Cytokinopathy with aberrant cytotoxic lymphocytes and pro-fibrotic myeloid response in SARS-CoV-2 mRNA vaccine-associated myocarditis

Rare immune-mediated cardiac tissue inflammation can occur after vaccination, including after SARS-CoV-2 mRNA vaccines. However, the underlying immune cellular and molecular mechanisms driving this pathology remain poorly understood. Here, we investigated a cohort of patients who developed myocarditis and/or pericarditis with elevated troponin, B-type natriuretic peptide, and C-reactive protein levels as well as cardiac imaging abnormalities shortly after SARS-CoV-2 mRNA vaccination. Contrary to early hypotheses, patients did not demonstrate features of hypersensitivity myocarditis, nor did they have exaggerated SARS-CoV-2–specific or neutralizing antibody responses consistent with a hyperimmune humoral mechanism. We additionally found no evidence of cardiac-targeted autoantibodies. Instead, unbiased systematic immune serum profiling revealed elevations in circulating interleukins (IL-1β, IL-1RA, and IL-15), chemokines (CCL4, CXCL1, and CXCL10), and matrix metalloproteases (MMP1, MMP8, MMP9, and TIMP1). Subsequent deep immune profiling using single-cell RNA and repertoire sequencing of peripheral blood mononuclear cells during acute disease revealed expansion of activated CXCR3+ cytotoxic T cells and NK cells, both phenotypically resembling cytokine-driven killer cells. In addition, patients displayed signatures of inflammatory and profibrotic CCR2+ CD163+ monocytes, coupled with elevated serum-soluble CD163, that may be linked to the late gadolinium enhancement on cardiac MRI, which can persist for months after vaccination. Together, our results demonstrate up-regulation in inflammatory cytokines and corresponding lymphocytes with tissue-damaging capabilities, suggesting a cytokine-dependent pathology, which may further be accompanied by myeloid cell–associated cardiac fibrosis. These findings likely rule out some previously proposed mechanisms of mRNA vaccine–-associated myopericarditis and point to new ones with relevance to vaccine development and clinical care. Description Cytokine-driven killer lymphocytes and inflammatory monocytes characterize myopericarditis after SARS-CoV-2 mRNA vaccination. Immunopathology signatures in myocarditis Myocarditis and/or pericarditis are rare adverse cardiac events observed after SARS-CoV-2 mRNA vaccination with a predilection for adolescent and young adult males. To investigate the pathogenesis of myopericarditis in this setting, Barmada and Klein et al. used unbiased immune profiling techniques to search for immune signatures that distinguished patients who developed myopericarditis from healthy vaccinated controls. Immune events associated with myopericarditis included elevated systemic levels of cytokines, an increased frequency of activated T and NK cells, and induction of inflammatory monocytes with profibrotic features. Neither immune targeting of cardiac autoantigens nor enhanced clonal expansion of B and T lymphocytes was detected. These findings provide deeper insights into the chain of events that can rarely lead to myopericarditis in the mRNA vaccine setting. —IRW

[1]  A. Ring,et al.  SARS-CoV-2 mRNA vaccines decouple anti-viral immunity from humoral autoimmunity , 2023, Nature Communications.

[2]  W. Lau,et al.  Influenza vaccination reveals sex dimorphic imprints of prior mild COVID-19 , 2023, Nature.

[3]  A. Randolph,et al.  Circulating Spike Protein Detected in Post–COVID-19 mRNA Vaccine Myocarditis , 2023, Circulation.

[4]  Lisa M. Domke,et al.  Autopsy-based histopathological characterization of myocarditis after anti-SARS-CoV-2-vaccination , 2022, Clinical Research in Cardiology.

[5]  B. Fireman,et al.  Incidence of Myocarditis/Pericarditis Following mRNA COVID-19 Vaccination Among Children and Younger Adults in the United States , 2022, Annals of Internal Medicine.

[6]  A. Keren,et al.  IL-1RA Antibodies in Myocarditis after SARS-CoV-2 Vaccination , 2022, The New England journal of medicine.

[7]  M. Bertrand,et al.  Age and sex-specific risks of myocarditis and pericarditis following Covid-19 messenger RNA vaccines , 2022, Nature Communications.

[8]  M. Flesch,et al.  Intramyocardial Inflammation after COVID-19 Vaccination: An Endomyocardial Biopsy-Proven Case Series , 2022, International journal of molecular sciences.

