Vous êtes sur la page 1sur 7

European Heart Journal Advance Access published December 8, 2015

European Heart Journal REVIEW


doi:10.1093/eurheartj/ehv639

Basic science for the clinician

Role of T-cells in myocardial infarction


Ulrich Hofmann 1,2 and Stefan Frantz 1*
1
Klinik und Poliklinik für Innere Medizin III, Universitätsklinikum der Martin-Luther-Universität Halle-Wittenberg, Ernst-Grube-Straße 40, Halle (Saale) 06120, Germany; and
2
Deutsches Zentrum für Herzinsuffizienz, Universitätsklinikum Würzburg, Straubmühlweg 2a, Würzburg 97078, Germany

Received 6 July 2015; revised 4 November 2015; accepted 4 November 2015

Innate immunity has been studied for several decades in the context of ischaemia-reperfusion injury, myocardial remodelling, and healing. In the

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


last years, a number of experimental and clinical studies focused on adaptive immunity in these processes. Meanwhile, there is considerable
evidence especially on the role of CD4+ T-cells in myocardial injury and healing, whereas their role in remodelling is less clear. Innate leukocytes
are able to recognize a wide array of self and foreign molecular patterns, whereas the activation of adaptive immunity requires the highly specific
cooperation of antigen-presenting cells and distinct antigen-specific receptors on lymphocytes. Relevant autoantigens have not yet been def-
initely identified but experimental evidence indicates that autoantigen recognition is necessary for T-cell activation after myocardial infarction.
Non-antigen-specific modes of activation might also play a role especially during acute ischaemia and reperfusion of the myocardium. This
review summarizes the current evidence from experimental studies and presents side-by-side recent clinical data on the role of T cells in
the pathophysiology of myocardial reperfusion injury and post myocardial infarction healing.
-----------------------------------------------------------------------------------------------------------------------------------------------------------
Keywords Innate immunity † Adaptive immunity † Myocardial infarction † Atherosclerosis † Lymphocyte

and the clinical significance regarding potential therapeutic implica-


Introduction tions will be discussed.
Atherosclerosis constitutes the pathophysiological basis for disab-
ling diseases like stroke and myocardial infarction, both constituting
the leading causes for morbidity and mortality in the Western Basic principles of T-cell activation
World.1 Myocardial infarction is one of the most detrimental ath- For the better understanding of the data summarized below, we first
erosclerosis related complications leading to considerable mortality would like to briefly envision some basic principles of T-cell gener-
and morbidity due to heart failure. Despite the encouraging effects ation, migration, and age-related changes in T-cell function. The cen-
of the current state-of-the-art primary and secondary prevention tral organ for the generation of T-cells is the thymus. The current
measures, there remains a substantial residual risk for ischaemic nomenclature of T-cell subsets is based on several markers (Table 1)
complications in individuals with sub-clinic or manifest coronary which are mostly analysed by fluorescence-activated cell sorting
heart disease.2 Furthermore, it is now widely accepted that despite (FACS). The thymic release of naı̈ve CD4+ and CD8+ T-cells
prompt reperfusion, which is today achieved in the majority of our declines with age.4 The process of thymus involution starts in
patients, the myocardium is subject to additional injury induced by human childhood which is much earlier than observed in mice.5
re-establishment of blood flow.3 The role of innate immunity has This is associated with an age-dependent decline in the T-cell recep-
been a subject for experimental research in the field of atheroscler- tor diversity and the predominance of highly differentiated periph-
osis and myocardial ischaemia-reperfusion injury for decades eral memory T-cells within an ‘aged’ T-cell compartment. The
whereas T-cells have only recently come into focus. age-related changes in the memory T-cell repertoire underlies a
Therefore, the present review summarizes established knowl- strong modulation by chronic virus infection especially cytomegaly
edge regarding the role of T-cells in the pathogenesis of myocardial virus (CMV). Cytomegaly virus infection, having an age-dependent
ischaemia-reperfusion injury and post myocardial infarction healing. prevalence of 70% in Western world, induces an expansion of
A special focus will be on the juxtaposition of mechanistic concepts the so-called effector-memory CD4+ and CD8+ compartment
mainly derived from rodent studies and the relating recent data from coming along with a shrinkage of the T-cell receptor diversity.5
human specimens. Current knowledge gaps, inherent problems in The memory T-cell subsets in elderly are poised for proinflamma-
the transfer from small animal data to human pathophysiology, tory cells which is aggravated by both CMV infection and traditional

