1
|
Hrovatin K, Sikkema L, Shitov VA, Heimberg G, Shulman M, Oliver AJ, Mueller MF, Ibarra IL, Wang H, Ramírez-Suástegui C, He P, Schaar AC, Teichmann SA, Theis FJ, Luecken MD. Considerations for building and using integrated single-cell atlases. Nat Methods 2024:10.1038/s41592-024-02532-y. [PMID: 39672979 DOI: 10.1038/s41592-024-02532-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/22/2024] [Indexed: 12/15/2024]
Abstract
The rapid adoption of single-cell technologies has created an opportunity to build single-cell 'atlases' integrating diverse datasets across many laboratories. Such atlases can serve as a reference for analyzing and interpreting current and future data. However, it has become apparent that atlasing approaches differ, and the impact of these differences are often unclear. Here we review the current atlasing literature and present considerations for building and using atlases. Importantly, we find that no one-size-fits-all protocol for atlas building exists, but rather we discuss context-specific considerations and workflows, including atlas conceptualization, data collection, curation and integration, atlas evaluation and atlas sharing. We further highlight the benefits of integrated atlases for analyses of new datasets and deriving biological insights beyond what is possible from individual datasets. Our overview of current practices and associated recommendations will improve the quality of atlases to come, facilitating the shift to a unified, reference-based understanding of single-cell biology.
Collapse
Affiliation(s)
- Karin Hrovatin
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lisa Sikkema
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Vladimir A Shitov
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Graham Heimberg
- Department of OMNI Bioinformatics, Genentech, South San Francisco, CA, USA
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
| | - Maiia Shulman
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Michaela F Mueller
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Ignacio L Ibarra
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Hanchen Wang
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
- Department of Computer Science, Stanford University, Palo Alto, CA, USA
| | - Ciro Ramírez-Suástegui
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Anna C Schaar
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR MacMillan Multiscale Human Programme, Toronto, Ontario, Canada
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Garching, Germany.
| | - Malte D Luecken
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
2
|
Pedroni A, Yilmaz E, Del Vecchio L, Bhattarai P, Vidal IT, Dai YWE, Koutsogiannis K, Kizil C, Ampatzis K. Decoding the molecular, cellular, and functional heterogeneity of zebrafish intracardiac nervous system. Nat Commun 2024; 15:10483. [PMID: 39632839 PMCID: PMC11618350 DOI: 10.1038/s41467-024-54830-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024] Open
Abstract
The proper functioning of the heart relies on the intricate interplay between the central nervous system and the local neuronal networks within the heart itself. While the central innervation of the heart has been extensively studied, the organization and functionality of the intracardiac nervous system (IcNS) remain largely unexplored. Here, we present a comprehensive taxonomy of the IcNS, utilizing single-cell RNA sequencing, anatomical studies, and electrophysiological techniques. Our findings reveal a diverse array of neuronal types within the IcNS, exceeding previous expectations. We identify a subset of neurons exhibiting characteristics akin to pacemaker/rhythmogenic neurons similar to those found in Central Pattern Generator networks of the central nervous system. Our results underscore the heterogeneity within the IcNS and its key role in regulating the heart's rhythmic functionality. The classification and characterization of the IcNS presented here serve as a valuable resource for further exploration into the mechanisms underlying heart functionality and the pathophysiology of associated cardiac disorders.
Collapse
Affiliation(s)
- Andrea Pedroni
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Elanur Yilmaz
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Lisa Del Vecchio
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Prabesh Bhattarai
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA
| | - Inés Talaya Vidal
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Yu-Wen E Dai
- Department of Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | | | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA.
- Department of Neurology, Columbia University Irving Medical Center, Columbia University, New York, NY, 10032, USA.
| | | |
Collapse
|
3
|
Vascular cells of blood vessels and organs across the human body. Nat Med 2024; 30:3431-3432. [PMID: 39643674 DOI: 10.1038/s41591-024-03410-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
|
4
|
Li R, Strobl J, Poyner EFM, Balbaa A, Torabi F, Mazin PV, Chipampe NJ, Stephenson E, Ramírez-Suástegi C, Shanmugiah VBM, Gardner L, Olabi B, Coulthard R, Botting RA, Zila N, Prigmore E, Gopee NH, Chroscik MA, Kritikaki E, Engelbert J, Goh I, Chan HM, Johnson HF, Ellis J, Rowe V, Tun W, Reynolds G, Yang D, Foster AR, Gambardella L, Winheim E, Admane C, Rumney B, Steele L, Jardine L, Nenonen J, Pickard K, Lumley J, Hampton P, Hu S, Liu F, Liu X, Horsfall D, Basurto-Lozada D, Grimble L, Bacon CM, Weatherhead SC, Brauner H, Wang Y, Bai F, Reynolds NJ, Allen JE, Jonak C, Brunner PM, Teichmann SA, Haniffa M. Cutaneous T cell lymphoma atlas reveals malignant T H2 cells supported by a B cell-rich tumor microenvironment. Nat Immunol 2024; 25:2320-2330. [PMID: 39558094 PMCID: PMC11588665 DOI: 10.1038/s41590-024-02018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 10/11/2024] [Indexed: 11/20/2024]
Abstract
Cutaneous T cell lymphoma (CTCL) is a potentially fatal clonal malignancy of T cells primarily affecting the skin. The most common form of CTCL, mycosis fungoides, can be difficult to diagnose, resulting in treatment delay. We performed single-cell and spatial transcriptomics analysis of skin from patients with mycosis fungoides-type CTCL and an integrated comparative analysis with human skin cell atlas datasets from healthy and inflamed skin. We revealed the co-optation of T helper 2 (TH2) cell-immune gene programs by malignant CTCL cells and modeling of the tumor microenvironment to support their survival. We identified MHC-II+ fibroblasts and dendritic cells that can maintain TH2 cell-like tumor cells. CTCL tumor cells are spatially associated with B cells, forming tertiary lymphoid structure-like aggregates. Finally, we validated the enrichment of B cells in CTCL and its association with disease progression across three independent patient cohorts. Our findings provide diagnostic aids, potential biomarkers for disease staging and therapeutic strategies for CTCL.
Collapse
Affiliation(s)
- Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| | - Johanna Strobl
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Elizabeth F M Poyner
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Aya Balbaa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Pavel V Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Emily Stephenson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | | | - Louis Gardner
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Bayanne Olabi
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rowen Coulthard
- NovoPath, Department of Cellular Pathology, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Rachel A Botting
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Nina Zila
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
- Section Biomedical Science, University of Applied Sciences FH Campus Wien, Vienna, Austria
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nusayhah H Gopee
- Biosciences Institute, Newcastle University, Newcastle, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marta A Chroscik
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Efpraxia Kritikaki
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Justin Engelbert
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Issac Goh
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Hon Man Chan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Jasmine Ellis
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Victoria Rowe
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Win Tun
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Gary Reynolds
- Biosciences Institute, Newcastle University, Newcastle, UK
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Dexin Yang
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | | | | | - Elena Winheim
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Chloe Admane
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Benjamin Rumney
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Lloyd Steele
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Julia Nenonen
- Division of Dermatology, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Keir Pickard
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Jennifer Lumley
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Philip Hampton
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Simeng Hu
- Biomedical Pioneering Innovation Center and School of Life Sciences, Peking University, Beijing, China
| | - Fengjie Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Xiangjun Liu
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - David Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Daniela Basurto-Lozada
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Louise Grimble
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Chris M Bacon
- Wolfson Childhood Cancer Research Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sophie C Weatherhead
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Hanna Brauner
- Division of Dermatology, Department of Medicine, Solna and Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Dermatology, Karolinska University Hospital, Stockholm, Sweden
| | - Yang Wang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China
| | - Fan Bai
- Biomedical Pioneering Innovation Center and School of Life Sciences, Peking University, Beijing, China
| | - Nick J Reynolds
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Judith E Allen
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Biosciences Institute, Newcastle University, Newcastle, UK.
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle, Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Barnett SN, Cujba AM, Yang L, Maceiras AR, Li S, Kedlian VR, Pett JP, Polanski K, Miranda AMA, Xu C, Cranley J, Kanemaru K, Lee M, Mach L, Perera S, Tudor C, Joseph PD, Pritchard S, Toscano-Rivalta R, Tuong ZK, Bolt L, Petryszak R, Prete M, Cakir B, Huseynov A, Sarropoulos I, Chowdhury RA, Elmentaite R, Madissoon E, Oliver AJ, Campos L, Brazovskaja A, Gomes T, Treutlein B, Kim CN, Nowakowski TJ, Meyer KB, Randi AM, Noseda M, Teichmann SA. An organotypic atlas of human vascular cells. Nat Med 2024; 30:3468-3481. [PMID: 39566559 PMCID: PMC11645277 DOI: 10.1038/s41591-024-03376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
The human vascular system, comprising endothelial cells (ECs) and mural cells, covers a vast surface area in the body, providing a critical interface between blood and tissue environments. Functional differences exist across specific vascular beds, but their molecular determinants across tissues remain largely unknown. In this study, we integrated single-cell transcriptomics data from 19 human organs and tissues and defined 42 vascular cell states from approximately 67,000 cells (62 donors), including angiotypic transitional signatures along the arterial endothelial axis from large to small caliber vessels. We also characterized organotypic populations, including splenic littoral and blood-brain barrier ECs, thus clarifying the molecular profiles of these important cell states. Interrogating endothelial-mural cell molecular crosstalk revealed angiotypic and organotypic communication pathways related to Notch, Wnt, retinoic acid, prostaglandin and cell adhesion signaling. Transcription factor network analysis revealed differential regulation of downstream target genes in tissue-specific modules, such as those of FOXF1 across multiple lung vascular subpopulations. Additionally, we make mechanistic inferences of vascular drug targets within different vascular beds. This open-access resource enhances our understanding of angiodiversity and organotypic molecular signatures in human vascular cells, and has therapeutic implications for vascular diseases across tissues.
Collapse
Affiliation(s)
- Sam N Barnett
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Ana-Maria Cujba
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lu Yang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Ana Raquel Maceiras
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Shuang Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Veronika R Kedlian
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - J Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kazumasa Kanemaru
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Lukas Mach
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | - Zewen K Tuong
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Alik Huseynov
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Ioannis Sarropoulos
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rasheda A Chowdhury
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK
| | - Elo Madissoon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Lia Campos
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Tomás Gomes
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Barbara Treutlein
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Chang N Kim
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Tomasz J Nowakowski
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Anna M Randi
- National Heart and Lung Institute, Imperial College London, London, UK
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK.
- British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
6
|
Juhasz V, Charlier FT, Zhao TX, Tsiantoulas D. Targeting the adaptive immune continuum in atherosclerosis and post-MI injury. Atherosclerosis 2024; 399:118616. [PMID: 39546915 DOI: 10.1016/j.atherosclerosis.2024.118616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024]
Abstract
Atherosclerotic disease is a cholesterol-rich lipoprotein particle-driven disease resulting in the formation of atherosclerotic plaques in large and medium size arteries. Rupture or erosion of atherosclerotic plaques can trigger the formation of a thrombus causing the obstruction of the blood flow in the coronary artery and thereby leading to myocardial infarction (MI). Inflammation is a crucial pillar of the mechanisms leading to atherosclerosis and governing the cardiac repair post-MI. Dissecting the complex and sophisticated networks of the immune responses underlying the formation of atherosclerotic plaques and affecting the healing of the heart after MI will allow the designing of highly precise immunomodulatory therapies for these settings. Notably, MI also accelerates atherosclerosis via modulating the response of the immune system. Therefore, for the identification of effective and safe therapeutic targets, it is critical to consider the inflammatory continuum that interconnects the two pathologies and identify immunomodulatory strategies that confer a protective effect in both settings or at least, affect each pathology independently. Adaptive immunity, which consists of B and T lymphocytes, is a major regulator of atherosclerosis and post-MI cardiac repair. Here, we review and discuss the effect of potential adaptive immunity-targeting therapies, such as cell-depleting therapies, in atherosclerosis and post-MI cardiac injury.