[9]  S. Lane,et al.  Reports of myocarditis and pericarditis following mRNA COVID-19 vaccination: a systematic review of spontaneously reported data from the UK, Europe and the USA and of the scientific literature , 2022, BMJ Open.

[10]  M. Oster,et al.  Comparison of Multisystem Inflammatory Syndrome in Children–Related Myocarditis, Classic Viral Myocarditis, and COVID‐19 Vaccine‐Related Myocarditis in Children , 2022, Journal of the American Heart Association.

[11]  S. D. de Ferranti,et al.  SARS-CoV-2 Infection and Associated Cardiovascular Manifestations and Complications in Children and Young Adults: A Scientific Statement From the American Heart Association , 2022, Circulation.

[12]  A. Venkatakrishnan,et al.  Surveillance of Safety of 3 Doses of COVID-19 mRNA Vaccination Using Electronic Health Records , 2022, JAMA network open.

[13]  O. Pappo,et al.  Severe Acute Myocarditis after the Third (Booster) Dose of mRNA COVID-19 Vaccination , 2022, Vaccines.

[14]  C. Forrest,et al.  Cardiac Complications After SARS-CoV-2 Infection and mRNA COVID-19 Vaccination — PCORnet, United States, January 2021–January 2022 , 2022, MMWR. Morbidity and mortality weekly report.

[15]  T. Shimabukuro,et al.  Safety Monitoring of COVID-19 Vaccine Booster Doses Among Persons Aged 12–17 Years — United States, December 9, 2021–February 20, 2022 , 2022, MMWR. Morbidity and mortality weekly report.

[16]  T. Shimabukuro,et al.  Safety of mRNA vaccines administered during the initial 6 months of the US COVID-19 vaccination programme: an observational study of reports to the Vaccine Adverse Event Reporting System and v-safe , 2022, The Lancet Infectious Diseases.

[17]  C. Steinwender,et al.  Myocarditis following mRNA COVID‐19 vaccination: call for endomyocardial biopsy , 2022, ESC heart failure.

[18]  C. Steenbergen,et al.  Increased Interleukin 18-Dependent Immune Responses Are Associated With Myopericarditis After COVID-19 mRNA Vaccination , 2022, Frontiers in Immunology.

[19]  M. Askenazi,et al.  Immunopathological signatures in multisystem inflammatory syndrome in children and pediatric COVID-19 , 2022, Nature Medicine.

[20]  C. Szoeke,et al.  COVID-19 vaccines: Considering sex differences in efficacy and safety , 2022, Contemporary Clinical Trials.

[21]  Benjamin Bowe,et al.  Long-term cardiovascular outcomes of COVID-19 , 2022, Nature Medicine.

[22]  E. Meffre,et al.  High-throughput identification of autoantibodies that target the human exoproteome , 2022, Cell reports methods.

[23]  T. Murohara,et al.  Biopsy-Proven Fulminant Myocarditis Requiring Mechanical Circulatory Support Following COVID-19 mRNA Vaccination , 2022, CJC Open.

[24]  A. Chiti,et al.  Advanced Imaging Supports the Mechanistic Role of Autoimmunity and Plaque Rupture in COVID-19 Heart Involvement , 2022, Clinical Reviews in Allergy & Immunology.

[25]  Michael John Smith,et al.  Myocarditis Cases Reported After mRNA-Based COVID-19 Vaccination in the US From December 2020 to August 2021. , 2022, JAMA.

[26]  Bjoern Peters,et al.  Lack of evidence of significant homology of SARS-CoV-2 spike sequences to myocarditis-associated antigens , 2022, eBioMedicine.

[27]  M. Artyomov,et al.  Single Cell Transcriptomics Reveals Cell Type Specific Diversification in Human Heart Failure , 2021, bioRxiv.

[28]  I. Mellman,et al.  IL-1 and IL-1ra are key regulators of the inflammatory response to RNA vaccines , 2022, Nature Immunology.

[29]  W. Gellad Myocarditis after vaccination against covid-19 , 2021, BMJ.

[30]  H. Sørensen,et al.  SARS-CoV-2 vaccination and myocarditis or myopericarditis: population based cohort study , 2021, BMJ.

