PubMed Abstract | Crossref Full Text | Google Scholar

18. Robertson H, Kim HJ, Li J, Robertson N, Robertson P, Jimenez-Vera E, et al. Decoding the hallmarks of allograft dysfunction with a comprehensive pan-organ transcriptomic atlas. Nat Med. (2024) 30:3748–57. doi: 10.1038/s41591-024-03030-6

PubMed Abstract | Crossref Full Text | Google Scholar

19. Li X, Turaga D, Li RG, Tsai C-R, Quinn JN, Zhao Y, et al. The macrophage landscape across the lifespan of a human cardiac allograft. Circulation. (2024) 149:1650–66. doi: 10.1161/CIRCULATIONAHA.123.065294

PubMed Abstract | Crossref Full Text | Google Scholar

20. Schmauss D, Weis M. Cardiac allograft vasculopathy. Circulation. (2008) 117:2131–41. doi: 10.1161/CIRCULATIONAHA.107.711911

PubMed Abstract | Crossref Full Text | Google Scholar

21. Avery RK. Cardiac-allograft vasculopathy. N Engl J Med. (2003) 349:829–30. doi: 10.1056/NEJMp038124

PubMed Abstract | Crossref Full Text | Google Scholar

22. Loupy A, Coutance G, Bonnet G, Van Keer J, Raynaud M, Aubert O, et al. Identification and characterization of trajectories of cardiac allograft vasculopathy after heart transplantation. Circulation. (2020) 141:1954–67. doi: 10.1161/CIRCULATIONAHA.119.044924

PubMed Abstract | Crossref Full Text | Google Scholar

23. Nicolás-Ávila JA, Lechuga-Vieco AV, Esteban-Martínez L, Sánchez-Díaz M, Díaz-García E, Santiago DJ, et al. A network of macrophages supports mitochondrial homeostasis in the heart. Cell. (2020) 183:94–109.e123. doi: 10.1016/j.cell.2020.08.031

PubMed Abstract | Crossref Full Text | Google Scholar

24. DeBerge M, Yeap XY, Dehn S, Zhang S, Grigoryeva L, Misener S, et al. MerTK cleavage on resident cardiac macrophages compromises repair after myocardial ischemia reperfusion injury. Circ Res. (2017) 121:930–40. doi: 10.1161/circresaha.117.311327

PubMed Abstract | Crossref Full Text | Google Scholar

25. Zhang S, Weinberg S, DeBerge M, Gainullina A, Schipma M, Kinchen JM, et al. Efferocytosis fuels requirements of fatty acid oxidation and the electron transport chain to polarize macrophages for tissue repair. Cell Metab. (2019) 29:443–56.e445. doi: 10.1016/j.cmet.2018.12.004

PubMed Abstract | Crossref Full Text | Google Scholar

26. Owen MC, Kopecky BJ. Targeting macrophages in organ transplantation: a step toward personalized medicine. Transplantation. (2024) 108:2045–56. doi: 10.1097/TP.0000000000004978

PubMed Abstract | Crossref Full Text | Google Scholar

27. Wu YL, Ye Q, Eytan DF, Liu L, Rosario BL, Hitchens TK, et al. Magnetic resonance imaging investigation of macrophages in acute cardiac allograft rejection after heart transplantation. Circ Cardiovasc Imaging. (2013) 6:965–73. doi: 10.1161/CIRCIMAGING.113.000674

PubMed Abstract | Crossref Full Text | Google Scholar

28. Glinton K, DeBerge M, Fisher E, Schroth S, Sinha A, Wang J-J, et al. Bone marrow-derived AXL tyrosine kinase promotes mitogenic crosstalk and cardiac allograft vasculopathy. J Heart Lung Transplant. (2021) 40:435–46. doi: 10.1016/j.healun.2021.03.006

PubMed Abstract | Crossref Full Text | Google Scholar

29. Kitchens WH, Chase CM, Uehara S, Cornell LD, Colvin RB, Russell PS, et al. Macrophage depletion suppresses cardiac allograft vasculopathy in mice. Am J Transplant. (2007) 7:2675–82. doi: 10.1111/j.1600-6143.2007.01997.x

PubMed Abstract | Crossref Full Text | Google Scholar

30. Glinton K, DeBerge M, Yeap X-Y, Zhang J, Forbess J, Luo X, et al. Acute and chronic phagocyte determinants of cardiac allograft vasculopathy. Semin Immunopathol. (2018) 40:593–603. doi: 10.1007/s00281-018-0699-4

PubMed Abstract | Crossref Full Text | Google Scholar

31. Liu X, Lu Y, Lian Y, Chen Z, Xia J, Meng L, et al. Macrophage depletion improves chronic rejection in rats with allograft heart transplantation. Transplant Proc. (2020) 52:992–1000. doi: 10.1016/j.transproceed.2019.12.037

PubMed Abstract | Crossref Full Text | Google Scholar

32. Hashimoto D, Chow A, Noizat C, Teo P, Beasley MB, Leboeuf M, et al. Tissue-Resident macrophages self-maintain locally throughout adult life with minimal contribution from circulating monocytes. Immunity. (2013) 38:792–804. doi: 10.1016/j.immuni.2013.04.004

PubMed Abstract | Crossref Full Text | Google Scholar

33. Schulz C, Perdiguero EG, Chorro L, Szabo-Rogers H, Cagnard N, Kierdorf K, et al. A lineage of myeloid cells independent of myb and hematopoietic stem cells. Science. (2012) 336:86–90. doi: 10.1126/science.1219179

PubMed Abstract | Crossref Full Text | Google Scholar

34. Epelman S, Lavine KJ, Beaudin AE, Sojka DK, Carrero JA, Calderon B, et al. Embryonic and adult-derived resident cardiac macrophages are maintained through distinct mechanisms at steady state and during inflammation. Immunity. (2014) 40:91–104. doi: 10.1016/j.immuni.2013.11.019