* Corresponding author. Tel: +49 345 557 2601, Fax: +49 345 557 2072, Email: stefan.frantz@uk-halle.de
Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2015. For permissions please email: journals.permissions@oup.com.
Page 2 of 7 U. Hofmann and S. Frantz

priming is believed to mainly influence the pattern of their periph-


Table 1 Key markers for immuno-phenotyping of eral homing.7 For the myocardium, hepatocyte growth factor signal-
T-cells referred in the text ling via its receptor c-Met was recently shown to instruct primed
CD Cluster of differentiation nomenclature denotes surface T-cells in heart draining lymph nodes to express a pattern of chemo-
markers for the immune-phenotyping of cells kine receptors, including CCR4 and CXCR3, which allow their spe-
CD4 CD4 antigen mainly identifies T-helper cells cific homing to the myocardium.8 After antigen recognition, clonally
CD8 CD8 antigen mainly identifies cytotoxic T-cells expanding T-cells develop into tissue-homing effector cells and dif-
CD25 Subunit of the interleukin-2 receptor, expressed on ferent memory T-cell subsets which are defined by their recircula-
activated T-cells and CD4+ T-regulatory T-cells tion characteristics. Besides recirculating memory T-cell subsets
Foxp3 Forkhead-Box-Protein 3 transcription factor serves tissue resident memory cells are becoming more and more recog-
as marker for CD4+ T-regulatory cells in mice nized in different organs including the myocardium9 which cannot
CD39 Ectonucleoside triphosphate diphosphohydrolase, be monitored in peripheral blood. Their biological significance in
membrane bound enzyme hydrolyzes ATP and
myocardial homeostasis and disease is still enigmatic.
ADP to AMP
The very brief summary of the biological principles of T-cell acti-
CD73 ecto-5′ -nucleotidase, membrane bound enzyme converts
AMP to adenosine vation and migration should illustrate that analysis of the peripheral
blood alone will allow studying certain subsets of recirculating
T-cells which, at best, contain only a very small portion of disease-

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


specific cells (Figure 1). This poses a major limitation in the interpret-
ation of data from a considerable number of studies analysing T-cells
cardiovascular risk factors. These processes are only poorly re-
flected in rodent models. in peripheral blood. However, it is obvious that tissue specimen
For adaptive immune responses, naı̈ve T-cells have to recognize from the myocardium or its draining lymph nodes which would pro-
vide much more specific insights are not widely available.
their antigens in the context of major histocompatibility complex
surface molecules. This process takes place in secondary lymphatic
organs, mainly in the draining lymph nodes and the spleen. Besides
antigen recognition, additional stimuli are required for activation of
Clinical evidence for the
T-cells (‘costimulatory signals’). T-cell activation by contact with its involvement of T-cells in acute
specific antigen is called T-cell priming. Primed CD4+ and CD8+ coronary syndromes
T-cells can then both exert effector/proinflammatory and regula-
tory/anti-inflammatory function depending on their differentiation There is meanwhile a considerable number of studies that have ana-
profile which is defined by the signals the T-cell receives from lysed T-cells in peripheral blood in patients with unstable angina or
antigen-presenting cells and the environment in lymphatic organs. acute coronary syndromes. These data might partly reflect activa-
So-called natural CD4+ regulatory T-cells constitute a particular tion of T-cells secondary to coronary atherothrombosis and pre-
anti-inflammatory immune-regulatory T-cell subset which is gener- ceding coronary artery wall inflammation. However, they might
ated in the thymus and is highly enriched for T-cells with autoantigen also represent system-wide changes in the T-cell activation status
specificity. and their migratory behaviour in response acute myocardial injury
and failure. Activation of the renin – angiotensin – aldosterone or
the sympathetic nervous system is also likely to influence the com-
Principles of T-cell migration position of the circulating T-cell subsets analysed in the periphery.
In adults, memory T-cells predominate the peripheral T-cell com- Hence, specific peripheral T-cell patterns observed in patients
partment which upon antigen recognition are poised for rapid pro- with acute coronary events may not necessarily be informative on
liferation and effector function execution. T-cells enter lymph nodes the role of particular T-cell subsets in the pathophysiology of ather-
either from the afferent lymphatics or from the blood and exit othrombosis or myocardial injury. For space restrictions, we would
lymph nodes via the efferent lymph which is mainly collected in like to refer the reader to a very recent comprehensive review on
the thoracic duct where it is drained to the blood. Naı̈ve T-cells re- clinical studies describing T-cell subsets in patients with atheroscler-
circulate between blood and lymph nodes. Also different subsets of osis and acute coronary syndromes.10
the memory T-cell pool recirculate through the body in steady state.
Antigen-presenting cells collect antigens in peripheral tissues and
lymph nodes to present them as processed peptides to T-cells. Role of T-cells in
T-cell activation by recognition of antigens leads to the transient re- ischaemia-reperfusion injury
tention of T-cells in secondary lymphoid organs. However, the ac-
tivation and clonal expansion of antigen experienced T-cells is of the myocardium
coupled with migration cues enabling activated T-cells to leave sec-
ondary lymphoid organs and to become recruited to sides of inflam- Experimental evidence from mouse
mation.6 Both transient T-cell retention in secondary lymphatic studies
organs for efficient antigen recognition and activation and recruit- First experimental evidence that CD4+ T-cells contribute to myo-
ment to peripheral non-lymphatic organs is strongly regulated by cardial ischaemia-reperfusion injury came from a study analysing
chemokine receptor signalling. Hereby, the location of T-cell wild-type and lymphocyte-deficient RAG1 KO mice. RAG1 KO
Role of T cells in myocardial infarction Page 3 of 7