[31]  K. Khunti,et al.  Risks of myocarditis, pericarditis, and cardiac arrhythmias associated with COVID-19 vaccination or SARS-CoV-2 infection , 2021, Nature Medicine.

[32]  S. Heymans,et al.  Myocarditis after COVID-19 mRNA vaccination: clinical observations and potential mechanisms , 2021, Nature Reviews Cardiology.

[33]  S. Pittaluga,et al.  Evidence of SARS-CoV-2-Specific T-Cell-Mediated Myocarditis in a MIS-A Case , 2021, Frontiers in Immunology.

[34]  S. Pittaluga,et al.  Evidence of SARS-CoV-2-Specific T-Cell-Mediated Myocarditis in a MIS-A Case , 2021, Frontiers in Immunology.

[35]  M. Dalamaga,et al.  Potential implications of lipid nanoparticles in the pathogenesis of myocarditis associated with the use of mRNA vaccines against SARS-CoV-2 , 2021, Metabolism Open.

[36]  J. Biliński,et al.  Sex Differences in Incidence, Clinical Characteristics and Outcomes in Children and Young Adults Hospitalized for Clinically Suspected Myocarditis in the Last Ten Years—Data from the MYO-PL Nationwide Database , 2021, Journal of clinical medicine.

[37]  D. Longo,et al.  A Possible Role for Anti-idiotype Antibodies in SARS-CoV-2 Infection and Vaccination. , 2021, The New England journal of medicine.

[38]  Fabian J Theis,et al.  SARS-CoV-2 infection triggers profibrotic macrophage responses and lung fibrosis , 2021, Cell.

[39]  B. Igyártó,et al.  The mRNA-LNP platform's lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory , 2021, iScience.

[40]  Sadeer G. Al-Kindi,et al.  COVID-lateral damage: cardiovascular manifestations of SARS-CoV-2 infection , 2021, Translational Research.

[41]  Aaron M. Rosenfeld,et al.  Lipid nanoparticles enhance the efficacy of mRNA and protein subunit vaccines by inducing robust T follicular helper cell and humoral responses , 2021, Immunity.

[42]  R. Balicer,et al.  Myocarditis after Covid-19 Vaccination in a Large Health Care Organization , 2021, The New England journal of medicine.

[43]  Shengshou Hu,et al.  Resolving the intertwining of inflammation and fibrosis in human heart failure at single-cell level , 2021, Basic Research in Cardiology.

[44]  C. Olson,et al.  Preliminary Findings of mRNA Covid-19 Vaccine Safety in Pregnant Persons. , 2021, The New England journal of medicine.

[45]  R. Poppiti,et al.  Lymphohistocytic myocarditis after Ad26.COV2.S viral vector COVID-19 vaccination , 2021, IJC Heart & Vasculature.

[46]  K. Klingel,et al.  Biopsy-proven lymphocytic myocarditis following first mRNA COVID-19 vaccination in a 40-year-old male: case report , 2021, Clinical Research in Cardiology.

[47]  Ben Y. Reis,et al.  Safety of the BNT162b2 mRNA Covid-19 Vaccine in a Nationwide Setting , 2021, The New England journal of medicine.

[48]  D. Weissman,et al.  mRNA vaccines for infectious diseases: principles, delivery and clinical translation , 2021, Nature Reviews Drug Discovery.

[49]  R. Kerbl [Myocarditis after COVID-19 mRNA vaccination]. , 2021, Monatsschrift Kinderheilkunde : Organ der Deutschen Gesellschaft fur Kinderheilkunde.

[50]  R. Langer,et al.  Lipid nanoparticles for mRNA delivery , 2021, Nature Reviews Materials.

[51]  A. Robicsek,et al.  Myocarditis and Pericarditis After Vaccination for COVID-19. , 2021, JAMA.

[52]  J. Polesel,et al.  Sex Disparities in Efficacy in COVID-19 Vaccines: A Systematic Review and Meta-Analysis , 2021, Vaccines.

[53]  M. Dimopoulos,et al.  Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients , 2021, Cell Reports.

[54]  P. Hotez,et al.  Myocarditis With COVID-19 mRNA Vaccines , 2021, Circulation.

[55]  Mark M. Davis,et al.  Systems vaccinology of the BNT162b2 mRNA vaccine in humans , 2021, Nature.