PubMed Abstract | Crossref Full Text | Google Scholar

35. Lavine KJ, Epelman S, Uchida K, Weber KJ, Nichols CG, Schilling JD, et al. Distinct macrophage lineages contribute to disparate patterns of cardiac recovery and remodeling in the neonatal and adult heart. Proc Natl Acad Sci USA. (2014) 111:16029–34. doi: 10.1073/pnas.1406508111

PubMed Abstract | Crossref Full Text | Google Scholar

36. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. (2010) 330:841–5. doi: 10.1126/science.1194637

PubMed Abstract | Crossref Full Text | Google Scholar

37. Leid J, Carrelha J, Boukarabila H, Epelman S, Jacobsen SEW, Lavine KJ. Primitive embryonic macrophages are required for coronary development and maturation. Circ Res. (2016) 118:1498–511. doi: 10.1161/CIRCRESAHA.115.308270

PubMed Abstract | Crossref Full Text | Google Scholar

38. Dick SA, Wong A, Hamidzada H, Nejat S, Nechanitzky R, Vohra S, et al. Three tissue resident macrophage subsets coexist across organs with conserved origins and life cycles. Science Immunology. (2022) 7:eabf7777. doi: 10.1126/sciimmunol.abf7777

PubMed Abstract | Crossref Full Text | Google Scholar

39. Dick SA, Macklin JA, Nejat S, Momen A, Clemente-Casares X, Althagafi MG, et al. Self-renewing resident cardiac macrophages limit adverse remodeling following myocardial infarction. Nat Immunol. (2019) 20:29–39. doi: 10.1038/s41590-018-0272-2

PubMed Abstract | Crossref Full Text | Google Scholar

40. Nahrendorf M, Swirski FK, Aikawa E, Stangenberg L, Wurdinger T, Figueiredo J-L, et al. The healing myocardium sequentially mobilizes two monocyte subsets with divergent and complementary functions. J Exp Med. (2007) 204:3037–47. doi: 10.1084/jem.20070885

PubMed Abstract | Crossref Full Text | Google Scholar

41. Bajpai G, Schneider C, Wong N, Bredemeyer A, Hulsmans M, Nahrendorf M, et al. The human heart contains distinct macrophage subsets with divergent origins and functions. Nat Med. (2018) 24:1234–45. doi: 10.1038/s41591-018-0059-x

PubMed Abstract | Crossref Full Text | Google Scholar

42. Malone AF, Wu H, Fronick C, Fulton R, Gaut JP, Humphreys BD. Harnessing expressed single nucleotide variation and single cell RNA sequencing to define immune cell chimerism in the rejecting kidney transplant. J Am Soc Nephrol. (2020) 31:1977–86. doi: 10.1681/ASN.2020030326

PubMed Abstract | Crossref Full Text | Google Scholar

43. Kang HM, Subramaniam M, Targ S, Nguyen M, Maliskova L, McCarthy E, et al. Multiplexed droplet single-cell RNA-sequencing using natural genetic variation. Nat Biotechnol. (2018) 36:89–94. doi: 10.1038/nbt.4042

PubMed Abstract | Crossref Full Text | Google Scholar

44. Kopecky BJ, Dun H, Amrute JM, Lin C-Y, Bredemeyer AL, Terada Y, et al. Donor macrophages modulate rejection after heart transplantation. Circulation. (2022) 146:623–38. doi: 10.1161/CIRCULATIONAHA.121.057400

PubMed Abstract | Crossref Full Text | Google Scholar

45. Amancherla K, Schlendorf KH, Chow N, Sheng Q, Freedman JE, Rathmell JC. Single-cell RNA-sequencing identifies unique cell-specific gene expression profiles in high-grade cardiac allograft vasculopathy. bioRxiv. (2024). 2007.2010.602989. doi: 10.1101/2024.07.10.602989

PubMed Abstract | Crossref Full Text | Google Scholar

46. Hodge S, Dean M, Hodge G, Holmes M, Reynolds PN. Decreased efferocytosis and mannose binding lectin in the airway in bronchiolitis obliterans syndrome. J Heart Lung Transplant. (2011) 30:589–95. doi: 10.1016/j.healun.2011.01.710

PubMed Abstract | Crossref Full Text | Google Scholar

47. Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. (2014) 510:92–101. doi: 10.1038/nature13479

PubMed Abstract | Crossref Full Text | Google Scholar

48. Chen G, Kheradmand T, Bryant J, Wang S, Tasch J, Wang Jj, et al. Intragraft CD11b + IDO+ cells mediate cardiac allograft tolerance by ECDI-fixed donor splenocyte infusions. Am J Transplant. (2012) 12:2920–9. doi: 10.1111/j.1600-6143.2012.04203.x

PubMed Abstract | Crossref Full Text | Google Scholar

49. Sun E, Gao Y, Chen J, Roberts AI, Wang X, Chen Z, et al. Allograft tolerance induced by donor apoptotic lymphocytes requires phagocytosis in the recipient. Cell Death Differ. (2004) 11:1258–64. doi: 10.1038/sj.cdd.4401500

PubMed Abstract | Crossref Full Text | Google Scholar

50. Bryant J, Lerret NM, Wang J-j, Kang H-K, Tasch J, Zhang Z, et al. Preemptive donor apoptotic cell infusions induce IFN-γ–producing myeloid-derived suppressor cells for cardiac allograft protection. J Immunol. (2014) 192:6092–101. doi: 10.4049/jimmunol.1302771

PubMed Abstract | Crossref Full Text | Google Scholar

51. Gallini R, Lindblom P, Bondjers C, Betsholtz C, Andrae J. PDGF-A and PDGF-B induces cardiac fibrosis in transgenic mice. Exp Cell Res. (2016) 349:282–90. doi: 10.1016/j.yexcr.2016.10.022

