Elsevier

Journal of Autoimmunity

Volume 45, September 2013, Pages 80-89
Journal of Autoimmunity

Review
Learning from rejection: What transplantation teaches us about (other) vascular pathologies

https://doi.org/10.1016/j.jaut.2013.05.006Get rights and content

Abstract

Allograft vasculopathy is an accelerated intimal hyperplastic lesion leading to progressive vascular stenosis; it represents the major long-term limitation to successful solid organ transplant. Although allograft vasculopathy is not formally an autoimmune disease, nor does it constitute a major cause of cardiovascular disease on a purely numerical basis, its pathogenesis provides an important window on the mechanisms by which immune injury can drive more common vascular pathologic entities. Thus, insights gleaned from vascularized solid organ transplants can shed new mechanistic (and therapeutic) light on: 1) the intimal vascular responses accompanying typical atherosclerosis and other inflammatory vessel diseases (e.g., scleroderma); 2) the pathogenesis of vascular stenosis versus aneurysm formation; 3) the sources of intimal smooth muscle cells in the healing of any vascular injury; and 4) the mechanisms by which smooth muscle cells are recruited into intimal lesions. Indeed, research on allograft vasculopathy has led to the understanding that interferon-γ plays a similar pathogenic role in a host of vascular stenosing lesions—and that Th2 cytokines can drive vascular remodeling and aneurysm formation. Moreover, circulating precursors (and not just medial smooth muscle cells) contribute to the intimal hyperplasia seen in atherosclerosis and in-stent restenosis. That non-vessel smooth muscle cells can be recruited to sites of vessel injury further suggests that chemokine and adhesion molecule interactions may be viable targets to limit vascular stenosis in a wide range of vascular lesions. This review will describe the pathogenesis of allograft vasculopathy, and will relate how understanding the underlying pathways informs our understanding of both human transplant-associated disease, as well as other human vascular pathologies.

Introduction

Allograft vasculopathy (AV) is a progressive and diffuse intimal hyperplastic lesion of arteries that leads to insidious vessel narrowing, and eventually to allograft ischemia. As opposed to atherosclerosis which typically requires decades before becoming clinically significant, the pace of vessel stenosis in AV is accelerated, accruing over the course of just months to years. Even in an era of immunosuppressive therapies that can effectively block acute cellular and humoral rejection, AV remains the major cause of long-term allograft failure [1]; the “half-life” of most transplanted hearts, for example, remains stubbornly pinned at approximately a decade—largely unchanged since the 1980's [2].

In kidneys, the disease manifests as a progressive renal insufficiency; in liver, ischemic damage is reflected by bile duct drop-out; and in the lung—in addition to the vascular compromise—a kindred process leads to airway narrowing called bronchiolitis obliterans. In cardiac allografts, AV involvement of the coronary and intramyocardial arterioles results in gradual ischemic congestive heart failure and/or lethal arrhythmias.

Although AV has been called “chronic rejection”, it should not be considered the consequence of a smoldering parenchymal rejection or even an ongoing rejection of the vessel wall; indeed, AV can progress even in the absence of allospecific responses [3]. Rather, AV is probably better understood as a variation on stereotypical healing. Functionally, the repertoire of vessel responses after injury are extremely limited, and vascular repair—regardless of the underlying cause—ultimately funnels through a final common pathway that conscripts smooth muscle cells and extracellular matrix synthesis to buttress the “damaged” wall. Thus, the vascular wall thickening that accompanies atherosclerosis is a consequence of a lifetime of accumulated insults ranging from smoking to hypertension to hypercholesterolemia to diabetes. Similarly, the restenosis that occurs within months to years after venous grafting into arterial circulation or endovascular stenting results from the mechanical trauma (with associated thrombosis and inflammation) of the procedure and/or device. In the case of AV, immune-mediated vascular injury clearly initiates the process, but it is also worth noting that non-immune pathways (e.g., ischemic injury, free radical damage, hypercholesterolemia, etc.) can contribute. This is by way of justifying that lessons to be gleaned from AV will undoubtedly provide pathogenic insights (and suggest potential therapeutic targets) for the more common vascular lesions that confront cardiologists, rheumatologists, and vascular surgeons.