Collapse
Affiliation(s)
- Viktoria Juhasz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fiona T Charlier
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | | |
Collapse
|
7
|
Yoshida T, Yoshida S, Inukai K, Kato K, Yura Y, Hattori T, Taki K, Enomoto A, Ohashi K, Okumura T, Ouchi N, Kawase H, Wettschureck N, Offermanns S, Murohara T, Takefuji M. ALPK2 prevents cardiac diastolic dysfunction in heart failure with preserved ejection fraction. FASEB J 2024; 38:e70192. [PMID: 39556326 PMCID: PMC11599786 DOI: 10.1096/fj.202402103r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/23/2024] [Accepted: 11/06/2024] [Indexed: 11/19/2024]
Abstract
Protein phosphorylation, controlled by protein kinases, is central to regulating various pathophysiological processes, including cardiac systolic function. The dysregulation of protein kinase activity plays a significant role in the pathogenesis of cardiac systolic dysfunction. While cardiac contraction mechanisms are well documented, the mechanisms underlying cardiac diastole remain elusive. This gap persists owing to the historical focus on systolic dysfunction in heart failure research. Recently, heart failure with preserved ejection fraction (HFpEF), an age-related disease characterized by cardiac diastolic dysfunction, has emerged as a major public health concern. However, its underlying mechanism remains unclear. In this study, we investigated cardiac protein kinases by analyzing the gene expression of 518 protein kinases in human tissues. We identified alpha-kinase 2 (ALPK2) as a novel cardiac-specific atypical kinase and generated tamoxifen-inducible, cardiomyocyte-specific Alpk2-knockout mice and Alpk2-overexpressing mice. Alpk2 deficiency did not affect cardiac systolic dysfunction in the myocardial infarction model or the pressure-overload-induced heart failure model. Notably, cardiomyocyte-specific Alpk2 deficiency exacerbated cardiac diastolic dysfunction induced by aging and in the HFpEF model. Conversely, Alpk2 overexpression increased the phosphorylation of tropomyosin 1, a major regulator that binds myosin to actin, and mitigated cardiac stiffness in HFpEF. This study provides novel evidence that ALPK2 represents a potential therapeutic target for cardiac diastolic dysfunction in HFpEF and age-related cardiac impairments.
Collapse
Affiliation(s)
- Tatsuya Yoshida
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Satoya Yoshida
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Kohei Inukai
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Katsuhiro Kato
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Yoshimitsu Yura
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Tomoki Hattori
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Kentaro Taki
- Division for Medical Research EngineeringNagoya University School of MedicineNagoyaJapan
| | - Atsushi Enomoto
- Department of PathologyNagoya University School of MedicineNagoyaJapan
| | - Koji Ohashi
- Department of Molecular Medicine and CardiologyNagoya University School of MedicineNagoyaJapan
| | - Takahiro Okumura
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and CardiologyNagoya University School of MedicineNagoyaJapan
| | - Haruya Kawase
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
- Department of PharmacologyMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Nina Wettschureck
- Department of PharmacologyMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Stefan Offermanns
- Department of PharmacologyMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Toyoaki Murohara
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| | - Mikito Takefuji
- Department of CardiologyNagoya University School of MedicineNagoyaJapan
| |
Collapse
|
8
|
Lim AA, Pouyabahar D, Ashraf M, Huang K, Lohbihler M, Murareanu BM, Chang ML, Kwan M, Alibhai FJ, Tran T, Mazine A, Laflamme MA, Bader GD, Laksman Z, Protze S. Single-cell transcriptome analysis reveals CD34 as a marker of human sinoatrial node pacemaker cardiomyocytes. Nat Commun 2024; 15:10206. [PMID: 39604360 PMCID: PMC11603134 DOI: 10.1038/s41467-024-54337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
The sinoatrial node regulates the heart rate throughout life. Failure of this primary pacemaker results in life-threatening, slow heart rhythm. Despite its critical function, the cellular and molecular composition of the human sinoatrial node is not resolved. Particularly, no cell surface marker to identify and isolate sinoatrial node pacemaker cells has been reported. Here we use single-nuclei/cell RNA sequencing of fetal and human pluripotent stem cell-derived sinoatrial node cells to reveal that they consist of three subtypes of pacemaker cells: Core Pacemaker, Sinus Venosus, and Transitional Cells. Our study identifies a host of sinoatrial node pacemaker markers including MYH11, BMP4, and the cell surface antigen CD34. We demonstrate that sorting for CD34+ cells from stem cell differentiation cultures enriches for sinoatrial node cells exhibiting a functional pacemaker phenotype. This sinoatrial node pacemaker cell surface marker is highly valuable for stem cell-based disease modeling, drug discovery, cell replacement therapies, and the targeted delivery of therapeutics to sinoatrial node cells in vivo using antibody-drug conjugates.
Collapse
Affiliation(s)
- Amos A Lim
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Delaram Pouyabahar
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Mishal Ashraf
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kate Huang
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- Experimental Medicine Program, University of British Columbia, Vancouver, BC, Canada
| | - Michelle Lohbihler
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Brandon M Murareanu
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Matthew L Chang
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Maggie Kwan
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Faisal J Alibhai
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Thinh Tran
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amine Mazine
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Michael A Laflamme
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Gary D Bader
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Zachary Laksman
- Centre for Heart and Lung Innovation, University of British Columbia and St. Paul's Hospital, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Experimental Medicine Program, University of British Columbia, Vancouver, BC, Canada
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie Protze
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
9
|
Ma XR, Conley SD, Kosicki M, Bredikhin D, Cui R, Tran S, Sheth MU, Qiu WL, Chen S, Kundu S, Kang HY, Amgalan D, Munger CJ, Duan L, Dang K, Rubio OM, Kany S, Zamirpour S, DePaolo J, Padmanabhan A, Olgin J, Damrauer S, Andersson R, Gu M, Priest JR, Quertermous T, Qiu X, Rabinovitch M, Visel A, Pennacchio L, Kundaje A, Glass IA, Gifford CA, Pirruccello JP, Goodyer WR, Engreitz JM. Molecular convergence of risk variants for congenital heart defects leveraging a regulatory map of the human fetal heart. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.20.24317557. [PMID: 39606363 PMCID: PMC11601760 DOI: 10.1101/2024.11.20.24317557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Congenital heart defects (CHD) arise in part due to inherited genetic variants that alter genes and noncoding regulatory elements in the human genome. These variants are thought to act during fetal development to influence the formation of different heart structures. However, identifying the genes, pathways, and cell types that mediate these effects has been challenging due to the immense diversity of cell types involved in heart development as well as the superimposed complexities of interpreting noncoding sequences. As such, understanding the molecular functions of both noncoding and coding variants remains paramount to our fundamental understanding of cardiac development and CHD. Here, we created a gene regulation map of the healthy human fetal heart across developmental time, and applied it to interpret the functions of variants associated with CHD and quantitative cardiac traits. We collected single-cell multiomic data from 734,000 single cells sampled from 41 fetal hearts spanning post-conception weeks 6 to 22, enabling the construction of gene regulation maps in 90 cardiac cell types and states, including rare populations of cardiac conduction cells. Through an unbiased analysis of all 90 cell types, we find that both rare coding variants associated with CHD and common noncoding variants associated with valve traits converge to affect valvular interstitial cells (VICs). VICs are enriched for high expression of known CHD genes previously identified through mapping of rare coding variants. Eight CHD genes, as well as other genes in similar molecular pathways, are linked to common noncoding variants associated with other valve diseases or traits via enhancers in VICs. In addition, certain common noncoding variants impact enhancers with activities highly specific to particular subanatomic structures in the heart, illuminating how such variants can impact specific aspects of heart structure and function. Together, these results implicate new enhancers, genes, and cell types in the genetic etiology of CHD, identify molecular convergence of common noncoding and rare coding variants on VICs, and suggest a more expansive view of the cell types instrumental in genetic risk for CHD, beyond the working cardiomyocyte. This regulatory map of the human fetal heart will provide a foundational resource for understanding cardiac development, interpreting genetic variants associated with heart disease, and discovering targets for cell-type specific therapies.
Collapse
Affiliation(s)
- X Rosa Ma
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Stephanie D Conley
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Michael Kosicki
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Danila Bredikhin
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Ran Cui
- Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Steven Tran
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Maya U Sheth
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Wei-Lin Qiu
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sijie Chen
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Soumya Kundu
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Helen Y Kang
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Current address: PhD Program in Computational and Systems Biology, MIT, Cambridge, MA, USA
| | - Dulguun Amgalan
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
| | - Chad J Munger
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Lauren Duan
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Katherine Dang
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Oriane Matthys Rubio
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Siavash Zamirpour
- School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - John DePaolo
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arun Padmanabhan
- Gladstone Institutes, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco School of Medicine, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Jeffrey Olgin
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Scott Damrauer
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Corporal Michael Crescenz VA Medical Center, Philadelphia, PA, USA
- Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Robin Andersson
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati School of Medicine, Cincinnati, OH, USA
| | - James R Priest
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Tenaya Therapeutics, South San Francisco, CA, USA
| | - Thomas Quertermous
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Xiaojie Qiu
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Stanford Bio-X, Stanford University, Stanford, CA, USA
| | - Marlene Rabinovitch
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Vera Moulton Wall Center for Pulmonary Vascular Diseases, Stanford University, Stanford, CA, USA
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California, Merced, Merced, CA, USA
| | - Len Pennacchio
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Ian A Glass
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Department of Pediatrics and Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Casey A Gifford
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, Department of Medicine and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, CA, USA
- Bakar Computation Health Sciences Institute, University of California, San Francisco, CA, USA
| | - William R Goodyer
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Jesse M Engreitz
- Basic Science and Engineering (BASE) Initiative, Stanford Children's Health, Betty Irene Moore Children's Heart Center, Stanford, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Maternal and Child Health Research Institute, Stanford University, Stanford, CA, USA
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA
- Gene Regulation Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
10
|
Wang Z, Dai R, Wang M, Lei L, Zhang Z, Han K, Wang Z, Guo Q. KanCell: dissecting cellular heterogeneity in biological tissues through integrated single-cell and spatial transcriptomics. J Genet Genomics 2024:S1673-8527(24)00310-2. [PMID: 39577768 DOI: 10.1016/j.jgg.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 11/07/2024] [Accepted: 11/10/2024] [Indexed: 11/24/2024]
Abstract
KanCell is a deep learning model based on Kolmogorov-Arnold networks (KAN) designed to enhance cellular heterogeneity analysis by integrating single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) data. ST technologies provide insights into gene expression within tissue context, revealing cellular interactions and microenvironments. To fully leverage this potential, effective computational models are crucial. We evaluate KanCell on both simulated and real datasets from technologies such as STARmap, Slide-seq, Visium, and Spatial Transcriptomics. Our results demonstrate that KanCell outperforms existing methods across metrics like PCC, SSIM, COSSIM, RMSE, JSD, ARS, and ROC, with robust performance under varying cell numbers and background noise. Real-world applications on human lymph nodes, hearts, melanoma, breast cancer, dorsolateral prefrontal cortex, and mouse embryo brains confirmed its reliability. Compared to traditional approaches, KanCell effectively captures non-linear relationships and optimizes computational efficiency through KAN, providing an accurate and efficient tool for ST. By improving data accuracy and resolving cell type composition, KanCell reveals cellular heterogeneity, clarifies disease microenvironments, and identifies therapeutic targets, addressing complex biological challenges.
Collapse
Affiliation(s)
- Zhenghui Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Ruoyan Dai
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Mengqiu Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Lixin Lei
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zhiwei Zhang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Kaitai Han
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Zijun Wang
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China
| | - Qianjin Guo
- Academy of Artificial Intelligence, Beijing Institute of Petrochemical Technology, Beijing 102617, China.
| |
Collapse
|
11
|
Tomida S, Ishima T, Nagai R, Aizawa K. T-Type Voltage-Gated Calcium Channels: Potential Regulators of Smooth Muscle Contractility. Int J Mol Sci 2024; 25:12420. [PMID: 39596484 PMCID: PMC11594734 DOI: 10.3390/ijms252212420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/16/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Emerging evidence has indicated a possible link between attenuation of contractility in aortic smooth muscle cells and pathogenesis of aortic dissection, as revealed through comprehensive, multi-omic analyses of familial thoracic aortic aneurysm and dissection models. While L-type voltage-gated calcium channels have been extensively investigated for their roles in smooth muscle contraction, more recent investigations have suggested that downregulation of T-type voltage-gated calcium channels, rather than their L-type counterparts, may be more closely associated with impaired contractility observed in vascular smooth muscle cells. This review provides a detailed examination of T-type voltage-gated calcium channels, highlighting their structure, electrophysiology, biophysics, expression patterns, functional roles, and potential mechanisms through which their downregulation may contribute to reduced contractile function. Furthermore, the application of multi-omic approaches in investigating calcium channels is discussed.