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


Figure 1 T-cells migration and recirculation pathways. The scheme depicts where particular T-cells subsets get accessible for analysis in experi-
mental and clinical studies.

mice revealed significantly smaller infarct sizes compared with injurious action of CD4+ T-cells can be mitigated by endogenous
control mice. Furthermore, CD4+ T-cell-depleted mice, but not adenosine signalling.
CD8+-depleted mice showed a significantly decreased infarct size Corresponding to the above-mentioned clinical data,16 CD4+
compared with control mice. Reconstitution of RAG1 KO mice T-regulatory cells were recently reported to rapidly accumulate
by adoptive transfer with CD4+ T cells reversed this protection in murine hearts following ischaemia-reperfusion. The adoptive
seen in RAG1 KO mice. RAG1 KO mice reconstituted with transfer of in vitro activated T-regulatory cells was able to decrease
CD4+ T-cells from IFN-g KO mice did not increase myocardial myocardial injury, whereas transfer of non-activated T-cells had
infarct size, indicating that CD4 + T-cells promote ischaemia- no effect. Mechanistically, the protective effect of activated
reperfusion injury by IFN-g expression.11 T-regulatory cells was related to ectonucleoidase (CD39) ex-
The involvement of T-cells in ischaemia-reperfusion injury has pression.17 From these data, one can speculate that recruitment
been studied in animal reperfusion injury models in several organs of (in vitro) activated T-regulatory cells adds to the local
but there are few data explaining how CD4+ T-cells become acti- CD39-mediated adenosine formation which counteracts the dele-
vated under these conditions.12 The timeframe in which myocardial terious effects of other leukocytes subsets, including conventional
injury occurs in the presence of CD4+ T-cells would rather indicate T-cells in the context of myocardial ischaemia-reperfusion.
a non-T-cell receptor dependent way of T-cell activation, e.g. by rec-
ognition of ‘alarmins’13 from injured cell which are recognized by
pattern recognition receptors, such as toll-like receptors.14 Clinical evidence for the involvement of
T-cells in myocardial reperfusion injury
There are only few studies assessing the kinetics of blood T-cell sub-
Role of adenosine signalling and sets during early reperfusion. A novel approach using 13-parameter
T-regulatory cells for protection against FACS immuno-phenotyping followed by hierarchical cluster analysis
reperfusion injury was applied to identify novel subsets of peripheral T-cells in patients
In the reperfused myocardium, the ectoenzymes CD39 and CD73 with acute STEMI.18 This study reported a specific loss of CD4+
are expressed by a broad range of leukocytes and cardiomyocytes.15 chemokine receptor CCR7+ T-cells during early reperfusion. These
They promote the extracellular degradation of nucleotides released subsets include naı̈ve, central memory, and T-regulatory cells. At the
by cellular injury to anti-inflammatory adenosine. Treatment with an moment, one can definitely not clarify whether this observation re-
adenosine (A2A) receptor agonist blocked the increase in infarct flects a CCR7-mediated redistribution of this T-cell subset to per-
size in Rag1 KO mice reconstituted with wild type but not A2A re- ipheral (lymphatic) organs or a specific accumulation of these
ceptor deficient CD4+ T cells.11 This observation indicates that the subsets in the reperfused microvascular bed of the myocardium.
Page 4 of 7 U. Hofmann and S. Frantz