Given that this review appears in Autoimmunity, it may also be germane to point out that AV can actually be thought of as a self-directed immune response, except that “self” in this case is a transplanted organ. Such AV-associated intimal hyperplasia is also strongly reminiscent of the vascular changes that occur in the setting of scleroderma, where the fundamental target of autoimmune injury is the vessel wall [4]. Similar intimal lesions are also seen in vasculitis, where the endothelium and media are subjected to the manifold effects of inflammatory mediators secondarily recruited in the wake of immune complex deposition. Interestingly, graft-versus-host disease after mismatched bone marrow transplantation can also manifest as a vasculopathy that entirely mimics AV [5], [6].

The following will include an overview of the current thinking regarding the mechanisms that contribute to AV, highlighting the important contributions of innate and adaptive immunity, and emphasizing the role of interferon-γ (IFNγ). Additional insights from animal models and human disease will then be summarized as they pertain to vascular remodeling and aneurysm formation. After encapsulating the experiments that show the various sources of intimal smooth muscle cells, the review will conclude with a discussion of the mechanisms by which intimal smooth muscle cells may be recruited to sites of vascular injury. Throughout, it is worth comparing AV and more common lesions like atherosclerosis (Fig. 1). As noted, they likely share pathogenic mechanisms. Moreover, the relatively accelerated onset of AV makes it more amenable to time-course analysis, and because therapeutic end-points also occur earlier, AV may well represent a surrogate disease for developing effective interventions.

Section snippets

Overview: AV characteristics and pathogenesis [7]

Although allograft veins can develop AV lesions, the most clinically relevant effects are all in the arterial circulation. Thus, even though all vessels should be subject to similar immune injury, an arterial predilection probably reflects the consequences of higher shear stresses. It may also be a consequence of the greater volume of (or functionally different) medial smooth muscle cells. Regardless, it is noteworthy that other vascular pathologies, ranging from atherosclerosis to vasculitis,

Role of cytokines in vascular disease

Cytokines are central mediators in AV pathogenesis. Although innate and adaptive immune cells are traditionally considered the major producers, EC, SMC, and epithelium are also potent sources in the intragraft milieu [42], [43].

Interferonγ (IFNγ), produced primarily by infiltrating T-helper 1 cells (but also to a lesser extent by NK cells, CD8+ T cells, and dendritic cells), is the most important. Among other activities, it increases MHC I and II expression, potently drives macrophage

Vascular remodeling in vascular disease: stenosis or aneurysm?

Luminal stenosis is the major clinical consequence of AV, leading to allograft ischemia [7], [9]]. Such negative (inward) remodeling is chiefly attributable to intimal ECM and cell accumulation, but will also be influenced by vascular scarring and increased vasomotor tone. Conversely, positive (outward) remodeling can result from SMC and ECM turnover [58], maintaining a normal lumenal diameter even as the intima expands [59]. On the other hand, excessive matrix degradation or SMC loss can

Source of smooth muscle cells in vascular disease

Although long assumed to derive from the recruitment of medial SMC, the source of intimal SMC in vascular lesions is actually a bit more involved. Our understanding of these pathways owes much to the transplant literature and the understanding of AV pathogenesis.

It had been known for almost two decades that intimal SMC—comprising the bulk of the cellularity in atherosclerosis and other stenosing vascular pathologies—are phenotypically dissimilar from medial SMC, having substantially greater

Recruitment SMC into intimal lesions

A corollary to the observation that substantial numbers of SMC can derive from the circulation is that access to vessel walls almost certainly involves recruitment pathways akin to those used by inflammatory cells. The route of access is of more than just academic interest since these pathways become potential targets of therapy.

Formal evidence exists for cellular entry either from the vessel lumen or from the adventitial face [79]. In venous bypass grafts for example, EC denudation or

Conclusion

Allograft vasculopathy (AV) is an intimal proliferative lesion; the resulting concentric hyperplasia leads to a diffuse vascular stenosis that remains the most important long-term limit on the success of solid-organ transplantation. The primary inciting insult is an alloresponse, although ischemia and other forms of injury are involved. Both innate and adaptive immunity contribute, and a potentially autonomous IFNγ axis is a central driving component. Host SMC precursors of various sources

Final comments

It is a distinct pleasure and honor to be asked to write a brief reflection on the mentorship and guidance that I received—lo these years gone by—working with Abul Abbas in this special issue of the Journal of Autoimmunity. This issue is devoted entirely to recognizing the contributions of Abul Abbas and part of the Journal's efforts to recognize unique topics, themes and particularly outstanding autoimmunologists [114], [115], [116], [117].