[56]  A. Haimovich,et al.  Myopericarditis in young adults presenting to the emergency department after receiving a second COVID‐19 mRNA vaccine , 2021, Academic emergency medicine : official journal of the Society for Academic Emergency Medicine.

[57]  L. Cooper,et al.  Myocarditis Following Immunization With mRNA COVID-19 Vaccines in Members of the US Military. , 2021, JAMA cardiology.

[58]  A. Khera,et al.  Myocarditis Temporally Associated with COVID-19 Vaccination. , 2021, Circulation.

[59]  L. Cooper,et al.  Myocarditis after BNT162b2 and mRNA-1273 Vaccination. , 2021, Circulation.

[60]  K. Batra,et al.  In-Depth Evaluation of a Case of Presumed Myocarditis After the Second Dose of COVID-19 mRNA Vaccine , 2021, Circulation.

[61]  C. Fuss,et al.  Symptomatic Acute Myocarditis in 7 Adolescents After Pfizer-BioNTech COVID-19 Vaccination , 2021, Pediatrics.

[62]  G. Cicero,et al.  Myocarditis After SARS-CoV-2 Vaccination: A Vaccine-Induced Reaction? , 2021, Canadian Journal of Cardiology.

[63]  V. Regitz-Zagrosek,et al.  Sex-Specific Differences of the Inflammatory State in Experimental Autoimmune Myocarditis , 2021, Frontiers in Immunology.

[64]  J. Casanova,et al.  Polyclonal expansion of TCR Vbeta 21.3+ CD4+ and CD8+ T cells is a hallmark of Multisystem Inflammatory Syndrome in Children , 2021, Science Immunology.

[65]  Shuangge Ma,et al.  Diverse functional autoantibodies in patients with COVID-19 , 2021, Nature.

[66]  J. Schultze Deutsche COVID-19 Omics Initiative (DeCOI) , 2021, BIOspektrum.

[67]  Joseph C. Sun,et al.  Natural Killer Cells: From Innate to Adaptive Features. , 2021, Annual review of immunology.

[68]  D. Hafler,et al.  Immune dysregulation and autoreactivity correlate with disease severity in SARS-CoV-2-associated multisystem inflammatory syndrome in children , 2021, Immunity.

[69]  M. H. Cheng,et al.  HLA class I-associated expansion of TRBV11-2 T cells in Multisystem Inflammatory Syndrome in Children. , 2021, The Journal of clinical investigation.

[70]  B. Igyártó,et al.  The mRNA-LNP platform’s lipid nanoparticle component used in preclinical vaccine studies is highly inflammatory , 2021, bioRxiv.

[71]  Benjamin Bowe,et al.  High-dimensional characterization of post-acute sequelae of COVID-19 , 2021, Nature.

[72]  J. Garot,et al.  Prognostic Impact of Late Gadolinium Enhancement by Cardiovascular Magnetic Resonance in Myocarditis: A Systematic Review and Meta-Analysis. , 2021, Circulation. Cardiovascular imaging.

[73]  M. Büttner,et al.  scCODA is a Bayesian model for compositional single-cell data analysis , 2020, Nature Communications.

[74]  Anushya Muruganujan,et al.  The Gene Ontology resource: enriching a GOld mine , 2020, Nucleic Acids Res..

[75]  Q. Yue,et al.  C-C chemokine receptor 5 signaling contributes to cardiac remodeling and dysfunction under pressure overload , 2020, Molecular medicine reports.

[76]  H. Møller,et al.  Macrophage Activation Markers, Soluble CD163 and Mannose Receptor, in Liver Fibrosis , 2021, Frontiers in Medicine.

[77]  Murugesan V. S. Rajaram,et al.  Role of Cardiac Macrophages on Cardiac Inflammation, Fibrosis and Tissue Repair , 2020, Cells.

[78]  J. Mascola,et al.  Efficacy and Safety of the mRNA-1273 SARS-CoV-2 Vaccine , 2020, The New England journal of medicine.

[79]  O. Blagova,et al.  Anti‐heart antibodies levels and their correlation with clinical symptoms and outcomes in patients with confirmed or suspected diagnosis COVID‐19 , 2020, European journal of immunology.

[80]  Elizabeth B White,et al.  Diverse Functional Autoantibodies in Patients with COVID-19 , 2020, Nature.