However, for the interpretation of the data, it is important to men- from a study demonstrating that adoptive transfer of splenocytes
tion that CCL19 and CCL21 are the ligands for CCR7. Both chemo- from rats after acute myocardial infarction produced myocarditis
kines are reported to rise in plasma following coronary in naı̈ve recipient rats.23 Inspired by this study, we recently demon-
interventions.19 Thus, it is conceivable that these CCR7 ligands strated in a mouse MI model that CD4+ T-cells get activated and
could play a role for the recruitment of T-cells to the reperfusion proliferate in heart draining lymph nodes.24 The activation process
myocardium. The authors further concentrated their in-depth ana- takes place within days and requires an intact T-cell receptor reper-
lysis on a subset of CD57+ T-cells likely resembling memory T-cells toire. Absence of CD4+ T-cells in CD4 deficient and MHC class II
which have recently undergone proliferation. Unfortunately, deficient mice was associated with worse outcome. A similar
T-regulatory cells which have been described to specifically phenotype was found in a mouse model harbouring a transgenic
accumulate in coronary thrombi16 were not further analysed in T-cell receptor for an in this context irrelevant ovalbumin-derived
this study. peptide (OT-II mouse) which showed no significant T-cell activation
T-cell retention within the myocardium is reflected by trans- in mediastinal lymph nodes in response to MI. The most likely inter-
coronary gradients. By such an experimental approach, a possible pretation of the data is that the presence of CD4+ T-cells that can
mechanistic hint for the injurious role of effector-memory T-cell re- be activated by autoantigens presented by MHC class II molecules is
tention in acutely reperfused myocardium was provided: the extent a prerequisite for proper wound healing. The pivotal role of T-cells
of T-cell retention in the coronary circulation correlated with activated by interaction with antigen-presenting cells was indirectly
microvascular obstruction as determined by cardiac magnet re- proven in another experimental study where CD11c+ cells, mainly

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


sonance tomography imaging.20 One of the chemokines potentially representing antigen-presenting cells, were ablated in a transgenic
mediating retention of T-cells in the reperfused microvascular mouse model.25 After MI there was a strikingly similar phenotype
bed might be fractalkine. Its receptor (CX3CR1) expression after as found in CD4+ T-cell deficient mouse models. Absence of
reperfusion was especially regulated in the subset of CCR72 effect- CD11c+ cells resulted in deteriorated left ventricular function and
or T-cells.20 remodelling after MI. However, the effect of CD11c + antigen-
Recently, the same group reported data on the CD8+ T-cell com- presenting cells on the activation of CD4+ T-cells was not further
partment in patients with reperfused MI. They found a particular explored in this study.
early and long-lasting fall in the peripheral frequency of terminally A major open question in the field is whether CD4+ T-cells re-
differentiated effector-memory CD8+ T-cells in CMV-positive pa- spond to tissue-specific antigens or ‘universal’ tissue autoantigens.
tients.21 The CD8+ subset that was depleted from peripheral blood Mainly for methodological reasons it has not yet been experimental-
included CMV-specific T-cells. Their data further indicate that the ly addressed which antigens activate T-cells in experimental models
surface molecule PD-1 might be involved in the long-lasting loss of ischaemic tissue injury like MI.
of this subset which was specifically found in CMV-positive patients. Our experiments in mice showed that both conventional effector
Whether the putative preferential recruitment of this CD8+ T-cell CD4+ T-cells having mainly a Th1 cytokine profile and Foxp3+
subset to the reperfused myocardium impacts reperfusion injury T-regulatory cells infiltrate the myocardium within a few days after
and clinical outcome remains to be determined. Of note, a longitu- MI.24 However, the exact pathophysiological role of conventional
dinal cohort study found a significant association of coronary artery and regulatory T-cells in myocardial healing was initially not clear.
disease-related mortality and CMV status suggesting a potential Another group then reported in this context that T-regulatory
mechanistic link between the changes in the CD8+ T-cell compart- cell function is compromised in mice that underwent experimental
ment induced by CMV infection and the pathophysiology of acute infarction.26 Therefore, we concentrated our studies on the differ-
coronary events.22 ential effects of both CD4+ T-cell subsets.
Collectively, there is consistent evidence from mouse studies that Mechanistically, we found that CD4+ T-cells control the infiltra-
proinflammatory CD4+ T-cells contribute to ischemia-reperfusion tion and most likely the differentiation of recruited proinflammatory
injury whereas the role of CD4+ T-regulatory T-cells which were monocytes, characterized by high surface expression of the antigen
found to be enriched in coronary thrombi is less well established. Ly6C. The absence of CD4+ T cells increased the number of Ly6-
However, the available experimental data indicate that they might Chigh monocytes in the infarcted myocardium.24 Depletion of CD4+
constitute a therapeutic target to mitigate myocardial ischaemia- T-regulatory cells induced a preferential M1-like phenotype, where-
reperfusion injury. Data from clinical studies further indicate that as activation of T-regulatory cells induced a M2-like phenotype in
besides CD4+ T-cells, CD8+ memory T-cell subsets become myocardial macrophages. Stimulation of M2-like macrophage differ-
entrapped in the coronary microcirculation early after the onset entiation by activation of T-regulatory cells induced expression of
of reperfusion and/or might even actively infiltrate the reperfused arginase-1, Interleukin-13, osteopontin, and TGF-b in macro-
myocardium where they likely contribute to reperfusion. phages.27 As these proteins are all involved in the extracellular ma-
trix formation, the control of macrophage differentiation likely
constitutes the main mechanism how T-regulatory cells improve
Role of lymphocytes in non- myocardial healing. Accordingly, CD4+ T-cell deficient mice
reperfused myocardial infarction showed disturbed extracellular matrix formation in the scar and
died from myocardial rupture. Another experimental study in the
Experimental evidence on CD41 T-cells mouse MI model reported data fitting well to our concept that
First experimental evidence for the activation of T-cells with reactiv- T-regulatory cells may modulate the fibroblast phenotype and func-
ity to myocardial autoantigens in response to experimental MI came tion in the infarcted myocardium.28 Further adding to the concept of
Role of T cells in myocardial infarction Page 5 of 7