Collapse
Affiliation(s)
- Shota Tomida
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
- School of Medicine, Faculty of Medicine, Gunma University, Maebashi 371-8511, Japan
| | - Tamaki Ishima
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Ryozo Nagai
- Jichi Medical University, Shimotsuke 329-0498, Japan
| | - Kenichi Aizawa
- Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, Shimotsuke 329-0498, Japan
- Clinical Pharmacology Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
- Division of Translational Research, Clinical Research Center, Jichi Medical University Hospital, Shimotsuke 329-0498, Japan
| |
Collapse
|
12
|
Hill MC, Simonson B, Roselli C, Xiao L, Herndon CN, Chaffin M, Mantineo H, Atwa O, Bhasin H, Guedira Y, Bedi KC, Margulies KB, Klattenhoff CA, Tucker NR, Ellinor PT. Large-scale single-nuclei profiling identifies role for ATRNL1 in atrial fibrillation. Nat Commun 2024; 15:10002. [PMID: 39562555 PMCID: PMC11576987 DOI: 10.1038/s41467-024-54296-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia in humans, yet the molecular basis of AF remains incompletely understood. To determine the cell type-specific transcriptional changes underlying AF, we perform single-nucleus RNA-seq (snRNA-seq) on left atrial (LA) samples from patients with AF and controls. From more than 175,000 nuclei we find that only cardiomyocytes (CMs) and macrophages (MΦs) have a significant number of differentially expressed genes in patients with AF. Attractin Like 1 (ATRNL1) was overexpressed in CMs among patients with AF and localized to the intercalated disks. Further, in both knockdown and overexpression experiments we identify a potent role for ATRNL1 in cell stress response, and in the modulation of the cardiac action potential. Finally, we detect an unexpected expression pattern for a leading AF candidate gene, KCNN3. In sum, we uncover a role for ATRNL1 which may serve as potential therapeutic target for this common arrhythmia.
Collapse
Affiliation(s)
- Matthew C Hill
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Bridget Simonson
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carolina Roselli
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ling Xiao
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Caroline N Herndon
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Helene Mantineo
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ondine Atwa
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Harshit Bhasin
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yasmine Guedira
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kenneth C Bedi
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Nathan R Tucker
- Departments of Pharmacology and Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative & Precision Cardiology Laboratory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Wang MG, Chen L, Zhang XF. Dual decoding of cell types and gene expression in spatial transcriptomics with PANDA. Nucleic Acids Res 2024; 52:12173-12190. [PMID: 39404057 PMCID: PMC11551751 DOI: 10.1093/nar/gkae876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/24/2024] [Accepted: 09/24/2024] [Indexed: 11/12/2024] Open
Abstract
Sequencing-based spatial transcriptomics technologies have revolutionized our understanding of complex biological systems by enabling transcriptome profiling while preserving spatial context. However, spot-level expression measurements often amalgamate signals from diverse cells, obscuring potential heterogeneity. Existing methods aim to deconvolute spatial transcriptomics data into cell type proportions for each spot using single-cell RNA sequencing references but overlook cell-type-specific gene expression, essential for uncovering intra-type heterogeneity. We present PANDA (ProbAbilistic-based decoNvolution with spot-aDaptive cell type signAtures), a novel method that concurrently deciphers spot-level gene expression into both cell type proportions and cell-type-specific gene expression. PANDA integrates archetypal analysis to capture within-cell-type heterogeneity and dynamically learns cell type signatures for each spot during deconvolution. Simulations demonstrate PANDA's superior performance. Applied to real spatial transcriptomics data from diverse tissues, including tumor, brain, and developing heart, PANDA reconstructs spatial structures and reveals subtle transcriptional variations within specific cell types, offering a comprehensive understanding of tissue dynamics.
Collapse
Affiliation(s)
- Meng-Guo Wang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
| | - Luonan Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai 519031, Guangdong, China
| | - Xiao-Fei Zhang
- School of Mathematics and Statistics, and Hubei Key Lab–Math. Sci., Central China Normal University, Wuhan 430079, Hubei, China
- Key Laboratory of Nonlinear Analysis & Applications (Ministry of Education), Central China Normal University, Wuhan 430079, Hubei, China
| |
Collapse
|
14
|
Lee CYC, McCaffrey J, McGovern D, Clatworthy MR. Profiling immune cell tissue niches in the spatial -omics era. J Allergy Clin Immunol 2024:S0091-6749(24)01178-3. [PMID: 39522655 DOI: 10.1016/j.jaci.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Immune responses require complex, spatially coordinated interactions between immune cells and their tissue environment. For decades, we have imaged tissue sections to visualize a limited number of immune-related macromolecules in situ, functioning as surrogates for cell types or processes of interest. However, this inevitably provides a limited snapshot of the tissue's immune landscape. Recent developments in high-throughput spatial -omics technologies, particularly spatial transcriptomics, and its application to human samples has facilitated a more comprehensive understanding of tissue immunity by mapping fine-grained immune cell states to their precise tissue location while providing contextual information about their immediate cellular and tissue environment. These data provide opportunities to investigate mechanisms underlying the spatial distribution of immune cells and its functional implications, including the identification of immune niches, although the criteria used to define this term have been inconsistent. Here, we review recent technological and analytic advances in multiparameter spatial profiling, focusing on how these methods have generated new insights in translational immunology. We propose a 3-step framework for the definition and characterization of immune niches, which is powerfully facilitated by new spatial profiling methodologies. Finally, we summarize current approaches to analyze adaptive immune repertoires and lymphocyte clonal expansion in a spatially resolved manner.
Collapse
Affiliation(s)
- Colin Y C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James McCaffrey
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Dominic McGovern
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
15
|
Li J, Wiesinger A, Fokkert L, Bakker P, de Vries DK, Tijsen AJ, Pinto YM, Verkerk AO, Christoffels VM, Boink GJJ, Devalla HD. Modeling the atrioventricular conduction axis using human pluripotent stem cell-derived cardiac assembloids. Cell Stem Cell 2024; 31:1667-1684.e6. [PMID: 39260368 PMCID: PMC11546832 DOI: 10.1016/j.stem.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/19/2024] [Accepted: 08/19/2024] [Indexed: 09/13/2024]
Abstract
The atrioventricular (AV) conduction axis provides electrical continuity between the atrial and ventricular chambers. The "nodal" cardiomyocytes populating this region (AV canal in the embryo, AV node from fetal stages onward) propagate impulses slowly, ensuring sequential contraction of the chambers. Dysfunction of AV nodal tissue causes severe disturbances in rhythm and contraction, and human models that capture its salient features are limited. Here, we report an approach for the reproducible generation of AV canal cardiomyocytes (AVCMs) with in vivo-like gene expression and electrophysiological profiles. We created the so-called "assembloids" composed of atrial, AVCM, and ventricular spheroids, which effectively recapitulated unidirectional conduction and the "fast-slow-fast" activation pattern typical for the vertebrate heart. We utilized these systems to reveal intracellular calcium mishandling as the basis of LMNA-associated AV conduction block. In sum, our study introduces novel cell differentiation and tissue construction strategies to facilitate the study of complex disorders affecting heart rhythm.
Collapse
Affiliation(s)
- Jiuru Li
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Alexandra Wiesinger
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Lianne Fokkert
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Priscilla Bakker
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Dylan K de Vries
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Anke J Tijsen
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Yigal M Pinto
- Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Arie O Verkerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Experimental Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands; Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands
| | - Harsha D Devalla
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, Amsterdam 1105 AZ, the Netherlands.
| |
Collapse
|
16
|
Psarras S. The Macrophage-Fibroblast Dipole in the Context of Cardiac Repair and Fibrosis. Biomolecules 2024; 14:1403. [PMID: 39595580 PMCID: PMC11591949 DOI: 10.3390/biom14111403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/21/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Stromal and immune cells and their interactions have gained the attention of cardiology researchers and clinicians in recent years as their contribution in cardiac repair is increasingly recognized. The repair process in the heart is a particularly critical constellation of complex molecular and cellular events and interactions that characteristically fail to ensure adequate recovery following injury, insult, or exposure to stress conditions in this regeneration-hostile organ. The tremendous consequence of this pronounced inability to maintain homeostatic states is being translated in numerous ways promoting progress into heart failure, a deadly, irreversible condition requiring organ transplantation. Fibrosis is in fact a repair response eventually promoting cardiac dysfunction and cardiac fibroblasts are the major cellular players in this process, overproducing collagens and other extracellular matrix components when activated. On the other hand, macrophages may differentially affect fibroblasts and cardiac repair depending on their status and subsets. The opposite interaction is also probable. We discuss here the multifaceted aspects and crosstalk of this cell dipole and the opportunities it may offer for beneficial manipulation approaches that will hopefully lead to progress in heart disease interventions.
Collapse
Affiliation(s)
- Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efesiou 4, 115 27 Athens, Greece
| |
Collapse
|
17
|
Seeler S, Arnarsson K, Dreßen M, Krane M, Doppler SA. Beyond the Heartbeat: Single-Cell Omics Redefining Cardiovascular Research. Curr Cardiol Rep 2024; 26:1183-1196. [PMID: 39158785 DOI: 10.1007/s11886-024-02117-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE OF REVIEW This review aims to explore recent advances in single-cell omics techniques as applied to various regions of the human heart, illuminating cellular diversity, regulatory networks, and disease mechanisms. We examine the contributions of single-cell transcriptomics, genomics, proteomics, epigenomics, and spatial transcriptomics in unraveling the complexity of cardiac tissues. RECENT FINDINGS Recent strides in single-cell omics technologies have revolutionized our understanding of the heart's cellular composition, cell type heterogeneity, and molecular dynamics. These advancements have elucidated pathological conditions as well as the cellular landscape in heart development. We highlight emerging applications of integrated single-cell omics, particularly for cardiac regeneration, disease modeling, and precision medicine, and emphasize the transformative potential of these technologies to advance cardiovascular research and clinical practice.
Collapse
Affiliation(s)
- Sabine Seeler
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Munich, Germany
| | - Kristjan Arnarsson
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Munich, Germany
| | - Martina Dreßen
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Division of Cardiac Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Stefanie A Doppler
- Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Lazarettstr. 36, 80636, Munich, Germany.
- Institute for Translational Cardiac Surgery (INSURE), Department of Cardiovascular Surgery, German Heart Center Munich, School of Medicine and Health, TUM University Hospital, Technical University Munich, Munich, Germany.
| |
Collapse
|
18
|
Oliver AJ, Huang N, Bartolome-Casado R, Li R, Koplev S, Nilsen HR, Moy M, Cakir B, Polanski K, Gudiño V, Melón-Ardanaz E, Sumanaweera D, Dimitrov D, Milchsack LM, FitzPatrick MEB, Provine NM, Boccacino JM, Dann E, Predeus AV, To K, Prete M, Chapman JA, Masi AC, Stephenson E, Engelbert J, Lobentanzer S, Perera S, Richardson L, Kapuge R, Wilbrey-Clark A, Semprich CI, Ellams S, Tudor C, Joseph P, Garrido-Trigo A, Corraliza AM, Oliver TRW, Hook CE, James KR, Mahbubani KT, Saeb-Parsy K, Zilbauer M, Saez-Rodriguez J, Høivik ML, Bækkevold ES, Stewart CJ, Berrington JE, Meyer KB, Klenerman P, Salas A, Haniffa M, Jahnsen FL, Elmentaite R, Teichmann SA. Single-cell integration reveals metaplasia in inflammatory gut diseases. Nature 2024; 635:699-707. [PMID: 39567783 PMCID: PMC11578898 DOI: 10.1038/s41586-024-07571-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 05/15/2024] [Indexed: 11/22/2024]
Abstract
The gastrointestinal tract is a multi-organ system crucial for efficient nutrient uptake and barrier immunity. Advances in genomics and a surge in gastrointestinal diseases1,2 has fuelled efforts to catalogue cells constituting gastrointestinal tissues in health and disease3. Here we present systematic integration of 25 single-cell RNA sequencing datasets spanning the entire healthy gastrointestinal tract in development and in adulthood. We uniformly processed 385 samples from 189 healthy controls using a newly developed automated quality control approach (scAutoQC), leading to a healthy reference atlas with approximately 1.1 million cells and 136 fine-grained cell states. We anchor 12 gastrointestinal disease datasets spanning gastrointestinal cancers, coeliac disease, ulcerative colitis and Crohn's disease to this reference. Utilizing this 1.6 million cell resource (gutcellatlas.org), we discover epithelial cell metaplasia originating from stem cells in intestinal inflammatory diseases with transcriptional similarity to cells found in pyloric and Brunner's glands. Although previously linked to mucosal healing4, we now implicate pyloric gland metaplastic cells in inflammation through recruitment of immune cells including T cells and neutrophils. Overall, we describe inflammation-induced changes in stem cells that alter mucosal tissue architecture and promote further inflammation, a concept applicable to other tissues and diseases.