[81]  P. Dormitzer,et al.  Safety and Efficacy of the BNT162b2 mRNA Covid-19 Vaccine , 2020, The New England journal of medicine.

[82]  A. Cohen-Solal,et al.  Delayed acute myocarditis and COVID‐19‐related multisystem inflammatory syndrome , 2020, ESC heart failure.

[83]  S. Heymans,et al.  Myocarditis and inflammatory cardiomyopathy: current evidence and future directions , 2020, Nature Reviews Cardiology.

[84]  Catherine L. Worth,et al.  Cells of the adult human heart , 2020, Nature.

[85]  M. Netea,et al.  Trained Innate Immunity, Epigenetics, and Covid-19. , 2020, The New England journal of medicine.

[86]  Eric Song,et al.  Longitudinal analyses reveal immunological misfiring in severe COVID-19 , 2020, Nature.

[87]  M. Netea,et al.  The specifics of innate immune memory , 2020, Science.

[88]  R. Flavell,et al.  NK cell receptor NKG2D enforces proinflammatory features and pathogenicity of Th1 and Th17 cells , 2020, The Journal of experimental medicine.

[89]  Philip L. Felgner,et al.  A serological assay to detect SARS-CoV-2 seroconversion in humans , 2020, medRxiv.

[90]  R. Beigel,et al.  Sex-Based Differences in Characteristics and In-Hospital Outcomes among Patients With Diagnosed Acute Myocarditis. , 2020, The American journal of cardiology.

[91]  R. Xavier,et al.  Defining trained immunity and its role in health and disease , 2020, Nature Reviews Immunology.

[92]  Nicholas Borcherding,et al.  scRepertoire: An R-based toolkit for single-cell immune receptor analysis , 2020, F1000Research.

[93]  Dose-Finding Trial to Evaluate the Safety and Immunogenicity of Cytomegalovirus Vaccine mRNA-1647 in Healthy Adults , 2020, Case Medical Research.

[94]  T. Lüscher,et al.  Clinical Presentation and Laboratory Findings in Men Versus Women with Myocarditis. , 2020, Journal of Women's Health.

[95]  A. Teixeira-Carvalho,et al.  Increased frequencies of circulating CCR5+ memory T cells are correlated to chronic chagasic cardiomyopathy progression , 2019, Journal of leukocyte biology.

[96]  Aviv Regev,et al.  Intra- and Inter-cellular Rewiring of the Human Colon during Ulcerative Colitis , 2019, Cell.

[97]  Reza Masoomi,et al.  National Trends, Gender, Management, and Outcomes of Patients Hospitalized for Myocarditis. , 2019, The American journal of cardiology.

[98]  M. Siedner,et al.  Immune Correlates of Diffuse Myocardial Fibrosis and Diastolic Dysfunction Among Aging Women With Human Immunodeficiency Virus. , 2019, The Journal of infectious diseases.

[99]  C. Hedrick,et al.  Nonclassical Monocytes in Health and Disease. , 2019, Annual review of immunology.

[100]  A. Lichtman,et al.  T cell checkpoint regulators in the heart , 2019, Cardiovascular research.

[101]  M. Aronovitz,et al.  CXCR3 regulates CD4+ T cell cardiotropism in pressure overload-induced cardiac dysfunction. , 2019, JCI insight.

[102]  Allon M Klein,et al.  Scrublet: Computational Identification of Cell Doublets in Single-Cell Transcriptomic Data. , 2019, Cell systems.

[103]  N. Frangogiannis,et al.  The Functional Heterogeneity of Resident Cardiac Macrophages in Myocardial InjuryCCR2+ Cells Promote Inflammation, Whereas CCR2- Cells Protect. , 2019, Circulation research.

[104]  Y. Fujio,et al.  Blockade of NKG2D/NKG2D ligand interaction attenuated cardiac remodelling after myocardial infarction , 2018, Cardiovascular research.

[105]  M. Aronovitz,et al.  CXCR 3 regulates CD 4 + T cell cardiotropism in pressure overload induced cardiac dysfunction , 2019 .

[106]  T. Yokochi,et al.  A case of biopsy-proven eosinophilic myocarditis related to tetanus toxoid immunization. , 2018, Cardiovascular pathology : the official journal of the Society for Cardiovascular Pathology.

[107]  Michael I. Jordan,et al.  Deep Generative Modeling for Single-cell Transcriptomics , 2018, Nature Methods.