a pivotal role of regulatory T-cell in myocardial post infarct healing, it However, at present there are no clinically applicable non-invasive
was recently reported that expansion of T-regulatory cells in vivo by imaging techniques to monitor T-cell migration. Histopathological
means of either adoptive transfer of Tregs or a CD28-superagonistic studies reported T-cell infiltrates in both remote and peri-infarction
antibody attenuated myocardial proinflammatory cytokine expres- myocardial regions and activated T cells within the coronary artery
sion and immune cell infiltration in a rat MI model.29 Accordingly an- wall of both the infarct- and non-infarct-related arteries in necropsy
other group reported that adoptive transfer of Treg cells in a mouse specimen from patients suffering from MI.34 This gives a valuable hint
permanent MI model attenuates both the post-infarction inflamma- that, in analogy to what was observed in mouse studies, myocardial
tory response and adverse remodeling.30 Collectively, endogenous injury in humans activates T-cells which home to the injured myo-
T-regulatory cells emerged as pivotal cellular players regulating re- cardium and likely modulate local inflammatory activity. Only one
pair of the myocardium. The precise mechanism and location of clinical study comparatively analysed thoracic lymph nodes from pa-
their interaction with myeloid progenitors, monocytes, and macro- tients with acute coronary syndromes and control individuals. How-
phages represent major gaps in our understanding of these inter- ever, the authors could not find differences in the cellular
action processes.31 A schematic summary on the local action of composition of lymph nodes between the two cohorts.35 Thus,
CD4+ T-cells in infarcted myocardium is presented in Figure 2. more data on T-cell activation and differentiation parameters
from human tissue specimen would be highly valuable to decipher
Experimental evidence on CD81 T-cells the role of T-cells post myocardial infarction.
Concerning the role of CD8+ T-cells, a subset of angiotensin 2 recep- Collectively, there is substantial experimental evidence showing
that CD4+ T-cells, especially T-regulatory cells promote myocardial

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


tor positive CD8+ T-cells was described in the peri-infarct zone of
rats 7 days after MI.32 This subset of CD8+ T-cells differed from clas- healing by interaction with macrophages and probably fibroblasts.
sical cytotoxic angiotensin 2 receptor negative CD8+ T-cells in their Mostly due to methodological limitations, there are no clinical data
capacity to secret IL-10 in response to angiotensin 2 stimulation to support this beneficial role of CD4+ T-cells which is obviously con-
in vitro. Their functional significance in myocardial healing was demon- trary to their immediate detrimental action during early reperfusion.
strated by transfer of these cells harvested from a donor after MI. This
procedure significantly reduced infarct size after experimental MI.
Correspondingly, the beneficial action of angiotensin 2 receptor ex- Conclusion
pressing CD4+ T-cells which includes a subset of Foxp3+ T-cells was The available experimental data from animal models provide us a
recently reported.33 Of note, animals with genetic CD8 deficiency relative clear picture regarding the role of CD4+ T-cells: In the
showed no significant clinical phenotype after experimental MI com- mouse ischaemia-reperfusion model CD4+ T-cells aggravate
pared with WT animals (own unpublished data). ischaemia-reperfusion injury. Subsequently, the activation of
CD4+ T-cells, most likely by recognition of autoantigens, is required
Evidence from human studies for proper healing. In this context, especially the subset of Foxp3+
Histopathological analysis of myocardial specimen and non-invasive CD4+ regulatory T-cells tunes the differentiation of macrophages
imaging modalities would be highly valuable to judge the relevance towards a pro-healing phenotype preventing from left ventricular
of the reported changes in the T-cell compartment in peripheral dilation and rupture. However, the role of newly primed effector
blood for myocardial injury and subsequent healing processes. T-cells, recirculating, and tissue resident memory T-cells has not

Figure 2 Role of conventional (Tconv) and regulatory (Treg) CD4+ T-cells and their interaction with other cell types in experimental models of
reperfusion injury and myocardial healing.
Page 6 of 7 U. Hofmann and S. Frantz