Collapse
Affiliation(s)
- Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Raquel Bartolome-Casado
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, US
| | - Simon Koplev
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Hogne R Nilsen
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Madelyn Moy
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Victoria Gudiño
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Elisa Melón-Ardanaz
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | | | - Daniel Dimitrov
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | | | - Michael E B FitzPatrick
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nicholas M Provine
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Emma Dann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Ken To
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Jonathan A Chapman
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Andrea C Masi
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Emily Stephenson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Justin Engelbert
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Sebastian Lobentanzer
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Laura Richardson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Rakeshlal Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | | | - Sophie Ellams
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Catherine Tudor
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Alba Garrido-Trigo
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Ana M Corraliza
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Thomas R W Oliver
- Department of Histopathology and Cytology, Cambridge University Hospitals, Cambridge, UK
| | | | - Kylie R James
- Translational Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
- Department of Haematology, Cambridge Stem Cell Institute, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Biorepository for Translational Medicine, Cambridge NIHR Biomedical Research Centre, Cambridge, UK
| | - Matthias Zilbauer
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- University Department of Paediatrics, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals, Cambridge, UK
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Bioquant, Heidelberg, Germany
| | - Marte Lie Høivik
- Department of Gastroenterology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen S Bækkevold
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | | | - Janet E Berrington
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Azucena Salas
- Inflammatory Bowel Disease Unit, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and National Institute for Health Research (NIHR) Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Frode L Jahnsen
- Department of Pathology, University of Oslo and Oslo University Hospital-Rikshospitalet, Oslo, Norway
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Cambridge, UK.
- Theory of Condensed Matter, Cavendish Laboratory/Department of Physics, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- CIFAR Macmillan Multi-scale Human Program, CIFAR, Toronto, Ontario, Canada.
| |
Collapse
|
19
|
To K, Fei L, Pett JP, Roberts K, Blain R, Polański K, Li T, Yayon N, He P, Xu C, Cranley J, Moy M, Li R, Kanemaru K, Huang N, Megas S, Richardson L, Kapuge R, Perera S, Tuck E, Wilbrey-Clark A, Mulas I, Memi F, Cakir B, Predeus AV, Horsfall D, Murray S, Prete M, Mazin P, He X, Meyer KB, Haniffa M, Barker RA, Bayraktar O, Chédotal A, Buckley CD, Teichmann SA. A multi-omic atlas of human embryonic skeletal development. Nature 2024; 635:657-667. [PMID: 39567793 PMCID: PMC11578895 DOI: 10.1038/s41586-024-08189-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/09/2024] [Indexed: 11/22/2024]
Abstract
Human embryonic bone and joint formation is determined by coordinated differentiation of progenitors in the nascent skeleton. The cell states, epigenetic processes and key regulatory factors that underlie lineage commitment of these cells remain elusive. Here we applied paired transcriptional and epigenetic profiling of approximately 336,000 nucleus droplets and spatial transcriptomics to establish a multi-omic atlas of human embryonic joint and cranium development between 5 and 11 weeks after conception. Using combined modelling of transcriptional and epigenetic data, we characterized regionally distinct limb and cranial osteoprogenitor trajectories across the embryonic skeleton and further described regulatory networks that govern intramembranous and endochondral ossification. Spatial localization of cell clusters in our in situ sequencing data using a new tool, ISS-Patcher, revealed mechanisms of progenitor zonation during bone and joint formation. Through trajectory analysis, we predicted potential non-canonical cellular origins for human chondrocytes from Schwann cells. We also introduce SNP2Cell, a tool to link cell-type-specific regulatory networks to polygenic traits such as osteoarthritis. Using osteolineage trajectories characterized here, we simulated in silico perturbations of genes that cause monogenic craniosynostosis and implicate potential cell states and disease mechanisms. This work forms a detailed and dynamic regulatory atlas of bone and cartilage maturation and advances our fundamental understanding of cell-fate determination in human skeletal development.
Collapse
Affiliation(s)
- Ken To
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Surgery, University of Cambridge, Cambridge, UK
| | - Lijiang Fei
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - J Patrick Pett
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Kenny Roberts
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Raphael Blain
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
| | | | - Tong Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Nadav Yayon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridge, UK
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - James Cranley
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Madelyn Moy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Ruoyan Li
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Stathis Megas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK
| | | | - Rakesh Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Shani Perera
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Elizabeth Tuck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Ilaria Mulas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Batuhan Cakir
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - David Horsfall
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Simon Murray
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Pavel Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Xiaoling He
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Newcastle University, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
- Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Roger A Barker
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Omer Bayraktar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Alain Chédotal
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, France
- Institut de Pathologie, Groupe Hospitalier Est, Hospices Civils de Lyon, Lyon, France
- University Claude Bernard Lyon 1, MeLiS, CNRS UMR5284, INSERM U1314, Lyon, France
| | | | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Cambridge Centre for AI in Medicine, Department of Applied Mathematics and Theoretical Physics, Cambridge, UK.
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK.
- CIFAR Macmillan Multi-scale Human Programme, CIFAR, Toronto, Canada.
| |
Collapse
|
20
|
Lubberding AF, Veedfald S, Achter JS, Nissen SD, Soattin L, Sorrentino A, Vega ET, Linz B, Eggertsen CHE, Mulvey J, Toräng S, Larsen SA, Nissen A, Petersen LG, Bilir SE, Bentzen BH, Rosenkilde MM, Hartmann B, Lilleør TNB, Qazi S, Møller CH, Tfelt-Hansen J, Sattler SM, Jespersen T, Holst JJ, Lundby A. Glucagon-like peptide-1 increases heart rate by a direct action on the sinus node. Cardiovasc Res 2024; 120:1427-1441. [PMID: 38832935 PMCID: PMC11472427 DOI: 10.1093/cvr/cvae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 02/01/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024] Open
Abstract
AIMS Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are increasingly used to treat type 2 diabetes and obesity. Albeit cardiovascular outcomes generally improve, treatment with GLP-1 RAs is associated with increased heart rate, the mechanism of which is unclear. METHODS AND RESULTS We employed a large animal model, the female landrace pig, and used multiple in vivo and ex vivo approaches including pharmacological challenges, electrophysiology, and high-resolution mass spectrometry to explore how GLP-1 elicits an increase in heart rate. In anaesthetized pigs, neither cervical vagotomy, adrenergic blockers (alpha, beta, or combined alpha-beta blockade), ganglionic blockade (hexamethonium), nor inhibition of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (ivabradine) abolished the marked chronotropic effect of GLP-1. GLP-1 administration to isolated perfused pig hearts also increased heart rate, which was abolished by GLP-1 receptor blockade. Electrophysiological characterization of GLP-1 effects in vivo and in isolated perfused hearts localized electrical modulation to the atria and conduction system. In isolated sinus nodes, GLP-1 administration shortened the action potential cycle length of pacemaker cells and shifted the site of earliest activation. The effect was independent of HCN blockade. Collectively, these data support a direct effect of GLP-1 on GLP-1 receptors within the heart. Consistently, single nucleus RNA sequencing showed GLP-1 receptor expression in porcine pacemaker cells. Quantitative phosphoproteomics analyses of sinus node samples revealed that GLP-1 administration leads to phosphorylation changes of calcium cycling proteins of the sarcoplasmic reticulum, known to regulate heart rate. CONCLUSION GLP-1 has direct chronotropic effects on the heart mediated by GLP-1 receptors in pacemaker cells of the sinus node, inducing changes in action potential morphology and the leading pacemaker site through a calcium signalling response characterized by PKA-dependent phosphorylation of Ca2+ cycling proteins involved in pacemaking. Targeting the pacemaker calcium clock may be a strategy to lower heart rate in people treated with GLP-1 RAs.
Collapse
Affiliation(s)
- Anniek Frederike Lubberding
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Simon Veedfald
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Sarah Dalgas Nissen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Luca Soattin
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Benedikt Linz
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Caroline Harriet Eggert Eggertsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - John Mulvey
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Signe Toräng
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sara Agnete Larsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nissen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lonnie Grove Petersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Secil Erbil Bilir
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Bo Hjorth Bentzen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | | | - Saddiq Qazi
- Department of Cardiothoracic Surgery, Rigshospitalet, Copenhagen, Denmark
| | | | - Jacob Tfelt-Hansen
- Department of Cardiology, Heart Centre, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Forensic Medicine, Faculty of Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefan Michael Sattler
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Department of Cardiology, Herlev and Gentofte University Hospital, Hellerup, Denmark
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
21
|
Easter QT, Alvarado-Martinez Z, Kunz M, Matuck BF, Rupp BT, Weaver T, Ren Z, Tata A, Caballero-Perez J, Oscarson N, Hasuike A, Ghodke AN, Kimple AJ, Tata PR, Randell SH, Koo H, Ko KI, Byrd KM. Polybacterial Intracellular Macromolecules Shape Single-Cell Epikine Profiles in Upper Airway Mucosa. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.08.617279. [PMID: 39416216 PMCID: PMC11482982 DOI: 10.1101/2024.10.08.617279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The upper airway, particularly the nasal and oral mucosal epithelium, serves as a primary barrier for microbial interactions throughout life. Specialized niches like the anterior nares and the tooth are especially susceptible to dysbiosis and chronic inflammatory diseases. To investigate host-microbial interactions in mucosal epithelial cell types, we reanalyzed our single-cell RNA sequencing atlas of human oral mucosa, identifying polybacterial signatures (20% Gram-positive, 80% Gram-negative) within both epithelial- and stromal-resident cells. This analysis revealed unique responses of bacterial-associated epithelia when compared to two inflammatory disease states of mucosa. Single-cell RNA sequencing, in situ hybridization, and immunohistochemistry detected numerous persistent macromolecules from Gram-positive and Gram-negative bacteria within human oral keratinocytes (HOKs), including bacterial rRNA, mRNA and glycolipids. Epithelial cells with higher concentrations of 16S rRNA and glycolipids exhibited enhanced receptor-ligand signaling in vivo. HOKs with a spectrum of polybacterial intracellular macromolecular (PIM) concentrations were challenged with purified exogenous lipopolysaccharide, resulting in the synergistic upregulation of select innate (CXCL8, TNFSF15) and adaptive (CXCL17, CCL28) epikines. Notably, endogenous lipoteichoic acid, rather than lipopolysaccharide, directly correlated with epikine expression in vitro and in vivo. Application of the Drug2Cell algorithm to health and inflammatory disease data suggested altered drug efficacy predictions based on PIM detection. Our findings demonstrate that PIMs persist within mucosal epithelial cells at variable concentrations, linearly driving single-cell effector cytokine expression and influencing drug responses, underscoring the importance of understanding host-microbe interactions and the implications of PIMs on cell behavior in health and disease at single-cell resolution.
Collapse
Affiliation(s)
- Quinn T Easter
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Zabdiel Alvarado-Martinez
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Meik Kunz
- The Bioinformatics CRO, Orlando, FL, USA
| | - Bruno Fernandes Matuck
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Brittany T Rupp
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Theresa Weaver
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Zhi Ren
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | | | - Nick Oscarson
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
| | - Akira Hasuike
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
- Department of Periodontology, Nihon University School of Dentistry, Tokyo, JP
| | - Ameer N Ghodke
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Adam J Kimple
- Department of Otolaryngology-Head and Neck Surgery, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Purushothama R Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Hyun Koo
- Biofilm Research Laboratories, Center for Innovation & Precision Dentistry, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kang I Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kevin M Byrd
- Lab of Oral & Craniofacial Innovation (LOCI), ADA Science & Research Institute, Gaithersburg, MD, USA
- UNC Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
22
|
Gómez-Del Arco P, Isern J, Jimenez-Carretero D, López-Maderuelo D, Piñeiro-Sabarís R, El Abdellaoui-Soussi F, Torroja C, Vera-Pedrosa ML, Grima-Terrén M, Benguria A, Simón-Chica A, Queiro-Palou A, Dopazo A, Sánchez-Cabo F, Jalife J, de la Pompa JL, Filgueiras-Rama D, Muñoz-Cánoves P, Redondo JM. The G4 resolvase Dhx36 modulates cardiomyocyte differentiation and ventricular conduction system development. Nat Commun 2024; 15:8602. [PMID: 39366945 PMCID: PMC11452623 DOI: 10.1038/s41467-024-52809-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/19/2024] [Indexed: 10/06/2024] Open
Abstract
Extensive genetic studies have elucidated cardiomyocyte differentiation and associated gene networks using single-cell RNA-seq, yet the intricate transcriptional mechanisms governing cardiac conduction system (CCS) development and working cardiomyocyte differentiation remain largely unexplored. Here we show that mice deleted for Dhx36 (encoding the Dhx36 helicase) in the embryonic or neonatal heart develop overt dilated cardiomyopathy, surface ECG alterations related to cardiac impulse propagation, and (in the embryonic heart) a lack of a ventricular conduction system (VCS). Heart snRNA-seq and snATAC-seq reveal the role of Dhx36 in CCS development and in the differentiation of working cardiomyocytes. Dhx36 deficiency directly influences cardiomyocyte gene networks by disrupting the resolution of promoter G-quadruplexes in key cardiac genes, impacting cardiomyocyte differentiation and CCS morphogenesis, and ultimately leading to dilated cardiomyopathy and atrioventricular block. These findings further identify crucial genes and pathways that regulate the development and function of the VCS/Purkinje fiber (PF) network.