PubMed Abstract | Crossref Full Text | Google Scholar

52. Iida M, Tanabe K, Matsushima-Nishiwaki R, Kozawa O, Iida H. Adenosine monophosphate-activated protein kinase regulates platelet-derived growth factor-BB-induced vascular smooth muscle cell migration. Arch Biochem Biophys. (2013) 530:83–92. doi: 10.1016/j.abb.2012.12.019

PubMed Abstract | Crossref Full Text | Google Scholar

53. Chakarov S, Lim HY, Tan L, Lim SY, See P, Lum J, et al. Two distinct interstitial macrophage populations coexist across tissues in specific subtissular niches. Science. (2019) 363:eaau0964. doi: 10.1126/science.aau0964

PubMed Abstract | Crossref Full Text | Google Scholar

54. Xu L, Collins J, Drachenberg C, KuKuruga D, Burke A. Increased macrophage density of cardiac allograft biopsies is associated with antibody-mediated rejection and alloantibodies to HLA antigens. Clin Transplant. (2014) 28:554–60. doi: 10.1111/ctr.12348

PubMed Abstract | Crossref Full Text | Google Scholar

55. Lu W-h, Palatnik K, Fishbein GA, Lai C, Levi DS, Perens G, et al. Diverse morphologic manifestations of cardiac allograft vasculopathy: a pathologic study of 64 allograft hearts. J Heart Lung Transplant. (2011) 30:1044–50. doi: 10.1016/j.healun.2011.04.008

PubMed Abstract | Crossref Full Text | Google Scholar

56. Tsuda H, Dvorina N, Keslar KS, Nevarez-Mejia J, Valenzuela NM, Reed EF, et al. Molecular signature of antibody-mediated chronic vasculopathy in heart allografts in a novel mouse model. Am J Pathol. (2022) 192:1053–65. doi: 10.1016/j.ajpath.2022.04.003

PubMed Abstract | Crossref Full Text | Google Scholar

57. Huibers MMH, Vink A, Kaldeway J, Huisman A, Timmermans K, Leenders M, et al. Distinct phenotypes of cardiac allograft vasculopathy after heart transplantation: a histopathological study. Atherosclerosis. (2014) 236:353–9. doi: 10.1016/j.atherosclerosis.2014.07.016

PubMed Abstract | Crossref Full Text | Google Scholar

58. Lim HY, Lim SY, Tan CK, Thiam CH, Goh CC, Carbajo D, et al. Hyaluronan receptor LYVE-1-expressing macrophages maintain arterial tone through hyaluronan-mediated regulation of smooth muscle cell collagen. Immunity. (2018) 49:326–41.e327. doi: 10.1016/j.immuni.2018.06.008

PubMed Abstract | Crossref Full Text | Google Scholar

59. Nevarez-Mejia J, Jin Y-P, Pickering H, Parmar R, Valenzuela NM, Sosa RA, et al. Human leukocyte antigen class I antibody-activated endothelium promotes CD206+ M2 macrophage polarization and MMP9 secretion through TLR4 signaling and P-selectin in a model of antibody-mediated rejection and allograft vasculopathy. Am J Transplant. (2024) 24:406–18. doi: 10.1016/j.ajt.2023.10.020

PubMed Abstract | Crossref Full Text | Google Scholar

60. Watanabe R, Maeda T, Zhang H, Berry GJ, Zeisbrich M, Brockett R, et al. MMP (Matrix metalloprotease)-9–producing monocytes enable T cells to invade the vessel wall and cause vasculitis. Circ Res. (2018) 123:700–15. doi: 10.1161/CIRCRESAHA.118.313206

PubMed Abstract | Crossref Full Text | Google Scholar

61. Ankersmit HJ, Moser B, Roedler S, Teufel I, Zuckermann A, Roth G, et al. Death-inducing receptors and apoptotic changes in lymphocytes of patients with heart transplant vasculopathy. Clin Exp Immunol. (2002) 127:183–9. doi: 10.1046/j.1365-2249.2002.01741.x

PubMed Abstract | Crossref Full Text | Google Scholar

62. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. Functions of natural killer cells. Nat Immunol. (2008) 9:503–10. doi: 10.1038/ni1582

PubMed Abstract | Crossref Full Text | Google Scholar

63. Mocikat R, Braumüller H, Gumy A, Egeter O, Ziegler H, Reusch U, et al. Natural killer cells activated by MHC class ILow targets prime dendritic cells to induce protective CD8 T cell responses. Immunity. (2003) 19:561–9. doi: 10.1016/S1074-7613(03)00264-4

PubMed Abstract | Crossref Full Text | Google Scholar

64. Kim S, Iizuka K, Aguila HL, Weissman IL, Yokoyama WM. In vivo natural killer cell activities revealed by natural killer cell-deficient mice. Proc Natl Acad Sci USA. (2000) 97:2731–6. doi: 10.1073/pnas.050588297

PubMed Abstract | Crossref Full Text | Google Scholar

65. Kiessling R, Klein E, Wigzell H. Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol. (1975) 5:112–7. doi: 10.1002/eji.1830050208

PubMed Abstract | Crossref Full Text | Google Scholar

66. Caligiuri MA. Human natural killer cells. Blood. (2008) 112:461–9. doi: 10.1182/blood-2007-09-077438

PubMed Abstract | Crossref Full Text | Google Scholar

67. Cudkowicz G, Bennett M. Peculiar immunobiology of bone marrow allografts: i. Graft rejection by irradiated responder mice. J Exp Med. (1971) 134:83–102. doi: 10.1084/jem.134.1.83

PubMed Abstract | Crossref Full Text | Google Scholar

68. Cudkowicz G, Bennett M. Peculiar immunobiology of bone marrow allografts: iI. Rejection of parental grafts by resistant f1 hybrid mice. J Exp Med. (1971) 134:1513–28. doi: 10.1084/jem.134.6.1513