[108]  C. Dézsi,et al.  The Role of CXCR3 and Associated Chemokines in the Development of Atherosclerosis and During Myocardial Infarction , 2018, Front. Immunol..

[109]  Samuel L. Wolock,et al.  Scrublet: computational identification of cell doublets in single-cell transcriptomic data , 2018, bioRxiv.

[110]  D. Kreisel,et al.  The Human Heart Contains Distinct Macrophage Subsets with Divergent Origins and Functions , 2018, Nature Medicine.

[111]  Paul Hoffman,et al.  Integrating single-cell transcriptomic data across different conditions, technologies, and species , 2018, Nature Biotechnology.

[112]  B. Polić,et al.  NKG2D: A Master Regulator of Immune Cell Responsiveness , 2018, Front. Immunol..

[113]  G. Lip,et al.  Role of Monocytes in Heart Failure and Atrial Fibrillation , 2018, Journal of the American Heart Association.

[114]  W. Kang,et al.  Innate‐like Cytotoxic Function of Bystander‐Activated CD8+ T Cells Is Associated with Liver Injury in Acute Hepatitis A , 2018, Immunity.

[115]  Jixin Zhong,et al.  S100 Proteins As an Important Regulator of Macrophage Inflammation , 2018, Front. Immunol..

[116]  M. Delgado-Rodríguez,et al.  Systematic review and meta-analysis. , 2017, Medicina intensiva.

[117]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[118]  D. Raulet,et al.  Endothelial cells express NKG2D ligands and desensitize antitumor NK responses , 2017, eLife.

[119]  H. Swerdlow,et al.  Large-scale simultaneous measurement of epitopes and transcriptomes in single cells , 2017, Nature Methods.

[120]  Y. Arbel,et al.  Sex‐based differences in prevalence and clinical presentation among pericarditis and myopericarditis patients☆,☆☆,★,★★ , 2017, The American journal of emergency medicine.

[121]  Grace X. Y. Zheng,et al.  Massively parallel digital transcriptional profiling of single cells , 2016, Nature Communications.

[122]  A. Santoni,et al.  Regulation of NKG2D Expression and Signaling by Endocytosis. , 2016, Trends in immunology.

[123]  F. Zouein,et al.  The CXCL10/CXCR3 Axis and Cardiac Inflammation: Implications for Immunotherapy to Treat Infectious and Noninfectious Diseases of the Heart , 2016, Journal of immunology research.

[124]  M. Lederman,et al.  IL-15 promotes activation and expansion of CD8+ T cells in HIV-1 infection. , 2016, The Journal of clinical investigation.

[125]  M. Lindsey,et al.  MMP-9 signaling in the left ventricle following myocardial infarction. , 2016, American journal of physiology. Heart and circulatory physiology.

[126]  L. Cooper,et al.  Cardiac myosin-Th17 responses promote heart failure in human myocarditis. , 2016, JCI insight.

[127]  Steven H. Kleinstein,et al.  Change-O: a toolkit for analyzing large-scale B cell immunoglobulin repertoire sequencing data , 2015, Bioinform..

[128]  G. Anderson,et al.  Hepatocyte Growth Factor Receptor c-Met Instructs T Cell Cardiotropism and Promotes T Cell Migration to the Heart via Autocrine Chemokine Release , 2015, Immunity.

[129]  Charles P. Lin,et al.  Silencing of CCR2 in myocarditis. , 2015, European heart journal.

[130]  Benjamin G. Gowen,et al.  A shed NKG2D ligand that promotes natural killer cell activation and tumor rejection , 2015, Science.

[131]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[132]  Charles P. Lin,et al.  Silencing of CCR 2 in myocarditis , 2015 .

[133]  C. Kramer,et al.  Late Gadolinium Enhancement on Cardiac Magnetic Resonance Predicts Adverse Cardiovascular Outcomes in Nonischemic Cardiomyopathy: A Systematic Review and Meta-Analysis , 2014, Circulation. Cardiovascular imaging.

[134]  P. Rautava,et al.  [Clinical profile and influences on outcomes in patients hospitalized for acute pericarditis]. , 2014, Duodecim; laaketieteellinen aikakauskirja.

[135]  V. Kytö,et al.  The effects of gender and age on occurrence of clinically suspected myocarditis in adulthood , 2013, Heart.