Collapse
Affiliation(s)
- Pablo Gómez-Del Arco
- Institute for Rare Diseases Research, Instituto de Salud Carlos III (ISCIII). Majadahonda, Madrid, Spain.
- Gene Regulation in Cardiovascular Remodelling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Joan Isern
- Altos Labs, Inc., San Diego Institute of Science, San Diego, CA, USA
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Daniel Jimenez-Carretero
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Dolores López-Maderuelo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Microscopy and Dynamic Imaging Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Rebeca Piñeiro-Sabarís
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Fadoua El Abdellaoui-Soussi
- Institute for Rare Diseases Research, Instituto de Salud Carlos III (ISCIII). Majadahonda, Madrid, Spain
- Gene Regulation in Cardiovascular Remodelling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Center for Stem Cells and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carlos Torroja
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María Linarejos Vera-Pedrosa
- Cardiac Arrhythmia Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Mercedes Grima-Terrén
- Altos Labs, Inc., San Diego Institute of Science, San Diego, CA, USA
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Alberto Benguria
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ana Simón-Chica
- Novel Arrhythmogenic Mechanisms Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Antonio Queiro-Palou
- Institute for Rare Diseases Research, Instituto de Salud Carlos III (ISCIII). Majadahonda, Madrid, Spain
- Gene Regulation in Cardiovascular Remodelling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Ana Dopazo
- Genomics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José Jalife
- Cardiac Arrhythmia Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- University of Michigan, Ann Arbor, MI, USA
| | - José Luis de la Pompa
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Intercellular Signaling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - David Filgueiras-Rama
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Novel Arrhythmogenic Mechanisms Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Cardiovascular Institute, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Pura Muñoz-Cánoves
- Altos Labs, Inc., San Diego Institute of Science, San Diego, CA, USA.
- Tissue Regeneration Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Department of Experimental & Health Sciences, University Pompeu Fabra (UPF)/CIBERNED, Barcelona, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain.
| | - Juan Miguel Redondo
- Gene Regulation in Cardiovascular Remodelling and Inflammation Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
- Cell-Cell Communication & Inflammation Unit, Centro de Biología Molecular Severo Ochoa (CBMSO), Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
23
|
Singh H, Sanghvi SK. Top stories on intracellular potassium channels in cardiac arrhythmia. Heart Rhythm 2024; 21:2074-2075. [PMID: 39357933 DOI: 10.1016/j.hrthm.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 10/04/2024]
Affiliation(s)
- Harpreet Singh
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio; Department of Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio.
| | - Shridhar Kiran Sanghvi
- Department of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio; Department of Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
24
|
Lukovic D, Gyöngyösi M, Pavo IJ, Mester-Tonczar J, Einzinger P, Zlabinger K, Kastner N, Spannbauer A, Traxler D, Pavo N, Goliasch G, Pils D, Jakab A, Szankai Z, Michel-Behnke I, Zhang L, Devaux Y, Graf S, Beitzke D, Winkler J. Increased [ 18F]FDG uptake in the infarcted myocardial area displayed by combined PET/CMR correlates with snRNA-seq-detected inflammatory cell invasion. Basic Res Cardiol 2024; 119:807-829. [PMID: 38922408 PMCID: PMC11461641 DOI: 10.1007/s00395-024-01064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 06/17/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Combined [18F]FDG PET-cardiac MRI imaging (PET/CMR) is a useful tool to assess myocardial viability and cardiac function in patients with acute myocardial infarction (AMI). Here, we evaluated the prognostic value of PET/CMR in a porcine closed-chest reperfused AMI (rAMI) model. Late gadolinium enhancement by PET/CMR imaging displayed tracer uptake defect at the infarction site by 3 days after the rAMI in the majority of the animals (group Match, n = 28). Increased [18F]FDG uptake at the infarcted area (metabolism/contractility mismatch) with reduced tracer uptake in the remote viable myocardium (group Mismatch, n = 12) 3 days after rAMI was observed in the animals with larger infarct size and worse left ventricular ejection fraction (LVEF) (34 ± 8.7 vs 42.0 ± 5.2%), with lower LVEF also at the 1-month follow-up (35.8 ± 9.5 vs 43.0 ± 6.3%). Transcriptome analyses by bulk and single-nuclei RNA sequencing of the infarcted myocardium and border zones (n = 3 of each group, and 3 sham-operated controls) revealed a strong inflammatory response with infiltration of monocytes and macrophages in the infarcted and border areas in Mismatch animals. Our data indicate a high prognostic relevance of combined PET/MRI in the subacute phase of rAMI for subsequent impairment of heart function and underline the adverse effects of an excessive activation of the innate immune system in the initial phase after rAMI.
Collapse
Affiliation(s)
- Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria.
| | - Imre J Pavo
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040, Vienna, Austria
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Noemi Pavo
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Georg Goliasch
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietmar Pils
- Division of General Surgery, Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Andras Jakab
- Center for MR-Research, University Children's Hospital Zurich, Zurich, Switzerland
| | | | - Ina Michel-Behnke
- Division of Pediatric Cardiology, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Lu Zhang
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Senta Graf
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| | - Dietrich Beitzke
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
25
|
Zhu J, Pang K, Hu B, He R, Wang N, Jiang Z, Ji P, Zhao F. Custom microfluidic chip design enables cost-effective three-dimensional spatiotemporal transcriptomics with a wide field of view. Nat Genet 2024; 56:2259-2270. [PMID: 39256584 PMCID: PMC11525186 DOI: 10.1038/s41588-024-01906-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024]
Abstract
Spatial transcriptomic techniques offer unprecedented insights into the molecular organization of complex tissues. However, integrating cost-effectiveness, high throughput, a wide field of view and compatibility with three-dimensional (3D) volumes has been challenging. Here we introduce microfluidics-assisted grid chips for spatial transcriptome sequencing (MAGIC-seq), a new method that combines carbodiimide chemistry, spatial combinatorial indexing and innovative microfluidics design. This technique allows sensitive and reproducible profiling of diverse tissue types, achieving an eightfold increase in throughput, minimal cost and reduced batch effects. MAGIC-seq breaks conventional microfluidics limits by enhancing barcoding efficiency and enables analysis of whole postnatal mouse sections, providing comprehensive cellular structure elucidation at near single-cell resolution, uncovering transcriptional variations and dynamic trajectories of mouse organogenesis. Our 3D transcriptomic atlas of the developing mouse brain, consisting of 93 sections, reveals the molecular and cellular landscape, serving as a valuable resource for neuroscience and developmental biology. Overall, MAGIC-seq is a high-throughput, cost-effective, large field of view and versatile method for spatial transcriptomic studies.
Collapse
Affiliation(s)
- Junjie Zhu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Kun Pang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Beiyu Hu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Ruiqiao He
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zewen Jiang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peifeng Ji
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
26
|
Wu X, Yang X, Dai Y, Zhao Z, Zhu J, Guo H, Yang R. Single-cell sequencing to multi-omics: technologies and applications. Biomark Res 2024; 12:110. [PMID: 39334490 PMCID: PMC11438019 DOI: 10.1186/s40364-024-00643-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/17/2024] [Indexed: 09/30/2024] Open
Abstract
Cells, as the fundamental units of life, contain multidimensional spatiotemporal information. Single-cell RNA sequencing (scRNA-seq) is revolutionizing biomedical science by analyzing cellular state and intercellular heterogeneity. Undoubtedly, single-cell transcriptomics has emerged as one of the most vibrant research fields today. With the optimization and innovation of single-cell sequencing technologies, the intricate multidimensional details concealed within cells are gradually unveiled. The combination of scRNA-seq and other multi-omics is at the forefront of the single-cell field. This involves simultaneously measuring various omics data within individual cells, expanding our understanding across a broader spectrum of dimensions. Single-cell multi-omics precisely captures the multidimensional aspects of single-cell transcriptomes, immune repertoire, spatial information, temporal information, epitopes, and other omics in diverse spatiotemporal contexts. In addition to depicting the cell atlas of normal or diseased tissues, it also provides a cornerstone for studying cell differentiation and development patterns, disease heterogeneity, drug resistance mechanisms, and treatment strategies. Herein, we review traditional single-cell sequencing technologies and outline the latest advancements in single-cell multi-omics. We summarize the current status and challenges of applying single-cell multi-omics technologies to biological research and clinical applications. Finally, we discuss the limitations and challenges of single-cell multi-omics and potential strategies to address them.
Collapse
Affiliation(s)
- Xiangyu Wu
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Xin Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Yunhan Dai
- Medical School, Nanjing University, Nanjing, China
| | - Zihan Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China
| | - Junmeng Zhu
- Department of Oncology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| | - Rong Yang
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
27
|
Yin Y, Chen Y, Xu J, Liu B, Zhao Y, Tan X, Xiao M, Zhou Y, Zheng X, Xu Y, Han Z, Hu H, Li Z, Ou N, Lian W, Li Y, Su Z, Dai Y, Tang Y, Zhao L. Molecular and spatial signatures of human and rat corpus cavernosum physiopathological processes at single-cell resolution. Cell Rep 2024; 43:114760. [PMID: 39299236 DOI: 10.1016/j.celrep.2024.114760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/15/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
The composition and cellular heterogeneity of the corpus cavernosum (CC) microenvironment have been characterized, but the spatial heterogeneity at the molecular level remains unexplored. In this study, we integrate single-cell RNA sequencing (scRNA-seq) and spatial transcriptome sequencing to comprehensively chart the spatial cellular landscape of the human and rat CC under normal and disease conditions. We observe differences in the proportions of cell subtypes and marker genes between humans and rats. Based on the analysis of the fibroblast (FB) niche, we also find that the enrichment scores of mechanical force signaling vary across different regions and correlate with the spatial distribution of FB subtypes. In vitro, the soft and hard extracellular matrix (ECM) induces the differentiation of FBs into apolipoprotein (APO)+ FBs and cartilage oligomeric matrix protein (COMP)+ FBs, respectively. In summary, our study provides a cross-species and physiopathological transcriptomic atlas of the CC, contributing to a further understanding of the molecular anatomy and regulation of penile erection.
Collapse
Affiliation(s)
- Yinghao Yin
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yuzhuo Chen
- Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jiarong Xu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Biao Liu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yifan Zhao
- Department of Biostatistics & Informatics, Colorado School of Public Health, Aurora, CO, USA
| | - Xiaoli Tan
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Ming Xiao
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yihong Zhou
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaoping Zheng
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yanghua Xu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhitao Han
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Hongji Hu
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zitaiyu Li
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Ningjing Ou
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Wenfei Lian
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yawei Li
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhongzhen Su
- Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yingbo Dai
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Yuxin Tang
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| | - Liangyu Zhao
- Department of Urology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China; Department of Interventional Medicine, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China.
| |
Collapse
|
28
|
Yasumizu Y, Kinoshita M, Zhang MJ, Motooka D, Suzuki K, Nojima S, Koizumi N, Okuzaki D, Funaki S, Shintani Y, Ohkura N, Morii E, Okuno T, Mochizuki H. Spatial transcriptomics elucidates medulla niche supporting germinal center response in myasthenia gravis-associated thymoma. Cell Rep 2024; 43:114677. [PMID: 39180749 DOI: 10.1016/j.celrep.2024.114677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/26/2024] Open
Abstract
Myasthenia gravis (MG) is etiologically associated with thymus abnormalities, but its pathology in the thymus remains unclear. In this study, we attempt to narrow down the features associated with MG using spatial transcriptome analysis of thymoma and thymic hyperplasia samples. We find that the majority of thymomas are constituted by the cortical region. However, the small medullary region is enlarged in seropositive thymomas and contains polygenic enrichment and MG-specific germinal center structures. Neuromuscular medullary thymic epithelial cells, previously identified as MG-specific autoantigen-producing cells, are enriched in the cortico-medullary junction. The medulla is characterized by a specific chemokine pattern and immune cell composition, including migratory dendritic cells and effector regulatory T cells. Similar germinal center structures and immune microenvironments are also observed in the thymic hyperplasia medulla. This study shows that the medulla and junction areas are linked to MG pathology and provides insights into future MG research.
Collapse
Affiliation(s)
- Yoshiaki Yasumizu
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
| | - Makoto Kinoshita
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Martin Jinye Zhang
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Daisuke Motooka
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Koichiro Suzuki
- BIKEN-RIMD NGS Laboratory, Research Institute for Microbial Diseases, Osaka University, Suita, Japan; Biomedical Science Center, The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Suita, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naoshi Koizumi
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Daisuke Okuzaki
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan; Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Soichiro Funaki
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yasushi Shintani
- Department of General Thoracic Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Naganari Ohkura
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan; Department of Frontier Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Eiichi Morii
- Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tatsusada Okuno
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan.