Figure 3 Mechanism of activation and recruitment of T-cells to the post-ischaemic myocardium. Mechanism of immediate T-cell retention in

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


reperfused myocardium, chemokines/chemokine receptors determining homing of antigen-specific activated T cells, and factors controlling the
local retention in myocardium are largely unknown but might all constitute putative therapeutic targets.

yet been addressed in this context. Also, the role of T-cells in later Funding
stages of remodelling is not clear. For identifying therapeutic ap- The manuscript was supported by grants of the Deutsche Forschungsge-
proaches targeting T-cells to be further evaluated towards clinical meinschaft (DFG SFB688 to U.H. and S.F.) and by the Bundesministerium
application, we need to identify relevant antigens inducing T-cell ac- für Bildung und Forschung (BMBF01 EO1004 to S.F.).
tivation and to understand which signals induce their recruitment to
and retention in the infarcted myocardium (Figure 3). Conflict of interest: none declared.
However, regarding the particular differences in the T-cell com-
partment of humans and laboratory rodents we have to be cautious References
1. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, Blaha MJ, Dai S, Ford ES,
in translating the findings from experimental studies to the clinical Fox CS, Franco S, Fullerton HJ, Gillespie C, Hailpern SM, Heit JA, Howard VJ,
scenario. The immune system of our patients shows phenomena Huffman MD, Judd SE, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH,
subsumed under the term ‘immune-senescence’ that are not pre- Lisabeth LD, Mackey RH, Magid DJ, Marcus GM, Marelli A, Matchar DB,
McGuire DK, Mohler ER 3rd, Moy CS, Mussolino ME, Neumar RW, Nichol G,
sent in our laboratory animals. Further, subsets like CD4+ CD28null Pandey DK, Paynter NP, Reeves MJ, Sorlie PD, Stein J, Towfighi A, Turan TN,
T-cells which emerge in patients with chronic low-grade inflamma- Virani SS, Wong ND, Woo D, Turner MB, American Heart Association Statistics
tion, e.g. associated with atherosclerosis36 can exert proinflammatory C, Stroke Statistics S. Heart disease and stroke statistics – 2014 update: a report
from the American Heart Association. Circulation 2014;129:e28 –e292.
function without T-cell receptor ligation. These unconventional 2. Graham I, Atar D, Borch-Johnsen K, Boysen G, Burell G, Cifkova R, Dallongeville J,
T-cells are widely missing in our laboratory mice but might especially De Backer G, Ebrahim S, Gjelsvik B, Herrmann-Lingen C, Hoes A, Humphries S,
contribute to atherothrombosis and reperfusion injury in humans. Knapton M, Perk J, Priori SG, Pyorala K, Reiner Z, Ruilope L, Sans-Menendez S,
Scholte op Reimer W, Weissberg P, Wood D, Yarnell J, Zamorano JL, Walma E,
Thus, we further need a deeper understanding how, when, and Fitzgerald T, Cooney MT, Dudina A, European Society of Cardiology Committee
which T-cell subsets are recruited to the human coronary microvas- for Practice G. European guidelines on cardiovascular disease prevention in clinical
culature and subsequently to the reperfused myocardium. Also, we practice: executive summary: Fourth Joint Task Force of the European Society of
Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical
should not only focus on the ischemic myocardium but also on Practice (constituted by representatives of nine societies and by invited experts).
draining lymph nodes and other lymphatic organs including the Eur Heart J 2007;28:2375 –2414.
spleen and bone marrow where interactions with other cellular 3. Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med 2007;357:
1121 –1135.
components of immunity and haematopoiesis might take place in 4. Boehm T, Swann JB. Thymus involution and regeneration: two sides of the same
our patients. Therefore, a joint effort of cardiologists, immunolo- coin? Nat Rev Immunol 2013;13:831 – 838.
gists, and imaging specialists is warranted for designing new ap- 5. Nikolich-Zugich J. Aging of the T cell compartment in mice and humans: from no
naive expectations to foggy memories. J Immunol 2014;193:2622 –2629.
proaches to be tested in translational and clinical studies. They 6. von Andrian UH, Mackay CR. T-cell function and migration. Two sides of the same
should enable us to get deeper insights in the implications of coin. N Engl J Med 2000;343:1020 –1034.
T-cell activation in the clinical context and to validate new thera- 7. Masopust D, Schenkel JM. The integration of T cell migration, differentiation and
function. Nat Rev Immunol 2013;13:309 –320.
peutic approaches aiming at the modulation of T-cell immunity. 8. Komarowska I, Coe D, Wang G, Haas R, Mauro C, Kishore M, Cooper D,
Nadkarni S, Fu H, Steinbruchel DA, Pitzalis C, Anderson G, Bucy P, Lombardi G,
Breckenridge R, Marelli-Berg FM. Hepatocyte growth factor receptor c-Met in-
Authors’ contributions structs T cell cardiotropism and promotes T cell migration to the heart via auto-
crine chemokine release. Immunity 2015;42:1087 – 1099.
S.F. handled funding and supervision; S.F., U.H. drafted the manu- 9. Casey KA, Fraser KA, Schenkel JM, Moran A, Abt MC, Beura LK, Lucas PJ, Artis D,
Wherry EJ, Hogquist K, Vezys V, Masopust D. Antigen-independent differentiation
script; S.F., U.H. made critical revision of the manuscript for key and maintenance of effector-like resident memory T cells in tissues. J Immunol 2012;
intellectual content. 188:4866 –4875.
Role of T cells in myocardial infarction Page 7 of 7