I was a post-doc in Abul's lab almost a quarter

References (117)

  • S. Jiang et al.

    New spectrum of allorecognition pathways: implications for graft rejection and transplantation tolerance

    Curr Opin Immunol

    (2004)
  • O. Raisky et al.

    CD8(+) T cells induce graft vascular occlusion in a CD40 knockout donor/recipient combination

    J Heart Lung Transplant

    (2003)
  • Y. Furukawa et al.

    Association of B7-1 co-stimulation with the development of graft arterial disease: studies using mice lacking B7-1, B7-2, or B7-1/B7-2

    Am J Pathol

    (2000)
  • K. Kim et al.

    CD28-B7-mediated T cell costimulation in chronic cardiac allograft rejection: differential role of B7-1 in initiation versus progression of graft arteriosclerosis

    Am J Pathol

    (2001)
  • P. Terasaki et al.

    Predicting kidney graft failure by HLA antibodies: a prospective trial

    Am J Transplant

    (2004)
  • R. Frank et al.

    Correlation of circulating donor-specific anti-HLA antibodies and presence of C4d in endomyocardial biopsy with heart allograft outcomes: a single-center, retrospective study

    J Heart Lung Transpl

    (2013)
  • A. Zlotnik et al.

    The chemokine superfamily revisited

    Immunity

    (2012)
  • J. Suzuki et al.

    Tumor necrosis factor receptor -1 and -2 double deficiency reduces graft arterial disease in murine cardiac allografts

    Am J Transplant

    (2003)
  • J. Andersson et al.

    Adaptive immunity and atherosclerosis

    Clin Immunol

    (2010)
  • J. Mclaren et al.

    Interferon gamma: a master regulator of atherosclerosis

    Cytokine Growth Factor Rev

    (2009)
  • P. Kleinbongard et al.

    TNFalpha in atherosclerosis, myocardial ischemia/reperfusion and heart failure

    Pharmacol Ther

    (2010)
  • C. Wong et al.

    Role of vascular remodeling in the pathogenesis of early transplant coronary artery disease: a multicenter prospective intravascular ultrasound study

    J Heart Lung Transpl

    (2001)
  • U. Schonbeck et al.

    T(H)2 predominant immune responses prevail in human abdominal aortic aneurysm

    Am J Pathol

    (2002)
  • R. Shah et al.

    The role of stem cells in the response to myocardial and vascular wall injury

    Cardiovasc Pathol

    (2005)
  • J. George et al.

    Of mice and men: mouse models and the molecular mechanisms of post-transplant coronary artery disease

    J Heart Lung Transplant

    (2005)
  • S. Abele et al.

    Clopidogrel reduces the development of transplant arteriosclerosis

    J Thorac Cardiovasc Surg

    (2006)
  • P. Nelson et al.

    Chemokines, chemokine receptors, and allograft rejection

    Immunity

    (2001)
  • A. Schecter et al.

    Human smooth muscle cells possess functional CCR5

    J Biol Chem

    (2000)
  • A. Dhaliwal et al.

    Cardiac allograft vasculopathy: the Achilles' heel of long-term survival after cardiac transplantation

    Curr Atheroscler Rep

    (2006)
  • H. Nagano et al.

    Coronary arteriosclerosis after T-cell-mediated injury in transplanted mouse hearts. Role of interferon-g

    Am J Pathol

    (1998)
  • D. Pattanaik et al.

    Vascular involvement in systemic sclerosis (scleroderma)

    J Inflamm Res

    (2011)
  • D. Prevost et al.

    Coronary vessel involvement by chronic graft-versus-host disease presenting as sudden cardiac death

    Bone Marrow Transplant

    (2004)
  • R. Mitchell

    Graft vascular disease: immune response meets the vessel wall

    Annu Rev Pathol

    (2009)
  • M. Rahmani et al.

    Allograft vasculopathy versus atherosclerosis

    Circ Res

    (2006)
  • R. Mitchell et al.

    Vascular remodeling in transplant vasculopathy

    Circ Res

    (2007)
  • R. Salomon et al.

    Human coronary transplantation-associated arteriosclerosis: evidence for a chronic immune reaction to activated graft endothelial cells

    Am J Pathol

    (1991)
  • G. Hansson et al.