PubMed Abstract | Crossref Full Text | Google Scholar

69. Ljunggren H-G, Kärre K. In search of the ‘missing self’: mHC molecules and NK cell recognition. Immunol Today. (1990) 11:237–44. doi: 10.1016/0167-5699(90)90097-S

PubMed Abstract | Crossref Full Text | Google Scholar

70. Valiante NM, Uhrberg M, Shilling HG, Lienert-Weidenbach K, Arnett KL, D'Andrea A, et al. Functionally and structurally distinct NK cell receptor repertoires in the peripheral blood of two human donors. Immunity. (1997) 7:739–51. doi: 10.1016/S1074-7613(00)80393-3

PubMed Abstract | Crossref Full Text | Google Scholar

71. Koenig A, Chen C-C, Marçais A, Barba T, Mathias V, Sicard A, et al. Missing self triggers NK cell-mediated chronic vascular rejection of solid organ transplants. Nat Commun. (2019) 10:5350. doi: 10.1038/s41467-019-13113-5

PubMed Abstract | Crossref Full Text | Google Scholar

72. Uehara S, Chase CM, Kitchens WH, Rose HS, Colvin RB, Russell PS, et al. NK cells can trigger allograft vasculopathy: the role of hybrid resistance in solid organ Allografts1. J Immunol. (2005) 175:3424–30. doi: 10.4049/jimmunol.175.5.3424

PubMed Abstract | Crossref Full Text | Google Scholar

73. Zhang Z-X, Huang X, Jiang J, Lian D, Min W-p, Liu W, et al. Natural killer cells play a critical role in cardiac allograft vasculopathy in an interleukin-6–dependent manner. Transplantation. (2014) 98:1029–39. doi: 10.1097/TP.0000000000000405

PubMed Abstract | Crossref Full Text | Google Scholar

74. Graham JA, Wilkinson RA, Hirohashi T, Chase CM, Colvin RB, Madsen JC, et al. Viral infection induces de novo lesions of coronary allograft vasculopathy through a natural killer cell-dependent pathway. Am J Transplant. (2009) 9:2479–84. doi: 10.1111/j.1600-6143.2009.02801.x

PubMed Abstract | Crossref Full Text | Google Scholar

75. Maier S, Tertilt C, Chambron N, Gerauer K, Hüser N, Heidecke C-D, et al. Inhibition of natural killer cells results in acceptance of cardiac allografts in CD28−/− mice. Nat Med. (2001) 7:557–62. doi: 10.1038/87880

PubMed Abstract | Crossref Full Text | Google Scholar

76. Kim J, Chang CK, Hayden T, Liu F-C, Benjamin J, Hamerman JA, et al. The activating immunoreceptor NKG2D and its ligands are involved in allograft transplant Rejection1. J Immunol. (2007) 179:6416–20. doi: 10.4049/jimmunol.179.10.6416

PubMed Abstract | Crossref Full Text | Google Scholar

77. Chen H, Xia J, Zhang L, Jin X, Yang M, Li J, et al. NKG2D Blockade attenuated cardiac allograft vasculopathy in a mouse model of cardiac transplantation. Clin Exp Immunol. (2013) 173:544–52. doi: 10.1111/cei.12128

PubMed Abstract | Crossref Full Text | Google Scholar

78. Fabritius C, Ritschl PV, Resch T, Roth M, Ebner S, Günther J, et al. Deletion of the activating NK cell receptor NKG2D accelerates rejection of cardiac allografts. Am J Transplant. (2017) 17:3199–209. doi: 10.1111/ajt.14467

PubMed Abstract | Crossref Full Text | Google Scholar

79. Yu G, Xu X, Vu MD, Kilpatrick ED, Li XC. NK Cells promote transplant tolerance by killing donor antigen-presenting cells. J Exp Med. (2006) 203:1851–8. doi: 10.1084/jem.20060603

PubMed Abstract | Crossref Full Text | Google Scholar

80. Lin CM, Plenter RJ, Coulombe M, Gill RG. Interferon gamma and contact-dependent cytotoxicity are each rate limiting for natural killer cell–mediated antibody-dependent chronic rejection. Am J Transplant. (2016) 16:3121–30. doi: 10.1111/ajt.13865

PubMed Abstract | Crossref Full Text | Google Scholar

81. Hirohashi T, Chase CM, Della Pelle P, Sebastian D, Alessandrini A, Madsen JC, et al. A novel pathway of chronic allograft rejection mediated by NK cells and alloantibody. Am J Transplant. (2012) 12:313–21. doi: 10.1111/j.1600-6143.2011.03836.x

PubMed Abstract | Crossref Full Text | Google Scholar

82. Paul P, Picard C, Sampol E, Lyonnet L, Di Cristofaro J, Paul-Delvaux L, et al. Genetic and functional profiling of CD16-dependent natural killer activation identifies patients at higher risk of cardiac allograft vasculopathy. Circulation. (2018) 137:1049–59. doi: 10.1161/CIRCULATIONAHA.117.030435

PubMed Abstract | Crossref Full Text | Google Scholar

83. Parkes MD, Halloran PF, Hidalgo LG. Evidence for CD16a-mediated NK cell stimulation in antibody-mediated kidney transplant rejection. Transplantation. (2017) 101:e102–11. doi: 10.1097/TP.0000000000001586

PubMed Abstract | Crossref Full Text | Google Scholar

84. Agaugué S, Marcenaro E, Ferranti B, Moretta L, Moretta A. Human natural killer cells exposed to IL-2, IL-12, IL-18, or IL-4 differently modulate priming of naive T cells by monocyte-derived dendritic cells. Blood. (2008) 112:1776–83. doi: 10.1182/blood-2008-02-135871