10. Ammirati E, Moroni F, Magnoni M, Camici PG. The role of T and B cells in human 24. Hofmann U, Beyersdorf N, Weirather J, Podolskaya A, Bauersachs J, Ertl G,
atherosclerosis and atherothrombosis. Clin Exp Immunol 2015;179:173 –187. Kerkau T, Frantz S. Activation of CD4+ T lymphocytes improves wound healing
11. Yang Z, Day YJ, Toufektsian MC, Xu Y, Ramos SI, Marshall MA, French BA, Linden J. and survival after experimental myocardial infarction in mice. Circulation 2012;125:
Myocardial infarct-sparing effect of adenosine A2A receptor activation is due to its 1652– 1663.
action on CD4+ T lymphocytes. Circulation 2006;114:2056 – 2064. 25. Anzai A, Anzai T, Nagai S, Maekawa Y, Naito K, Kaneko H, Sugano Y, Takahashi T,
12. Linfert D, Chowdhry T, Rabb H. Lymphocytes and ischemia-reperfusion injury. Abe H, Mochizuki S, Sano M, Yoshikawa T, Okada Y, Koyasu S, Ogawa S, Fukuda K.
Transplant Rev (Orlando) 2009;23:1 –10. Regulatory role of dendritic cells in postinfarction healing and left ventricular re-
13. Matzinger P. The danger model: a renewed sense of self. Science 2002;296: modeling. Circulation 2012;125:1234 –1245.
301 –305. 26. Sharir R, Semo J, Shimoni S, Ben-Mordechai T, Landa-Rouben N, Maysel-
14. Reynolds JM, Dong C. Toll-like receptor regulation of effector T lymphocyte func- Auslender S, Shaish A, Entin-Meer M, Keren G, George J. Experimental myocardial
tion. Trends Immunol 2013;34:511–519. infarction induces altered regulatory T cell hemostasis, and adoptive transfer at-
15. Bonner F, Borg N, Burghoff S, Schrader J. Resident cardiac immune cells and ex- tenuates subsequent remodeling. PLoS ONE 2014;9:e113653.
pression of the ectonucleotidase enzymes CD39 and CD73 after ischemic injury. 27. Weirather J, Hofmann UD, Beyersdorf N, Ramos GC, Vogel B, Frey A, Ertl G,
PLoS ONE 2012;7:e34730. Kerkau T, Frantz S. Foxp3+ CD4+ T cells improve healing after myocardial infarc-
16. Klingenberg R, Brokopp CE, Grives A, Courtier A, Jaguszewski M, Pasqual N, Vlas- tion by modulating monocyte/macrophage differentiation. Circ Res 2014;115:
kou Badra E, Lewandowski A, Gaemperli O, Hoerstrup SP, Maier W, 55 –67.
Landmesser U, Luscher TF, Matter CM. Clonal restriction and predominance of 28. Saxena A, Dobaczewski M, Rai V, Haque Z, Chen W, Li N, Frangogiannis NG. Regu-
regulatory T cells in coronary thrombi of patients with acute coronary syndromes. latory T cells are recruited in the infarcted mouse myocardium and may modulate
Eur Heart J 2015;36:1041 –1048. fibroblast phenotype and function. Am J Physiol Heart Circ Physiol 2014;307:
17. Xia N, Jiao J, Tang TT, Lv BJ, Lu YZ, Wang KJ, Zhu ZF, Mao XB, Nie SF, Wang Q, H1233 –H1242.
Tu X, Xiao H, Liao YH, Shi GP, Cheng X. Activated regulatory T-cells attenuate
29. Tang TT, Yuan J, Zhu ZF, Zhang WC, Xiao H, Xia N, Yan XX, Nie SF, Liu J, Zhou SF,
myocardial ischaemia/reperfusion injury through a CD39-dependent mechanism.
Li JJ, Yao R, Liao MY, Tu X, Liao YH, Cheng X. Regulatory T cells ameliorate cardiac
Clin Sci (Lond) 2015;128:679 –693.