    The immune system in atherosclerosis

    Nat Immunol

    (2011)
  • G. Tellides et al.

    Interferon-gamma axis in graft arteriosclerosis

    Circ Res

    (2007)
  • D. Ramzy et al.

    Cardiac allograft vasculopathy: a review

    Can J Surg

    (2005)
  • J. Thurman et al.

    The central role of the alternative complement pathway in human disease

    J Immunol

    (2006)
  • J. Wehner et al.

    Antibody and complement in transplant vasculopathy

    Circ Res

    (2007)
  • K. Koh et al.

    T cell-mediated vascular dysfunction of human allografts results from IFN-gamma dysregulation of NO synthase

    J Clin Invest

    (2004)
  • A. Ardehali et al.

    Indirect alloreactivity and chronic rejection

    Transplantation

    (2002)
  • P. Hornick et al.

    Significant frequencies of T cells with indirect anti-donor specificity in heart graft recipients with chronic rejection

    Circulation

    (2000)
  • K. Shimizu et al.

    Host CD40 ligand deficiency induces long-term allograft survival and donor-specific tolerance in mouse cardiac transplantation but does not prevent graft arteriosclerosis

    J Immunol

    (2000)
  • T. Glysing-Jensen et al.

    Chronic blockade of CD28-B7-mediated T-cell costimulation by CTLA4Ig reduces intimal thickening in MHC class I and II incompatible mouse heart allografts

    Transplantation

    (1997)
  • H. Kosuge et al.

    Critical role of inducible costimulator signaling in the development of arteriosclerosis

    Arterioscl Thromb Vasc Biol

    (2006)
  • J. Yang et al.

    Critical role of donor tissue expression of programmed death ligand-1 in regulating cardiac allograft rejection and vasculopathy

    Circulation

    (2008)
  • P. Russell et al.

    Coronary atherosclerosis in transplanted mouse hearts. II. Importance of humoral immunity

    J Immunol

    (1994)
  • W. Hancock et al.

    Antibody-induced transplant arteriosclerosis is prevented by graft expression of anti-oxidant and anti-apoptotic genes

    Nat Med

    (1998)
  • Cited by (13)

    • Gene expression profile in cardiovascular disease and preeclampsia: A meta-analysis of the transcriptome based on raw data from human studies deposited in Gene Expression Omnibus

      2015, Placenta
      Citation Excerpt :

      Angiogenesis, TGF-β-signaling, graft-versus-host disease and allograft rejection were also found to be overlapping pathways in both conditions. These pathways together with immune factors are proposed to play a role in the pathogenesis of PE and atherosclerosis, leading to CVD [29,30]. Myc proto-oncogene was one of the other genes that was differentially expressed both in CVD and PE.

    • Activation of benign autoimmunity as both tumor and autoimmune disease immunotherapy: A comprehensive review

      2014, Journal of Autoimmunity
      Citation Excerpt :

      This issue of the Journal of Autoimmunity is dedicated to the honor of Michael Sela and Ruth Arnon. The Journal of Autoimmunity has honored distinguished immunologists as well as published dedicated themes on subjects of critical importance, including, for example, recognition of Abul Abbas, Noel Rose and Ian Mackay, and themes focused on the liver as a victim of autoimmunity [51–54]. All immunologists and many hundreds of thousands of patients owe thanks and approbation to Michael and Ruth for their pioneering work on the chemical basis of antigenicity and immunogenicity (the latter term coined by Michael Sela) and for their fundamental discoveries, together with the late Dvora Teitelbaum, leading to their development of an important therapy for multiple sclerosis – glatiramer acetate (Copaxone) [55].

    • Engineering of synthetic cellular microenvironments: Implications for immunity

      2014, Journal of Autoimmunity
      Citation Excerpt :

      Finally, it is with gratitude and appreciation that we provide this contribution to a special issue of the Journal of Autoimmunity that honors the distinguished careers of Ruth Arnon and Michael Sela. This special issue is part of a series that honors distinguished immunologists which have included such figures as Abul Abbas, Pierre Youinou, Noel Rose, and Ian Mackay and attempts to highlight subjects that are uniquely important to immunologists [132–134]. It is particularly noteworthy that the work described herein is a direct reflection of the classic paper in 1960 by Michael and Ruth on studies of the chemical basis of the antigenicity of proteins [1].

    View all citing articles on Scopus
    View full text