PubMed Abstract | Crossref Full Text | Google Scholar

85. Schmauch E, Piening B, Mohebnasab M, Xia B, Zhu C, Stern J, et al. Integrative multi-omics profiling in human decedents receiving pig heart xenografts. Nat Med. (2024) 30:1448–60. doi: 10.1038/s41591-024-02972-1

PubMed Abstract | Crossref Full Text | Google Scholar

86. Kong D, Huang S, Miao X, Li J, Wu Z, Shi Y, et al. The dynamic cellular landscape of grafts with acute rejection after heart transplantation. J Heart Lung Transplant. (2023) 42:160–72. doi: 10.1016/j.healun.2022.10.017

PubMed Abstract | Crossref Full Text | Google Scholar

87. Goldberg JF, deFilippi CR, Lockhart C, McNair ER, Sinha SS, Kong H, et al. Proteomics in acute heart transplant rejection, on behalf of the GRAfT investigators. Transplantation. (2024). doi: 10.1097/TP.0000000000005258

PubMed Abstract | Crossref Full Text | Google Scholar

88. Tellides G, Pober JS. Interferon-γ axis in graft arteriosclerosis. Circ Res. (2007) 100:622–32. doi: 10.1161/01.RES.0000258861.72279.29

PubMed Abstract | Crossref Full Text | Google Scholar

89. Zecher D, Li Q, Williams AL, Walters JT, Baddoura FK, Chalasani G, et al. Innate immunity alone is not sufficient for chronic rejection but predisposes healed allografts to T cell-mediated pathology. Transpl Immunol. (2012) 26:113–8. doi: 10.1016/j.trim.2011.12.006

PubMed Abstract | Crossref Full Text | Google Scholar

90. Loupy A, Toquet C, Rouvier P, Beuscart T, Bories MC, Varnous S, et al. Late failing heart allografts: pathology of cardiac allograft vasculopathy and association with antibody-mediated rejection. Am J Transplant. (2016) 16:111–20. doi: 10.1111/ajt.13529

PubMed Abstract | Crossref Full Text | Google Scholar

91. Skaro AI, Liwski RS, Zhou J, Vessie EL, Lee TDG, Hirsch GM. CD8+ T cells mediate aortic allograft vasculopathy by direct killing and an interferon-γ-dependent indirect pathway. Cardiovasc Res. (2005) 65:283–91. doi: 10.1016/j.cardiores.2004.09.023

PubMed Abstract | Crossref Full Text | Google Scholar

92. Gareau A, Hirsch GM, Lee TDG, Nashan B. Contribution of B cells and antibody to cardiac allograft vasculopathy. Transplantation. (2009) 88:470–7. doi: 10.1097/TP.0b013e3181b076cc

PubMed Abstract | Crossref Full Text | Google Scholar

93. Shi C, Lee WS, He Q, Zhang D, Fletcher DL, Newell JB, et al. Immunologic basis of transplant-associated arteriosclerosis. Proc Natl Acad Sci USA. (1996) 93:4051–6. doi: 10.1073/pnas.93.9.4051

PubMed Abstract | Crossref Full Text | Google Scholar

94. Russell PS, Chase CM, Colvin RB. Alloantibody- and T cell-mediated immunity in the pathogenesis of transplant arteriosclerosis: lack of progression to sclerotic lesions in B cell-deficient mice: 1. Transplantation. (1997) 64:1531–6. doi: 10.1097/00007890-199712150-00005

PubMed Abstract | Crossref Full Text | Google Scholar

95. Gill RG. Bringing clarity to the murky problem of cardiac allograft vasculopathy. Am J Pathol. (2022) 192:986–9. doi: 10.1016/j.ajpath.2022.05.002

PubMed Abstract | Crossref Full Text | Google Scholar

96. Wehner J, Morrell CN, Reynolds T, Rodriguez ER, Baldwin WM. Antibody and complement in transplant vasculopathy. Circ Res. (2007) 100:191–203. doi: 10.1161/01.RES.0000255032.33661.88

PubMed Abstract | Crossref Full Text | Google Scholar

97. Wehner JR, Fox-Talbot K, Halushka MK, Ellis C, Zachary AA, Baldwin WM III. B cells and plasma cells in coronaries of chronically rejected cardiac transplants. Transplantation. (2010) 89:1141–8. doi: 10.1097/TP.0b013e3181d3f271

PubMed Abstract | Crossref Full Text | Google Scholar

98. Moore C, Gao B, Roskin KM, Vasilescu E-RM, Addonizio L, Givertz MM, et al. B cell clonal expansion within immune infiltrates in human cardiac allograft vasculopathy. Am J Transplant. (2020) 20:1431–8. doi: 10.1111/ajt.15737

PubMed Abstract | Crossref Full Text | Google Scholar

99. Huibers MMH, Gareau AJ, Vink A, Kruit R, Feringa H, Beerthuijzen JMT, et al. The composition of ectopic lymphoid structures suggests involvement of a local immune response in cardiac allograft vasculopathy. J Heart Lung Transplant. (2015) 34:734–45. doi: 10.1016/j.healun.2014.11.022

PubMed Abstract | Crossref Full Text | Google Scholar

100. Chatterjee D, Moore C, Gao B, Clerkin KJ, See SB, Shaked D, et al. Prevalence of polyreactive innate clones among graft-­infiltrating B cells in human cardiac allograft vasculopathy. J Heart Lung Transplant. (2018) 37:385–93. doi: 10.1016/j.healun.2017.09.011

PubMed Abstract | Crossref Full Text | Google Scholar

101. Jurcevic S, Ainsworth ME, Pomerance A, Smith JD, Robinson DR, Dunn MJ, et al. Antivimentin antibodies are an independent predictor of transplant-associated coronary artery disease after cardiac transplantation. Transplantation. (2001) 71:886–92. doi: 10.1097/00007890-200104150-00011

