The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species

IntroductionCardiac immunology studies in murine models have identified a sizeable population of myocardial B-cells and have shown that its modulation represents a promising strategy to develop novel therapies for heart failure. However, scarce data on B-cells in the human heart leaves unclear wheth...

Full description

Bibliographic Details
Main Authors: Kevin C. Bermea, Nicolas Kostelecky, Sylvie T. Rousseau, Chieh-Yu Lin, Luigi Adamo
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-09-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fimmu.2022.973211/full
_version_ 1797997544082505728
author Kevin C. Bermea
Nicolas Kostelecky
Sylvie T. Rousseau
Chieh-Yu Lin
Luigi Adamo
author_facet Kevin C. Bermea
Nicolas Kostelecky
Sylvie T. Rousseau
Chieh-Yu Lin
Luigi Adamo
author_sort Kevin C. Bermea
collection DOAJ
description IntroductionCardiac immunology studies in murine models have identified a sizeable population of myocardial B-cells and have shown that its modulation represents a promising strategy to develop novel therapies for heart failure. However, scarce data on B-cells in the human heart leaves unclear whether findings in rodents are relevant to human biology.MethodsWe performed immunohistochemical stains to characterize the amount and distribution of B-cells in human hearts, analyzing both fresh and post-mortem tissue. To gain insight into the biology of human myocardial B-cells we analyzed publicly-available spatial transcriptomics and single-cell sequencing datasets of myocardial and peripheral blood mononuclear cells (PBMCs). We validated these findings on primary B-cells sorted from the heart and peripheral blood of left ventricular assistive device recipients. To identify biological pathways upregulated in myocardial B-cells across species, we compared differential gene expression in myocardial vs peripheral blood B-cells across the studied human datasets and published rodent datasets.ResultsIn healthy human heart samples, we found B-cells at a ratio of 1:8 compared to T-cells (2.41 ± 0.45 vs 19.36 ± 4.43, p-value <0.001). Myocardial B-cells were more abundant in the interstitium compared with the intravascular space (p-value=0.011), and also more abundant in the myocardium vs. epicardium (p-value=0.048). Single-cell gene expression analysis showed that the human myocardium harbored mostly naive B-cells with a gene expression profile distinct from that of PBMC B-cells. Cross-comparison of differentially-expressed genes in myocardial vs. PBMC B-cells across human and rodent datasets identified 703 genes with consistent differential gene expression across species (binomial p-value=2.9e-48). KEGG pathway analysis highlighted “B-cell receptor signaling pathway,” “Antigen processing and presentation,” and “Cytokine-cytokine receptor interaction” among the top pathways upregulated in cardiac B-cells (FDR <0.001) conserved between species.ConclusionsLike the murine heart, the human heart harbors naive B-cells that are both intravascular and extravascular. Human myocardial B-cells are fewer and more evenly distributed between these two compartments than rodent myocardial B-cells. However, analysis of single-gene expression data indicates that the biological function of myocardial B-cells is conserved across species.
first_indexed 2024-04-11T10:33:26Z
format Article
id doaj.art-2b1a252758f247c3ba38a152532ff328
institution Directory Open Access Journal
issn 1664-3224
language English
last_indexed 2024-04-11T10:33:26Z
publishDate 2022-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Immunology
spelling doaj.art-2b1a252758f247c3ba38a152532ff3282022-12-22T04:29:21ZengFrontiers Media S.A.Frontiers in Immunology1664-32242022-09-011310.3389/fimmu.2022.973211973211The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across speciesKevin C. Bermea0Nicolas Kostelecky1Sylvie T. Rousseau2Chieh-Yu Lin3Luigi Adamo4Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United StatesDepartment of Pathology, Washington University in St. Louis School of Medicine, St. Louis, MO, United StatesDivision of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United StatesDepartment of Pathology, Washington University in St. Louis School of Medicine, St. Louis, MO, United StatesDivision of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, United StatesIntroductionCardiac immunology studies in murine models have identified a sizeable population of myocardial B-cells and have shown that its modulation represents a promising strategy to develop novel therapies for heart failure. However, scarce data on B-cells in the human heart leaves unclear whether findings in rodents are relevant to human biology.MethodsWe performed immunohistochemical stains to characterize the amount and distribution of B-cells in human hearts, analyzing both fresh and post-mortem tissue. To gain insight into the biology of human myocardial B-cells we analyzed publicly-available spatial transcriptomics and single-cell sequencing datasets of myocardial and peripheral blood mononuclear cells (PBMCs). We validated these findings on primary B-cells sorted from the heart and peripheral blood of left ventricular assistive device recipients. To identify biological pathways upregulated in myocardial B-cells across species, we compared differential gene expression in myocardial vs peripheral blood B-cells across the studied human datasets and published rodent datasets.ResultsIn healthy human heart samples, we found B-cells at a ratio of 1:8 compared to T-cells (2.41 ± 0.45 vs 19.36 ± 4.43, p-value <0.001). Myocardial B-cells were more abundant in the interstitium compared with the intravascular space (p-value=0.011), and also more abundant in the myocardium vs. epicardium (p-value=0.048). Single-cell gene expression analysis showed that the human myocardium harbored mostly naive B-cells with a gene expression profile distinct from that of PBMC B-cells. Cross-comparison of differentially-expressed genes in myocardial vs. PBMC B-cells across human and rodent datasets identified 703 genes with consistent differential gene expression across species (binomial p-value=2.9e-48). KEGG pathway analysis highlighted “B-cell receptor signaling pathway,” “Antigen processing and presentation,” and “Cytokine-cytokine receptor interaction” among the top pathways upregulated in cardiac B-cells (FDR <0.001) conserved between species.ConclusionsLike the murine heart, the human heart harbors naive B-cells that are both intravascular and extravascular. Human myocardial B-cells are fewer and more evenly distributed between these two compartments than rodent myocardial B-cells. However, analysis of single-gene expression data indicates that the biological function of myocardial B-cells is conserved across species.https://www.frontiersin.org/articles/10.3389/fimmu.2022.973211/fullB-cellsheartcardiologycardiac immunologycardiac inflammationnaive B-cells
spellingShingle Kevin C. Bermea
Nicolas Kostelecky
Sylvie T. Rousseau
Chieh-Yu Lin
Luigi Adamo
The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
Frontiers in Immunology
B-cells
heart
cardiology
cardiac immunology
cardiac inflammation
naive B-cells
title The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
title_full The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
title_fullStr The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
title_full_unstemmed The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
title_short The human myocardium harbors a population of naive B-cells with a distinctive gene expression signature conserved across species
title_sort human myocardium harbors a population of naive b cells with a distinctive gene expression signature conserved across species
topic B-cells
heart
cardiology
cardiac immunology
cardiac inflammation
naive B-cells
url https://www.frontiersin.org/articles/10.3389/fimmu.2022.973211/full
work_keys_str_mv AT kevincbermea thehumanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT nicolaskostelecky thehumanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT sylvietrousseau thehumanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT chiehyulin thehumanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT luigiadamo thehumanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT kevincbermea humanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT nicolaskostelecky humanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT sylvietrousseau humanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT chiehyulin humanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies
AT luigiadamo humanmyocardiumharborsapopulationofnaivebcellswithadistinctivegeneexpressionsignatureconservedacrossspecies