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Osaka, Japan
| |
Collapse
|
29
|
Leblanc FJA, Jin X, Kang K, Lee CJM, Xu J, Xuan L, Ma W, Belhaj H, Benzaki M, Mehta N, Foo RSY, Reilly S, Anene-Nzelu CG, Pan Z, Nattel S, Yang B, Lettre G. Atrial fibrillation variant-to-gene prioritization through cross-ancestry eQTL and single-nucleus multiomic analyses. iScience 2024; 27:110660. [PMID: 39262787 PMCID: PMC11388022 DOI: 10.1016/j.isci.2024.110660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/28/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Atrial fibrillation (AF) is the most common arrhythmia in the world. Human genetics can provide strong AF therapeutic candidates, but the identification of the causal genes and their functions remains challenging. Here, we applied an AF fine-mapping strategy that leverages results from a previously published cross-ancestry genome-wide association study (GWAS), expression quantitative trait loci (eQTLs) from left atrial appendages (LAAs) obtained from two cohorts with distinct ancestry, and a paired RNA sequencing (RNA-seq) and ATAC sequencing (ATAC-seq) LAA single-nucleus assay (sn-multiome). At nine AF loci, our co-localization and fine-mapping analyses implicated 14 genes. Data integration identified several candidate causal AF variants, including rs7612445 at GNB4 and rs242557 at MAPT. Finally, we showed that the repression of the strongest AF-associated eQTL gene, LINC01629, in human embryonic stem cell-derived cardiomyocytes using CRISPR inhibition results in the dysregulation of pathways linked to genes involved in the development of atrial tissue and the cardiac conduction system.
Collapse
Affiliation(s)
- Francis J A Leblanc
- Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Xuexin Jin
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
- Department of Cardiology, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Kai Kang
- Department of Cardiovascular Surgery, The First Affiliated Hospital, Harbin Medical University, Harbin 150001, China
| | - Chang Jie Mick Lee
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Juan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Lina Xuan
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Wenbo Ma
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | | | - Marouane Benzaki
- Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Neelam Mehta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Roger Sik Yin Foo
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Chukwuemeka George Anene-Nzelu
- Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhenwei Pan
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Stanley Nattel
- Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
- IHU Liryc and Fondation Bordeaux Université, Bordeaux, France
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Baofeng Yang
- Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Disease, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Guillaume Lettre
- Montreal Heart Institute, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
30
|
Engesser J, Khatri R, Schaub DP, Zhao Y, Paust HJ, Sultana Z, Asada N, Riedel JH, Sivayoganathan V, Peters A, Kaffke A, Jauch-Speer SL, Goldbeck-Strieder T, Puelles VG, Wenzel UO, Steinmetz OM, Hoxha E, Turner JE, Mittrücker HW, Wiech T, Huber TB, Bonn S, Krebs CF, Panzer U. Immune profiling-based targeting of pathogenic T cells with ustekinumab in ANCA-associated glomerulonephritis. Nat Commun 2024; 15:8220. [PMID: 39300109 DOI: 10.1038/s41467-024-52525-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis is a life-threatening autoimmune disease that often results in kidney failure caused by crescentic glomerulonephritis (GN). To date, treatment of most patients with ANCA-GN relies on non-specific immunosuppressive agents, which may have serious adverse effects and be only partially effective. Here, using spatial and single-cell transcriptome analysis, we characterize inflammatory niches in kidney samples from 34 patients with ANCA-GN and identify proinflammatory, cytokine-producing CD4+ and CD8+ T cells as a pathogenic signature. We then utilize these transcriptomic profiles for digital pharmacology and identify ustekinumab, a monoclonal antibody targeting IL-12 and IL-23, as the strongest therapeutic drug to use. Moreover, four patients with relapsing ANCA-GN are treated with ustekinumab in combination with low-dose cyclophosphamide and steroids, with ustekinumab given subcutaneously (90 mg) at weeks 0, 4, 12, and 24. Patients are followed up for 26 weeks to find this treatment well-tolerated and inducing clinical responses, including improved kidney function and Birmingham Vasculitis Activity Score, in all ANCA-GN patients. Our findings thus suggest that targeting of pathogenic T cells in ANCA-GN patients with ustekinumab might represent a potential approach and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Jonas Engesser
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Khatri
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Darius P Schaub
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Yu Zhao
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Hans-Joachim Paust
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Zeba Sultana
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany
| | - Nariaki Asada
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Hendrik Riedel
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Varshi Sivayoganathan
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anett Peters
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kaffke
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Victor G Puelles
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich O Wenzel
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver M Steinmetz
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elion Hoxha
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Willi Mittrücker
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute for Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Pathology, Division of Nephropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Bonn
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Institute of Medical Systems Biology, Center for Biomedical AI, Center for Molecular Neurobiology Hamburg, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Christian F Krebs
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Ulf Panzer
- Department of Medicine III, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
31
|
Jin S, Plikus MV, Nie Q. CellChat for systematic analysis of cell-cell communication from single-cell transcriptomics. Nat Protoc 2024:10.1038/s41596-024-01045-4. [PMID: 39289562 DOI: 10.1038/s41596-024-01045-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 06/27/2024] [Indexed: 09/19/2024]
Abstract
Recent advances in single-cell sequencing technologies offer an opportunity to explore cell-cell communication in tissues systematically and with reduced bias. A key challenge is integrating known molecular interactions and measurements into a framework to identify and analyze complex cell-cell communication networks. Previously, we developed a computational tool, named CellChat, that infers and analyzes cell-cell communication networks from single-cell transcriptomic data within an easily interpretable framework. CellChat quantifies the signaling communication probability between two cell groups using a simplified mass-action-based model, which incorporates the core interaction between ligands and receptors with multisubunit structure along with modulation by cofactors. Importantly, CellChat performs a systematic and comparative analysis of cell-cell communication using a variety of quantitative metrics and machine-learning approaches. CellChat v2 is an updated version that includes additional comparison functionalities, an expanded database of ligand-receptor pairs along with rich functional annotations, and an Interactive CellChat Explorer. Here we provide a step-by-step protocol for using CellChat v2 on single-cell transcriptomic data, including inference and analysis of cell-cell communication from one dataset and identification of altered intercellular communication, signals and cell populations from different datasets across biological conditions. The R implementation of CellChat v2 toolkit and its tutorials together with the graphic outputs are available at https://github.com/jinworks/CellChat . This protocol typically takes ~5 min depending on dataset size and requires a basic understanding of R and single-cell data analysis but no specialized bioinformatics training for its implementation.
Collapse
Affiliation(s)
- Suoqin Jin
- School of Mathematics and Statistics, Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China.
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
32
|
Yoshida S, Yoshida T, Inukai K, Kato K, Yura Y, Hattori T, Enomoto A, Ohashi K, Okumura T, Ouchi N, Kawase H, Wettschureck N, Offermanns S, Murohara T, Takefuji M. Protein kinase N promotes cardiac fibrosis in heart failure by fibroblast-to-myofibroblast conversion. Nat Commun 2024; 15:7638. [PMID: 39266515 PMCID: PMC11392935 DOI: 10.1038/s41467-024-52068-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 08/26/2024] [Indexed: 09/14/2024] Open
Abstract
Chronic fibrotic tissue disrupts various organ functions. Despite significant advances in therapies, mortality and morbidity due to heart failure remain high, resulting in poor quality of life. Beyond the cardiomyocyte-centric view of heart failure, it is now accepted that alterations in the interstitial extracellular matrix (ECM) also play a major role in the development of heart failure. Here, we show that protein kinase N (PKN) is expressed in cardiac fibroblasts. Furthermore, PKN mediates the conversion of fibroblasts into myofibroblasts, which plays a central role in secreting large amounts of ECM proteins via p38 phosphorylation signaling. Fibroblast-specific deletion of PKN led to a reduction of myocardial fibrotic changes and cardiac dysfunction in mice models of ischemia-reperfusion or heart failure with preserved ejection fraction. Our results indicate that PKN is a therapeutic target for cardiac fibrosis in heart failure.
Collapse
Affiliation(s)
- Satoya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Tatsuya Yoshida
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Kohei Inukai
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Katsuhiro Kato
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Yoshimitsu Yura
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Tomoki Hattori
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University School of Medicine, Nagoya, Japan
| | - Koji Ohashi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Takahiro Okumura
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Molecular Medicine and Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Haruya Kawase
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Nina Wettschureck
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan
| | - Mikito Takefuji
- Department of Cardiology, Nagoya University School of Medicine, Nagoya, Japan.
| |
Collapse
|
33
|
Ambroise R, Takasugi P, Liu J, Qian L. Direct Cardiac Reprogramming in the Age of Computational Biology. J Cardiovasc Dev Dis 2024; 11:273. [PMID: 39330331 PMCID: PMC11432431 DOI: 10.3390/jcdd11090273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/28/2024] Open
Abstract
Heart disease continues to be one of the most fatal conditions worldwide. This is in part due to the maladaptive remodeling process by which ischemic cardiac tissue is replaced with a fibrotic scar. Direct cardiac reprogramming presents a unique solution for restoring injured cardiac tissue through the direct conversion of fibroblasts into induced cardiomyocytes, bypassing the transition through a pluripotent state. Since its inception in 2010, direct cardiac reprogramming using the transcription factors Gata4, Mef2c, and Tbx5 has revolutionized the field of cardiac regenerative medicine. Just over a decade later, the field has rapidly evolved through the expansion of identified molecular and genetic factors that can be used to optimize reprogramming efficiency. The integration of computational tools into the study of direct cardiac reprogramming has been critical to this progress. Advancements in transcriptomics, epigenetics, proteomics, genome editing, and machine learning have not only enhanced our understanding of the underlying mechanisms driving this cell fate transition, but have also driven innovations that push direct cardiac reprogramming closer to clinical application. This review article explores how these computational advancements have impacted and continue to shape the field of direct cardiac reprogramming.
Collapse
Affiliation(s)
- Rachelle Ambroise
- Department of Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Paige Takasugi
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
34
|
Liu Y, Sun X, Yuan M, Yu Z, Hou Q, Jia Z, Xu T, Xu B. Enhanced lipid metabolism reprogramming in CHF rats through IL-6-mediated cardiac glial cell modulation by digilanid C and electroacupuncture stimulation combination. Front Cell Dev Biol 2024; 12:1424395. [PMID: 39291267 PMCID: PMC11405320 DOI: 10.3389/fcell.2024.1424395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Background Cardiac lipid metabolism reprogramming is recognized as a critical pathological factor in the progression of chronic heart failure (CHF). The therapeutic potential of digilanid C and electroacupuncture stimulation (ES) in enhancing lipid metabolism and cardiac function has been established. However, the optimal synergistic regulatory strategies of these interventions on cardiac lipid metabolism have yet to be elucidated. Methods This study aimed to comprehensively evaluate the impact of a digilanid C-ES combination on cardiac steatosis remodeling in CHF. Assessments were conducted across various dimensions, including myocardial oxygen consumption, mitochondrial function, and lipid metabolism. Additionally, we sought to uncover the underlying neuromolecular mechanisms. Results Our findings, at both molecular and morphological levels, indicated that the synergistic application of digilanid C and ES significantly inhibited myocardial fibrosis and steatosis. This combination therapy facilitated the repair of cardiac neuro-vascular uncoupling and induced a reprogramming of lipid metabolism. Notably, the digilanid C-ES combination ameliorated cardiomyocyte apoptosis and enhanced mitochondrial biogenesis in CHF, leading to a restructured energy supply pattern. Cardiac immunofluorescence analyses revealed the aggregation of cardiac glial cells (CGCs) at sites of abnormal neurovascular coupling, a response to cardiac lipid degeneration. This was accompanied by a marked reduction in the abnormally elevated expression of interleukin 6 (IL-6) and glutamatergic signaling, which correlated with the severity of cardiac steatosis and the aberrant activation of CGCs. The combined therapy was found to activate the Janus kinase 1 (JAK1)/signal transducer and activator of transcription 3 (STAT3) pathway, effectively attenuated lipid accumulation and over-recruitment of CGCs and deprivation of glutamatergic nerves. Conclusion These findings underscore the potential of digilanid C and ES combination therapy as a novel approach to modulate the complex interplay between neurovascular dynamics and metabolic dysregulation in CHF.