Downloaded from http://eurheartj.oxfordjournals.org/ by guest on December 8, 2015


remodeling after myocardial infarction. Basic Res Cardiol 2012;107:232.
18. Hoffmann J, Fiser K, Weaver J, Dimmick I, Loeher M, Pircher H, Martin-Ruiz C,
30. Matsumoto K, Ogawa M, Suzuki J, Hirata Y, Nagai R, Isobe M. Regulatory T lympho-
Veerasamy M, Keavney B, von Zglinicki T, Spyridopoulos I. High-throughput
cytes attenuate myocardial infarction-induced ventricular remodeling in mice. Int
13-parameter immunophenotyping identifies shifts in the circulating T-cell com-
Heart J 2011;52:382 –387.
partment following reperfusion in patients with acute myocardial infarction. PLoS
31. Nahrendorf M, Swirski FK. Regulating repair: regulatory T cells in myocardial infarc-
ONE 2012;7:e47155.
tion. Circ Res 2014;115:7 –9.
19. Damas JK, Smith C, Oie E, Fevang B, Halvorsen B, Waehre T, Boullier A, Breland U,
32. Curato C, Slavic S, Dong J, Skorska A, Altarche-Xifro W, Miteva K, Kaschina E,
Yndestad A, Ovchinnikova O, Robertson AK, Sandberg WJ, Kjekshus J, Tasken K,
Froland SS, Gullestad L, Hansson GK, Quehenberger O, Aukrust P. Enhanced ex- Thiel A, Imboden H, Wang J, Steckelings U, Steinhoff G, Unger T, Li J. Identification
pression of the homeostatic chemokines CCL19 and CCL21 in clinical and experi- of noncytotoxic and IL-10-producing CD8+AT2R+ T cell population in response
mental atherosclerosis: possible pathogenic role in plaque destabilization. to ischemic heart injury. J Immunol 2010;185:6286 –6293.
Arterioscler Thromb Vasc Biol 2007;27:614 – 620. 33. Skorska A, von Haehling S, Ludwig M, Lux CA, Gaebel R, Kleiner G, Klopsch C,
20. Boag SE, Das R, Shmeleva EV, Bagnall A, Egred M, Howard N, Bennaceur K, Dong J, Curato C, Altarche-Xifro W, Slavic S, Unger T, Steinhoff G, Li J, David R.
Zaman A, Keavney B, Spyridopoulos I. T lymphocytes and fractalkine contribute The CD4 AT2R T cell subpopulation improves post-infarction remodelling and re-
to myocardial ischemia/reperfusion injury in patients. J Clin Invest 2015;125: stores cardiac function. J Cell Mol Med 2015;19:1975 –1985.
3063 –3076. 34. Abbate A, Bussani R, Liuzzo G, Biondi-Zoccai GG, Barresi E, Mellone P, Sinagra G,
21. Hoffmann J, Shmeleva EV, Boag SE, Fiser K, Bagnall A, Murali S, Dimmick I, Dobrina A, De Giorgio F, Sharma R, Bassan F, Severino A, Baldi F, Biasucci LM,
Pircher H, Martin-Ruiz C, Egred M, Keavney B, von Zglinicki T, Das R, Todryk S, Pandolfi F, Silvestri F, Vetrovec GW, Baldi A, Crea F. Sudden coronary death, fatal
Spyridopoulos I. Myocardial ischemia and reperfusion leads to transient CD8 im- acute myocardial infarction and widespread coronary and myocardial inflamma-
mune deficiency and accelerated immunosenescence in CMV-seropositive pa- tion. Heart 2008;94:737 –742.
tients. Circ Res 2015;116:87 –98. 35. Backteman K, Andersson C, Dahlin LG, Ernerudh J, Jonasson L. Lymphocyte
22. Savva GM, Pachnio A, Kaul B, Morgan K, Huppert FA, Brayne C, Moss PA, Medical Re- subpopulations in lymph nodes and peripheral blood: a comparison between
search Council Cognitive Function and Ageing Study. Cytomegalovirus infection is as- patients with stable angina and acute coronary syndrome. PLoS ONE 2012;7:
sociated with increased mortality in the older population. Aging Cell 2013;12:381–387. e32691.
23. Maisel A, Cesario D, Baird S, Rehman J, Haghighi P, Carter S. Experimental auto- 36. Liuzzo G, Goronzy JJ, Yang H, Kopecky SL, Holmes DR, Frye RL, Weyand CM.
immune myocarditis produced by adoptive transfer of splenocytes after myocardial Monoclonal T-cell proliferation and plaque instability in acute coronary syndromes.
infarction. Circ Res 1998;82:458 –463. Circulation 2000;101:2883 – 2888.

Vous aimerez peut-être aussi