[136]  P. Amouyel,et al.  Serum MMP-8: A Novel Indicator of Left Ventricular Remodeling and Cardiac Outcome in Patients after Acute Myocardial Infarction , 2013, PloS one.

[137]  P. Rautava,et al.  Gender differences in myocarditis: a nationwide study in Finland , 2013 .

[138]  M. Lindsey,et al.  Matrix metalloproteinase (MMP)-9: a proximal biomarker for cardiac remodeling and a distal biomarker for inflammation. , 2013, Pharmacology & therapeutics.

[139]  Ming-Ru Wu,et al.  NKG2D ligands as therapeutic targets. , 2013, Cancer immunity.

[140]  I. Taniguchi,et al.  Scoring of late gadolinium enhancement in cardiac magnetic resonance imaging can predict cardiac events in patients with hypertrophic cardiomyopathy. , 2011, Journal of cardiology.

[141]  M. Matloubian,et al.  Expression of chemokine receptor CXCR3 on T cells affects the balance between effector and memory CD8 T-cell generation , 2011, Proceedings of the National Academy of Sciences.

[142]  K. Matsushima,et al.  Chemokine receptor CXCR3 facilitates CD8+ T cell differentiation into short-lived effector cells leading to memory degeneration , 2011, The Journal of experimental medicine.

[143]  S. Sell,et al.  Inflammatory chemokine receptors regulate CD8+ T cell contraction and memory generation following infection , 2011, The Journal of experimental medicine.

[144]  R. Vasan,et al.  Biomarkers of Extracellular Matrix Metabolism (MMP-9 and TIMP-1) and Risk of Stroke, Myocardial Infarction, and Cause-Specific Mortality: Cohort Study , 2011, PloS one.

[145]  L. Lanier,et al.  Effect of NKG2D ligand expression on host immune responses , 2010, Immunological reviews.

[146]  Mark D. Robinson,et al.  edgeR: a Bioconductor package for differential expression analysis of digital gene expression data , 2009, Bioinform..

[147]  M. Friedrich,et al.  Age and gender effects on the extent of myocardial involvement in acute myocarditis: a cardiovascular magnetic resonance study , 2009, Heart.

[148]  B. Suarez-Alvarez,et al.  Potential Role of NKG2D and Its Ligands in Organ Transplantation: New Target for Immunointervention , 2009, American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons.

[149]  Y. Finkelstein,et al.  Eosinophilic Myocarditis Temporally Associated With Conjugate Meningococcal C and Hepatitis B Vaccines in Children , 2008, The Pediatric infectious disease journal.

[150]  R. Negrin,et al.  Silencing Human NKG2D, DAP10, and DAP12 Reduces Cytotoxicity of Activated CD8+ T Cells and NK Cells1 , 2005, The Journal of Immunology.

[151]  J. Coligan,et al.  Cutting Edge: NKG2D Is a Costimulatory Receptor for Human Naive CD8+ T Cells , 2005, The Journal of Immunology.

[152]  M. Rossi,et al.  CCR5 Plays a Critical Role in the Development of Myocarditis and Host Protection in Mice Infected with Trypanosoma cruzi , 2005, The Journal of infectious diseases.

[153]  W. Edwards,et al.  Eosinophilic-lymphocytic myocarditis after smallpox vaccination , 2003, The Lancet.

[154]  G. McMahon,et al.  Expression of the Chemokine Receptor CXCR3 and Its Ligand IP-10 During Human Cardiac Allograft Rejection , 2001 .

[155]  W. Hancock,et al.  Requirement of the Chemokine Receptor CXCR3 for Acute Allograft Rejection , 2000, The Journal of experimental medicine.

[156]  M. Ashburner,et al.  Gene Ontology: tool for the unification of biology , 2000, Nature Genetics.

[157]  M. Ashburner,et al.  The Gene Ontology Consortium , 2000 .

[158]  T. Waldmann,et al.  The multifaceted regulation of interleukin-15 expression and the role of this cytokine in NK cell differentiation and host response to intracellular pathogens. , 1999, Annual review of immunology.

[159]  M. Caligiuri,et al.  Interleukin (IL) 15 is a novel cytokine that activates human natural killer cells via components of the IL-2 receptor , 1994, The Journal of experimental medicine.