Collapse
Affiliation(s)
- Yun Liu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiao Sun
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Mingqian Yuan
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhi Yu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qun Hou
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhengxu Jia
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tiancheng Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Bin Xu
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
35
|
Lundby A, Achter JS, Goodyer WR. Utilizing multi-omics strategies to unravel the molecular basis of heart rhythm. Heart Rhythm 2024; 21:1761-1762. [PMID: 39209409 DOI: 10.1016/j.hrthm.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jonathan S Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - William R Goodyer
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, California; Department of Pediatrics, Stanford University, Stanford, California
| |
Collapse
|
36
|
Dance A. What is a cell type, really? The quest to categorize life's myriad forms. Nature 2024; 633:754-756. [PMID: 39317746 DOI: 10.1038/d41586-024-03073-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
|
37
|
Fayyaz AU, Eltony M, Prokop LJ, Koepp KE, Borlaug BA, Dasari S, Bois MC, Margulies KB, Maleszewski JJ, Wang Y, Redfield MM. Pathophysiological insights into HFpEF from studies of human cardiac tissue. Nat Rev Cardiol 2024:10.1038/s41569-024-01067-1. [PMID: 39198624 DOI: 10.1038/s41569-024-01067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/18/2024] [Indexed: 09/01/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major, worldwide health-care problem. Few therapies for HFpEF exist because the pathophysiology of this condition is poorly defined and, increasingly, postulated to be diverse. Although perturbations in other organs contribute to the clinical profile in HFpEF, altered cardiac structure, function or both are the primary causes of this heart failure syndrome. Therefore, studying myocardial tissue is fundamental to improve pathophysiological insights and therapeutic discovery in HFpEF. Most studies of myocardial changes in HFpEF have relied on cardiac tissue from animal models without (or with limited) confirmatory studies in human cardiac tissue. Animal models of HFpEF have evolved based on theoretical HFpEF aetiologies, but these models might not reflect the complex pathophysiology of human HFpEF. The focus of this Review is the pathophysiological insights gained from studies of human HFpEF myocardium. We outline the rationale for these studies, the challenges and opportunities in obtaining myocardial tissue from patients with HFpEF and relevant comparator groups, the analytical approaches, the pathophysiological insights gained to date and the remaining knowledge gaps. Our objective is to provide a roadmap for future studies of cardiac tissue from diverse cohorts of patients with HFpEF, coupling discovery biology with measures to account for pathophysiological diversity.
Collapse
Affiliation(s)
- Ahmed U Fayyaz
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Muhammad Eltony
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Larry J Prokop
- Mayo Clinic College of Medicine and Science, Library Reference Service, Rochester, MN, USA
| | - Katlyn E Koepp
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Barry A Borlaug
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Surendra Dasari
- Mayo Clinic College of Medicine and Science, Computational Biology, Rochester, MN, USA
| | - Melanie C Bois
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Kenneth B Margulies
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joesph J Maleszewski
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Ying Wang
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA
| | - Margaret M Redfield
- Department of Cardiovascular Disease, Division of Circulatory Failure, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
38
|
Combémorel N, Cavell N, Tyser RCV. Early heart development: examining the dynamics of function-form emergence. Biochem Soc Trans 2024; 52:1579-1589. [PMID: 38979619 DOI: 10.1042/bst20230546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/10/2024]
Abstract
During early embryonic development, the heart undergoes a remarkable and complex transformation, acquiring its iconic four-chamber structure whilst concomitantly contracting to maintain its essential function. The emergence of cardiac form and function involves intricate interplays between molecular, cellular, and biomechanical events, unfolding with precision in both space and time. The dynamic morphological remodelling of the developing heart renders it particularly vulnerable to congenital defects, with heart malformations being the most common type of congenital birth defect (∼35% of all congenital birth defects). This mini-review aims to give an overview of the morphogenetic processes which govern early heart formation as well as the dynamics and mechanisms of early cardiac function. Moreover, we aim to highlight some of the interplay between these two processes and discuss how recent findings and emerging techniques/models offer promising avenues for future exploration. In summary, the developing heart is an exciting model to gain fundamental insight into the dynamic relationship between form and function, which will augment our understanding of cardiac congenital defects and provide a blueprint for potential therapeutic strategies to treat disease.
Collapse
Affiliation(s)
- Noémie Combémorel
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| | - Natasha Cavell
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| | - Richard C V Tyser
- Cambridge Stem Cell Institute, University of Cambridge, Jeffrey Cheah Biomedical Centre, Cambridge CB2 0AW, U.K
| |
Collapse
|
39
|
Liu L, Chen A, Li Y, Mulder J, Heyn H, Xu X. Spatiotemporal omics for biology and medicine. Cell 2024; 187:4488-4519. [PMID: 39178830 DOI: 10.1016/j.cell.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 08/26/2024]
Abstract
The completion of the Human Genome Project has provided a foundational blueprint for understanding human life. Nonetheless, understanding the intricate mechanisms through which our genetic blueprint is involved in disease or orchestrates development across temporal and spatial dimensions remains a profound scientific challenge. Recent breakthroughs in cellular omics technologies have paved new pathways for understanding the regulation of genomic elements and the relationship between gene expression, cellular functions, and cell fate determination. The advent of spatial omics technologies, encompassing both imaging and sequencing-based methodologies, has enabled a comprehensive understanding of biological processes from a cellular ecosystem perspective. This review offers an updated overview of how spatial omics has advanced our understanding of the translation of genetic information into cellular heterogeneity and tissue structural organization and their dynamic changes over time. It emphasizes the discovery of various biological phenomena, related to organ functionality, embryogenesis, species evolution, and the pathogenesis of diseases.
Collapse
Affiliation(s)
| | - Ao Chen
- BGI Research, Shenzhen 518083, China
| | | | - Jan Mulder
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Holger Heyn
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China.
| |
Collapse
|
40
|
Momin N, Pabel S, Rudra A, Kumowski N, Lee IH, Mentkowski K, Yamazoe M, Stengel L, Muse CG, Seung H, Paccalet A, Gonzalez-Correa C, Jacobs EB, Grune J, Schloss MJ, Sossalla S, Wojtkiewicz G, Iwamoto Y, McMullen P, Mitchell RN, Ellinor PT, Anderson DG, Naxerova K, Nahrendorf M, Hulsmans M. Therapeutic Spp1 silencing in TREM2 + cardiac macrophages suppresses atrial fibrillation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.10.607461. [PMID: 39149373 PMCID: PMC11326243 DOI: 10.1101/2024.08.10.607461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Atrial fibrillation (AFib) and the risk of its lethal complications are propelled by fibrosis, which induces electrical heterogeneity and gives rise to reentry circuits. Atrial TREM2+ macrophages secrete osteopontin (encoded by Spp1), a matricellular signaling protein that engenders fibrosis and AFib. Here we show that silencing Spp1 in TREM2+ cardiac macrophages with an antibody-siRNA conjugate reduces atrial fibrosis and suppresses AFib in mice, thus offering a new immunotherapy for the most common arrhythmia.
Collapse
Affiliation(s)
- Noor Momin
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA
| | - Steffen Pabel
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Arnab Rudra
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard–MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nina Kumowski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - I-Hsiu Lee
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Blavatnik Institute, Genetics, Harvard Medical School, Boston, MA, USA
| | - Kyle Mentkowski
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Masahiro Yamazoe
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura Stengel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany
| | - Charlotte G. Muse
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Hana Seung
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexandre Paccalet
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Cristina Gonzalez-Correa
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emily B. Jacobs
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA
| | - Jana Grune
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Maximilian J. Schloss
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Samuel Sossalla
- Department of Cardiology, University Hospital Giessen, Kerckhoff Clinic Bad Nauheim, and DZHK, Partner site RhineMain, Germany
| | - Gregory Wojtkiewicz
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Patrick McMullen
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard–MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Richard N. Mitchell
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Patrick T. Ellinor
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard University, Cambridge, MA, USA
| | - Daniel G. Anderson
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital, Boston, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard–MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kamila Naxerova
- Blavatnik Institute, Genetics, Harvard Medical School, Boston, MA, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Internal Medicine I, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Maarten Hulsmans
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
41
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
42
|
Kamaraj US, Gautam P, Cheng T, Chin TS, Tay SK, Ho TH, Nadarajah R, Goh RCH, Wong SL, Mantoo S, Busmanis I, Li H, Le MT, Li QJ, Lim EH, Loh YH. Deciphering tumour microenvironment and elucidating the origin of cancer cells in ovarian clear cell carcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606821. [PMID: 39149248 PMCID: PMC11326226 DOI: 10.1101/2024.08.06.606821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Ovarian clear cell carcinoma (CCC) has an East Asian preponderance. It is associated with endometriosis, a benign condition where endometrial (inner lining of the uterus) tissue is found outside the uterus and on the peritoneal surface, in the abdominal or pelvic space. CCC is relatively more resistant to conventional chemotherapy compared to other ovarian cancer subtypes and is associated with a poorer prognosis. In this study, we recruited and obtained tumour tissues from seven patients across the four stages of CCC. The tumour and the tumour microenvironment (TME) from 7 CCC patients spanning clinical stages 1-4 were transcriptionally profiled using high-resolution scRNA-seq to gain insight into CCC's biological mechanisms. Firstly, we built a scRNA-seq resource for the CCC tumour microenvironment (TME). Secondly, we identified the different cell type proportions and found high levels of immune infiltration in CCC. Thirdly, since CCC is associated with endometriosis, we compared CCC with two publicly available endometriosis scRNA-seq datasets. The CCC malignant cells showed similarities with glandular secretory and ciliated epithelial cells found in endometriosis. Finally, we determined the differences in cell-cell communication between various cell types present in CCC TME and endometriosis conditions to gain insights into the transformations in CCC.
Collapse
Affiliation(s)
- Uma S Kamaraj
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Pradeep Gautam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Terence Cheng
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Tham Su Chin
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Sun Kuie Tay
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Tew Hong Ho
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Ravichandran Nadarajah
- Department of Obstetrics & Gynaecology, Singapore General Hospital, Outram Road, Singapore 169608
| | - Ronald Chin Hong Goh
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Shing Lih Wong
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Sangeeta Mantoo
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Inny Busmanis
- Department of Anatomical Pathology, Singapore General Hospital, Academia, College Road, Singapore 169856
| | - Hu Li
- Center for Individualized Medicine, Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Minh Tn Le
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Qi-Jing Li
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Elaine Hsuen Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583
| | - Yuin-Han Loh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
- Department of Physiology, NUS Yong Loo Lin School of Medicine, 2 Medical Drive, MD9, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- NUS Graduate School's Integrative Sciences and Engineering Programme, National University of Singapore, 28 Medical Drive, Singapore, Singapore
| |
Collapse
|
43
|
Oh Y, Abid R, Dababneh S, Bakr M, Aslani T, Cook DP, Vanderhyden BC, Park JG, Munshi NV, Hui CC, Kim KH. Transcriptional regulation of the postnatal cardiac conduction system heterogeneity. Nat Commun 2024; 15:6550. [PMID: 39095365 PMCID: PMC11297185 DOI: 10.1038/s41467-024-50849-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
The cardiac conduction system (CCS) is a network of specialized cardiomyocytes that coordinates electrical impulse generation and propagation for synchronized heart contractions. Although the components of the CCS, including the sinoatrial node, atrioventricular node, His bundle, bundle branches, and Purkinje fibers, were anatomically discovered more than 100 years ago, their molecular constituents and regulatory mechanisms remain incompletely understood. Here, we demonstrate the transcriptomic landscape of the postnatal mouse CCS at a single-cell resolution with spatial information. Integration of single-cell and spatial transcriptomics uncover region-specific markers and zonation patterns of expression. Network inference shows heterogeneous gene regulatory networks across the CCS. Notably, region-specific gene regulation is recapitulated in vitro using neonatal mouse atrial and ventricular myocytes overexpressing CCS-specific transcription factors, Tbx3 and/or Irx3. This finding is supported by ATAC-seq of different CCS regions, Tbx3 ChIP-seq, and Irx motifs. Overall, this study provides comprehensive molecular profiles of the postnatal CCS and elucidates gene regulatory mechanisms contributing to its heterogeneity.