PubMed Abstract | Crossref Full Text | Google Scholar

102. Mahesh B, Leong H-S, Nair KS, McCormack A, Sarathchandra P, Rose ML. Autoimmunity to vimentin potentiates graft vasculopathy in murine cardiac allografts. Transplantation. (2010) 90:4–13. doi: 10.1097/TP.0b013e3181dfa694

PubMed Abstract | Crossref Full Text | Google Scholar

103. Morishima N. Changes in nuclear morphology during apoptosis correlate with vimentin cleavage by different caspases located either upstream or downstream of bcl-2 action. Genes Cells. (1999) 4:401–14. doi: 10.1046/j.1365-2443.1999.00270.x

PubMed Abstract | Crossref Full Text | Google Scholar

104. Mantell BS, Cordero H, See SB, Clerkin KJ, Vasilescu R, Marboe CC, et al. Transcriptomic heterogeneity of antibody mediated rejection after heart transplant with or without donor specific antibodies. J Heart Lung Transplant. (2021) 40:1472–80. doi: 10.1016/j.healun.2021.06.012

PubMed Abstract | Crossref Full Text | Google Scholar

105. Thaunat O, Field A-C, Dai J, Louedec L, Patey N, Bloch M-F, et al. Lymphoid neogenesis in chronic rejection: evidence for a local humoral alloimmune response. Proc Natl Acad Sci USA. (2005) 102:14723–8. doi: 10.1073/pnas.0507223102

PubMed Abstract | Crossref Full Text | Google Scholar

106. Reed EF, Hong B, Ho E, Harris PE, Weinberger J, Suciu-Foca N. Monitoring of soluble hla alloantigens and anti-hla antibodies identifies heart allograft recipients at risk of transplant-associated coronary artery disease. Transplantation. (1996) 61:566–72. doi: 10.1097/00007890-199602270-00009

PubMed Abstract | Crossref Full Text | Google Scholar

107. Rose EA, Pepino P, Barr ML, Smith CR, Ratner AJ, Ho E, et al. Relation of HLA antibodies and graft atherosclerosis in human cardiac allograft recipients. J Heart Lung Transplant. (1992) 11:S120–3.1622990

PubMed Abstract | Google Scholar

108. Crudele V, Cacciatore F, Grimaldi V, Maiello C, Romano G, Amarelli C, et al. Human leukocyte antigen-DR mismatch is associated with increased in-hospital mortality after a heart transplant. Exp Clin Transplant. (2013) 11:346–51. doi: 10.6002/ect.2012.0276

PubMed Abstract | Crossref Full Text | Google Scholar

109. Hirohashi T, Uehara S, Chase CM, DellaPelle P, Madsen JC, Russell PS, et al. Complement independent antibody-mediated endarteritis and transplant arteriopathy in mice. Am J Transplant. (2010) 10:510–7. doi: 10.1111/j.1600-6143.2009.02958.x

PubMed Abstract | Crossref Full Text | Google Scholar

110. Galvani S, Augé N, Calise D, Thiers JC, Canivet C, Kamar N, et al. HLA Class I antibodies provoke graft arteriosclerosis in human arteries transplanted into SCID/beige mice. Am J Transplant. (2009) 9:2607–14. doi: 10.1111/j.1600-6143.2009.02804.x

PubMed Abstract | Crossref Full Text | Google Scholar

111. Das BB, Lacelle C, Zhang S, Gao A, Fixler D. Complement (C1q) binding de novo donor-specific antibodies and cardiac-allograft vasculopathy in pediatric heart transplant recipients. Transplantation. (2018) 102:502–9. doi: 10.1097/tp.0000000000001944

PubMed Abstract | Crossref Full Text | Google Scholar

112. Yamakuchi M, Kirkiles-Smith NC, Ferlito M, Cameron SJ, Bao C, Fox-Talbot K, et al. Antibody to human leukocyte antigen triggers endothelial exocytosis. Proc Natl Acad Sci USA. (2007) 104:1301–6. doi: 10.1073/pnas.0602035104

PubMed Abstract | Crossref Full Text | Google Scholar

113. Bian H, Reed EF. Alloantibody-Mediated class I signal transduction in endothelial cells and smooth muscle cells: enhancement by IFN-γ and TNF-α1. J Immunol. (1999) 163:1010–8. doi: 10.4049/jimmunol.163.2.1010

PubMed Abstract | Crossref Full Text | Google Scholar

114. Pabois A, Pagie S, Gérard N, Laboisse C, Pattier S, Hulin P, et al. Notch signaling mediates crosstalk between endothelial cells and macrophages via Dll4 and IL6 in cardiac microvascular inflammation. Biochem Pharmacol. (2016) 104:95–107. doi: 10.1016/j.bcp.2016.01.016

PubMed Abstract | Crossref Full Text | Google Scholar

115. Russell PS, Chase CM, Winn HJ, Colvin RB. Coronary atherosclerosis in transplanted mouse hearts. I. Time course and immunogenetic and immunopathological considerations. Am J Pathol. (1994) 144:260–74.7906094

PubMed Abstract | Google Scholar

116. Habal MV, Miller AMI, Rao S, Lin S, Obradovic A, Khosravi-Maharlooei M, et al. T cell repertoire analysis suggests a prominent bystander response in human cardiac allograft vasculopathy. Am J Transplant. (2021) 21:1465–76. doi: 10.1111/ajt.16333

PubMed Abstract | Crossref Full Text | Google Scholar

117. Hagemeijer MC, Van Oosterhout MFM, Van Wichen DF, Van Kuik J, Siera-de Koning E, Gmelig Meyling FHJ, et al. T cells in cardiac allograft vasculopathy are skewed to memory th-1 cells in the presence of a distinct th-2 population. Am J Transplant. (2008) 8:1040–50. doi: 10.1111/j.1600-6143.2008.02198.x

PubMed Abstract | Crossref Full Text | Google Scholar

118. Koh KP, Wang Y, Yi T, Shiao SL, Lorber MI, Sessa WC, et al. T cell–mediated vascular dysfunction of human allografts results from IFN-γ dysregulation of NO synthase. J Clin Invest. (2004) 114:846–56. doi: 10.1172/JCI21767

PubMed Abstract | Crossref Full Text | Google Scholar

119. Choy JC, Wang Y, Tellides G, Pober JS. Induction of inducible NO synthase in bystander human T cells increases allogeneic responses in the vasculature. Proc Natl Acad Sci USA. (2007) 104:1313–8. doi: 10.1073/pnas.0607731104

PubMed Abstract | Crossref Full Text | Google Scholar

120. Ravichandran R, Itabashi Y, Fleming T, Bansal S, Bowen S, Poulson C, et al. Low-dose IL-2 prevents murine chronic cardiac allograft rejection: role for IL-2-induced T regulatory cells and exosomes with PD-L1 and CD73. Am J Transplant. (2022) 22:2180–94. doi: 10.1111/ajt.17101

PubMed Abstract | Crossref Full Text | Google Scholar

121. Nadig SN, Więckiewicz J, Wu DC, Warnecke G, Zhang W, Luo S, et al. In vivo prevention of transplant arteriosclerosis by ex vivo–expanded human regulatory T cells. Nat Med. (2010) 16:809–13. doi: 10.1038/nm.2154

PubMed Abstract | Crossref Full Text | Google Scholar

122. Warnecke G, Bushell A, Nadig SN, Wood KJ. Regulation of transplant arteriosclerosis by CD25 + CD4+ T cells generated to alloantigen in vivo. Transplantation. (2007) 83:1459–65. doi: 10.1097/01.tp.0000265446.61754.d2

PubMed Abstract | Crossref Full Text | Google Scholar

123. Hirohashi T, Chase CM, DellaPelle P, Sebastian D, Farkesh E, Colvin RB, et al. Depletion of T regulatory cells promotes natural killer cell–mediated cardiac allograft vasculopathy. Transplantation. (2014) 98:828–34. doi: 10.1097/TP.0000000000000329

PubMed Abstract | Crossref Full Text | Google Scholar

124. Zhu H, Li J, Wang S, Liu K, Wang L, Huang L. Γδ T cell receptor deficiency attenuated cardiac allograft vasculopathy and promoted regulatory T cell expansion. Scand J Immunol. (2013) 78:44–9. doi: 10.1111/sji.12064

PubMed Abstract | Crossref Full Text | Google Scholar

125. Ge W, Jiang J, Liu W, Lian D, Saito A, Garcia B, et al. Regulatory T cells are critical to tolerance induction in presensitized mouse transplant recipients through targeting memory T cells. Am J Transplant. (2010) 10:1760–73. doi: 10.1111/j.1600-6143.2010.03186.x

PubMed Abstract | Crossref Full Text | Google Scholar

126. Pham MX, Teuteberg JJ, Kfoury AG, Starling RC, Deng MC, Cappola TP, et al. Gene-Expression profiling for rejection surveillance after cardiac transplantation. N Engl J Med. (2010) 362:1890–900. doi: 10.1056/NEJMoa0912965

PubMed Abstract | Crossref Full Text | Google Scholar

127. Yamani MH, Taylor DO, Rodriguez ER, Cook DJ, Zhou L, Smedira N, et al. Transplant vasculopathy is associated with increased AlloMap gene expression score. J Heart Lung Transplant. (2007) 26:403–6. doi: 10.1016/j.healun.2006.12.011

PubMed Abstract | Crossref Full Text | Google Scholar

128. Halloran PF, Potena L, Van Huyen J-PD, Bruneval P, Leone O, Kim DH, et al. Building a tissue-based molecular diagnostic system in heart transplant rejection: the heart molecular microscope diagnostic (MMDx) system. J Heart Lung Transplant. (2017) 36:1192–200. doi: 10.1016/j.healun.2017.05.029

PubMed Abstract | Crossref Full Text | Google Scholar

129. Loupy A, Duong Van Huyen JP, Hidalgo L, Reeve J, Racapé M, Aubert O, et al. Gene expression profiling for the identification and classification of antibody-mediated heart rejection. Circulation. (2017) 135:917–35. doi: 10.1161/CIRCULATIONAHA.116.022907

PubMed Abstract | Crossref Full Text | Google Scholar

130. Li JSY, Raghubar AM, Matigian NA, Ng MSY, Rogers NM, Mallett AJ. The utility of spatial transcriptomics for solid organ transplantation. Transplantation. (2023) 107:1463–71. doi: 10.1097/TP.0000000000004466

PubMed Abstract | Crossref Full Text | Google Scholar

131. De Vlaminck I, Valantine HA, Snyder TM, Strehl C, Cohen G, Luikart H, et al. Circulating cell-free DNA enables noninvasive diagnosis of heart transplant rejection. Sci Transl Med. (2014) 6:241ra277–241ra277. doi: 10.1126/scitranslmed.3007803

PubMed Abstract | Crossref Full Text | Google Scholar

132. Holzhauser L, Clerkin KJ, Fujino T, Alenghat FJ, Raikhelkar J, Kim G, et al. Donor-derived cell-free DNA is associated with cardiac allograft vasculopathy. Clin Transplant. (2021) 35:e14206. doi: 10.1111/ctr.14206

PubMed Abstract | Crossref Full Text | Google Scholar

133. Khush KK, Patel J, Pinney S, Kao A, Alharethi R, DePasquale E, et al. Noninvasive detection of graft injury after heart transplant using donor-derived cell-free DNA: a prospective multicenter study. Am J Transplant. (2019) 19:2889–99. doi: 10.1111/ajt.15339

PubMed Abstract | Crossref Full Text | Google Scholar

134. Truby LK, Kwee LC, Agarwal R, Grass E, DeVore AD, Patel CB, et al. Proteomic profiling identifies CLEC4C expression as a novel biomarker of primary graft dysfunction after heart transplantation. J Heart Lung Transplant. (2021) 40:1589–98. doi: 10.1016/j.healun.2021.07.024

PubMed Abstract | Crossref Full Text | Google Scholar

135. Wei D, Trenson S, Van Keer JM, Melgarejo J, Cutsforth E, Thijs L, et al. The novel proteomic signature for cardiac allograft vasculopathy. ESC Heart Failure. (2022) 9:1216–27. doi: 10.1002/ehf2.13796

PubMed Abstract | Crossref Full Text | Google Scholar

136. Hautbergue T, Laverdure F, Van SD, Vallee A, Sanchis-Borja M, Decante B, et al. Metabolomic profiling of cardiac allografts after controlled circulatory death. J Heart Lung Transplant. (2023) 42:870–9. doi: 10.1016/j.healun.2023.02.1492

PubMed Abstract | Crossref Full Text | Google Scholar

137. Deelen J, Kettunen J, Fischer K, van der Spek A, Trompet S, Kastenmüller G, et al. A metabolic profile of all-cause mortality risk identified in an observational study of 44,168 individuals. Nat Commun. (2019) 10:3346. doi: 10.1038/s41467-019-11311-9

PubMed Abstract | Crossref Full Text | Google Scholar

138. Deng K, Zhang F, Tan Q, Huang Y, Song W, Rong Z, et al. WaveICA: a novel algorithm to remove batch effects for large-scale untargeted metabolomics data based on wavelet analysis. Anal Chim Acta. (2019) 1061:60–9. doi: 10.1016/j.aca.2019.02.010

PubMed Abstract | Crossref Full Text | Google Scholar

139. Han W, Li L. Evaluating and minimizing batch effects in metabolomics. Mass Spectrom Rev. (2022) 41:421–42. doi: 10.1002/mas.21672

PubMed Abstract | Crossref Full Text | Google Scholar

140. Kharchenko PV. The triumphs and limitations of computational methods for scRNA-seq. Nat Methods. (2021) 18:723–32. doi: 10.1038/s41592-021-01171-x

PubMed Abstract | Crossref Full Text | Google Scholar

141. Leek JT, Scharpf RB, Bravo HC, Simcha D, Langmead B, Johnson WE, et al. Tackling the widespread and critical impact of batch effects in high-throughput data. Nat Rev Genet. (2010) 11:733–9. doi: 10.1038/nrg2825

PubMed Abstract | Crossref Full Text | Google Scholar

142. Jackson CA, Vogel C. New horizons in the stormy sea of multimodal single-cell data integration. Mol Cell. (2022) 82:248–59. doi: 10.1016/j.molcel.2021.12.012

PubMed Abstract | Crossref Full Text | Google Scholar

143. Kleino I, Frolovaitė P, Suomi T, Elo LL. Computational solutions for spatial transcriptomics. Comput Struct Biotechnol J. (2022) 20:4870–84. doi: 10.1016/j.csbj.2022.08.043

PubMed Abstract | Crossref Full Text | Google Scholar

144. Korsunsky I, Millard N, Fan J, Slowikowski K, Zhang F, Wei K, et al. Fast, sensitive and accurate integration of single-cell data with harmony. Nat Methods. (2019) 16:1289–96. doi: 10.1038/s41592-019-0619-0

PubMed Abstract | Crossref Full Text | Google Scholar

145. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM, et al. Comprehensive integration of single-cell data. Cell. (2019) 177:1888–902.e1821. doi: 10.1016/j.cell.2019.05.031

PubMed Abstract | Crossref Full Text | Google Scholar

146. Arevalo J, Su E, Ewald JD, van Dijk R, Carpenter AE, Singh S. Evaluating batch correction methods for image-based cell profiling. Nat Commun. (2024) 15:6516. doi: 10.1038/s41467-024-50613-5

PubMed Abstract | Crossref Full Text | Google Scholar

147. Puttarajappa CM, Mehta RB, Roberts MS, Smith KJ, Hariharan S. Economic analysis of screening for subclinical rejection in kidney transplantation using protocol biopsies and noninvasive biomarkers. Am J Transplant. (2021) 21:186–97. doi: 10.1111/ajt.16150

PubMed Abstract | Crossref Full Text | Google Scholar

148. Almufleh A, Zhang L, Mielniczuk LM, Stadnick E, Davies RA, Du Q, et al. Biomarker discovery in cardiac allograft vasculopathy using targeted aptamer proteomics. Clin Transplant. (2020) 34:e13765. doi: 10.1111/ctr.13765

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: immunity, bioinformatics & computational biology, cardiac allograft vasculopathy (CAV), macrophage - cell, transplantation (heart)

Citation: Thorp EB, Ananthakrishnan A and Lantz CW (2025) Decoding immune cell interactions during cardiac allograft vasculopathy: insights derived from bioinformatic strategies. Front. Cardiovasc. Med. 12:1568528. doi: 10.3389/fcvm.2025.1568528

Received: 30 January 2025; Accepted: 4 April 2025;
Published: 24 April 2025.

Edited by:

Masanori Aikawa, Brigham and Women's Hospital and Harvard Medical School, United States

Reviewed by:

Laura DiChiacchio, Cedars Sinai Medical Center, United States
Stefano Casarin, Houston Methodist Research Institute, United States

Copyright: © 2025 Thorp, Ananthakrishnan and Lantz. 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: Connor W. Lantz, Y29ubm9yLmxhbnR6QG5vcnRod2VzdGVybi5lZHU=

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.