Collapse
Affiliation(s)
- Yena Oh
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rimshah Abid
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Saif Dababneh
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marwan Bakr
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Termeh Aslani
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - David P Cook
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Barbara C Vanderhyden
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jin G Park
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Nikhil V Munshi
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, USA
- Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chi-Chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Kyoung-Han Kim
- University of Ottawa Heart Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
44
|
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, Georgakilas AG. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy. Pharmacol Ther 2024; 260:108670. [PMID: 38823489 DOI: 10.1016/j.pharmthera.2024.108670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece; Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | - Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Yağmur Kafalı
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris I Giotakis
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197 Berlin, Germany
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany; German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany; Genomics Core Facility, Philipps-University, 35043 Marburg, Germany; Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany; Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece.
| |
Collapse
|
45
|
Achter JS, Vega ET, Sorrentino A, Kahnert K, Galsgaard KD, Hernandez-Varas P, Wierer M, Holst JJ, Wojtaszewski JFP, Mills RW, Kjøbsted R, Lundby A. In-depth phosphoproteomic profiling of the insulin signaling response in heart tissue and cardiomyocytes unveils canonical and specialized regulation. Cardiovasc Diabetol 2024; 23:258. [PMID: 39026321 PMCID: PMC11264841 DOI: 10.1186/s12933-024-02338-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/26/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Insulin signaling regulates cardiac substrate utilization and is implicated in physiological adaptations of the heart. Alterations in the signaling response within the heart are believed to contribute to pathological conditions such as type-2 diabetes and heart failure. While extensively investigated in several metabolic organs using phosphoproteomic strategies, the signaling response elicited in cardiac tissue in general, and specifically in the specialized cardiomyocytes, has not yet been investigated to the same extent. METHODS Insulin or vehicle was administered to male C57BL6/JRj mice via intravenous injection into the vena cava. Ventricular tissue was extracted and subjected to quantitative phosphoproteomics analysis to evaluate the insulin signaling response. To delineate the cardiomyocyte-specific response and investigate the role of Tbc1d4 in insulin signal transduction, cardiomyocytes from the hearts of cardiac and skeletal muscle-specific Tbc1d4 knockout mice, as well as from wildtype littermates, were studied. The phosphoproteomic studies involved isobaric peptide labeling with Tandem Mass Tags (TMT), enrichment for phosphorylated peptides, fractionation via micro-flow reversed-phase liquid chromatography, and high-resolution mass spectrometry measurements. RESULTS We quantified 10,399 phosphorylated peptides from ventricular tissue and 12,739 from isolated cardiomyocytes, localizing to 3,232 and 3,128 unique proteins, respectively. In cardiac tissue, we identified 84 insulin-regulated phosphorylation events, including sites on the Insulin Receptor (InsrY1351, Y1175, Y1179, Y1180) itself as well as the Insulin receptor substrate protein 1 (Irs1S522, S526). Predicted kinases with increased activity in response to insulin stimulation included Rps6kb1, Akt1 and Mtor. Tbc1d4 emerged as a major phosphorylation target in cardiomyocytes. Despite limited impact on the global phosphorylation landscape, Tbc1d4 deficiency in cardiomyocytes attenuated insulin-induced Glut4 translocation and induced protein remodeling. We observed 15 proteins significantly regulated upon knockout of Tbc1d4. While Glut4 exhibited decreased protein abundance consequent to Tbc1d4-deficiency, Txnip levels were notably increased. Stimulation of wildtype cardiomyocytes with insulin led to the regulation of 262 significant phosphorylation events, predicted to be regulated by kinases such as Akt1, Mtor, Akt2, and Insr. In cardiomyocytes, the canonical insulin signaling response is elicited in addition to regulation on specialized cardiomyocyte proteins, such as Kcnj11Y12 and DspS2597. Details of all phosphorylation sites are provided. CONCLUSION We present a first global outline of the insulin-induced phosphorylation signaling response in heart tissue and in isolated adult cardiomyocytes, detailing the specific residues with changed phosphorylation abundances. Our study marks an important step towards understanding the role of insulin signaling in cardiac diseases linked to insulin resistance.
Collapse
Affiliation(s)
- Jonathan Samuel Achter
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Estefania Torres Vega
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andrea Sorrentino
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Konstantin Kahnert
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Douglas Galsgaard
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Pablo Hernandez-Varas
- Core Facility for Integrated Microscopy, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Wierer
- Proteomics Research Infrastructure, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Frank Pind Wojtaszewski
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Robert William Mills
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rasmus Kjøbsted
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Alicia Lundby
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
46
|
Jurida L, Werner S, Knapp F, Niemann B, Li L, Grün D, Wirth S, Weber A, Beuerlein K, Liebetrau C, Wiedenroth CB, Guth S, Kojonazarov B, Jafari L, Weissmann N, Günther S, Braun T, Bartkuhn M, Schermuly RT, Dorfmüller P, Yin X, Mayr M, Schmitz ML, Czech L, Schlüter KD, Schulz R, Rohrbach S, Kracht M. A common gene signature of the right ventricle in failing rat and human hearts. NATURE CARDIOVASCULAR RESEARCH 2024; 3:819-840. [PMID: 39196177 PMCID: PMC11358011 DOI: 10.1038/s44161-024-00485-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 05/02/2024] [Indexed: 08/29/2024]
Abstract
The molecular mechanisms of progressive right heart failure are incompletely understood. In this study, we systematically examined transcriptomic changes occurring over months in isolated cardiomyocytes or whole heart tissues from failing right and left ventricles in rat models of pulmonary artery banding (PAB) or aortic banding (AOB). Detailed bioinformatics analyses resulted in the identification of gene signature, protein and transcription factor networks specific to ventricles and compensated or decompensated disease states. Proteomic and RNA-FISH analyses confirmed PAB-mediated regulation of key genes and revealed spatially heterogeneous mRNA expression in the heart. Intersection of rat PAB-specific gene sets with transcriptome datasets from human patients with chronic thromboembolic pulmonary hypertension (CTEPH) led to the identification of more than 50 genes whose expression levels correlated with the severity of right heart disease, including multiple matrix-regulating and secreted factors. These data define a conserved, differentially regulated genetic network associated with right heart failure in rats and humans.
Collapse
Affiliation(s)
- Liane Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Sebastian Werner
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Fabienne Knapp
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Bernd Niemann
- Department of Cardiac and Vascular Surgery, Justus Liebig University, Giessen, Germany
| | - Ling Li
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Dimitri Grün
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Stefanie Wirth
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Axel Weber
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Knut Beuerlein
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany
| | - Christoph Liebetrau
- Department of Cardiology and Angiology, Justus Liebig University, Giessen, Germany
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | | | - Stefan Guth
- Department of Thoracic Surgery, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
| | - Baktybek Kojonazarov
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Leili Jafari
- Department of Cardiology, Kerckhoff Heart and Lung Center, Bad Nauheim, Germany
- Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Norbert Weissmann
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Günther
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Cardio-Pulmonary Institute, Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Marek Bartkuhn
- Cardio-Pulmonary Institute, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Institute for Lung Health, Justus Liebig University Giessen, Giessen, Germany
| | - Ralph T Schermuly
- Medical Clinic II, Justus Liebig University, Giessen, Germany
- Cardio-Pulmonary Institute, Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
| | - Peter Dorfmüller
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- German Center for Lung Research (DZL), Giessen, Germany
- Department of Internal Medicine, Justus Liebig University Giessen, Giessen, Germany
- Institute of Pathology, Justus Liebig University Giessen, Giessen, Germany
| | - Xiaoke Yin
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - Manuel Mayr
- National Heart and Lung Institute, Faculty of Medicine,Imperial College London, London, UK
| | - M Lienhard Schmitz
- German Center for Lung Research (DZL), Giessen, Germany
- Institute of Biochemistry, Justus Liebig University, Giessen, Germany
| | - Laureen Czech
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | | | - Rainer Schulz
- Department of Physiology, Justus Liebig University, Giessen, Germany
| | - Susanne Rohrbach
- Department of Physiology, Justus Liebig University, Giessen, Germany.
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University, Giessen, Germany.
- Cardio-Pulmonary Institute, Giessen, Germany.
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany.
- German Center for Lung Research (DZL), Giessen, Germany.
| |
Collapse
|
47
|
de Winter N, Ji J, Sintou A, Forte E, Lee M, Noseda M, Li A, Koenig AL, Lavine KJ, Hayat S, Rosenthal N, Emanueli C, Srivastava PK, Sattler S. Persistent transcriptional changes in cardiac adaptive immune cells following myocardial infarction: New evidence from the re-analysis of publicly available single cell and nuclei RNA-sequencing data sets. J Mol Cell Cardiol 2024; 192:48-64. [PMID: 38734060 DOI: 10.1016/j.yjmcc.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 03/17/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024]
Abstract
INTRODUCTION Chronic immunopathology contributes to the development of heart failure after a myocardial infarction. Both T and B cells of the adaptive immune system are present in the myocardium and have been suggested to be involved in post-MI immunopathology. METHODS We analyzed the B and T cell populations isolated from previously published single cell RNA-sequencing data sets (PMID: 32130914, PMID: 35948637, PMID: 32971526 and PMID: 35926050), of the mouse and human heart, using differential expression analysis, functional enrichment analysis, gene regulatory inferences, and integration with autoimmune and cardiovascular GWAS. RESULTS Already at baseline, mature effector B and T cells are present in the human and mouse heart, having increased activity in transcription factors maintaining tolerance (e.g. DEAF1, JDP2, SPI-B). Following MI, T cells upregulate pro-inflammatory transcript levels (e.g. Cd11, Gzmk, Prf1), while B cells upregulate activation markers (e.g. Il6, Il1rn, Ccl6) and collagen (e.g. Col5a2, Col4a1, Col1a2). Importantly, pro-inflammatory and fibrotic transcription factors (e.g. NFKB1, CREM, REL) remain active in T cells, while B cells maintain elevated activity in transcription factors related to immunoglobulin production (e.g. ERG, REL) in both mouse and human post-MI hearts. Notably, genes differentially expressed in post-MI T and B cells are associated with cardiovascular and autoimmune disease. CONCLUSION These findings highlight the varied and time-dependent dynamic roles of post-MI T and B cells. They appear ready-to-go and are activated immediately after MI, thus participate in the acute wound healing response. However, they subsequently remain in a state of pro-inflammatory activation contributing to persistent immunopathology.
Collapse
Affiliation(s)
- Natasha de Winter
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Jiahui Ji
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Amalia Sintou
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Elvira Forte
- The Jackson Laboratory, Bar Harbor, United States
| | - Michael Lee
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Michela Noseda
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; British Heart Foundation Centre For Research Excellence, Imperial College London, United Kingdom
| | - Aoxue Li
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; Department of Medicine Solna, Division of Cardiovascular Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Andrew L Koenig
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States
| | - Kory J Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States
| | | | - Nadia Rosenthal
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; The Jackson Laboratory, Bar Harbor, United States
| | - Costanza Emanueli
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; British Heart Foundation Centre For Research Excellence, Imperial College London, United Kingdom
| | - Prashant K Srivastava
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom
| | - Susanne Sattler
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, United Kingdom; Department of Cardiology, Medical University of Graz, Austria; Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Austria.
| |
Collapse
|
48
|
Au Yeung VPW, Obrezanova O, Zhou J, Yang H, Bowen TJ, Ivanov D, Saffadi I, Carter AS, Subramanian V, Dillmann I, Hall A, Corrigan A, Viant MR, Pointon A. Computational approaches identify a transcriptomic fingerprint of drug-induced structural cardiotoxicity. Cell Biol Toxicol 2024; 40:50. [PMID: 38940987 PMCID: PMC11213733 DOI: 10.1007/s10565-024-09880-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/15/2024] [Indexed: 06/29/2024]
Abstract
Structural cardiotoxicity (SCT) presents a high-impact risk that is poorly tolerated in drug discovery unless significant benefit is anticipated. Therefore, we aimed to improve the mechanistic understanding of SCT. First, we combined machine learning methods with a modified calcium transient assay in human-induced pluripotent stem cell-derived cardiomyocytes to identify nine parameters that could predict SCT. Next, we applied transcriptomic profiling to human cardiac microtissues exposed to structural and non-structural cardiotoxins. Fifty-two genes expressed across the three main cell types in the heart (cardiomyocytes, endothelial cells, and fibroblasts) were prioritised in differential expression and network clustering analyses and could be linked to known mechanisms of SCT. This transcriptomic fingerprint may prove useful for generating strategies to mitigate SCT risk in early drug discovery.
Collapse
Affiliation(s)
- Victoria P W Au Yeung
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK.
- Phenomics, Data Sciences & Quantitative Biology, R&D AstraZeneca, Cambridge, UK.
| | - Olga Obrezanova
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jiarui Zhou
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Hongbin Yang
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Tara J Bowen
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Delyan Ivanov
- High-Throughput Screening, R&D, AstraZeneca, Alderley Park, UK
| | - Izzy Saffadi
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Alfie S Carter
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Vigneshwari Subramanian
- Imaging and Data Analytics, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Inken Dillmann
- Disease Molecular Profiling, Discovery Biology, R&D AstraZeneca, Gothenburg, Sweden
| | - Andrew Hall
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Adam Corrigan
- Phenomics, Data Sciences & Quantitative Biology, R&D AstraZeneca, Cambridge, UK
| | - Mark R Viant
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, UK
- Phenome Centre Birmingham, University of Birmingham, Edgbaston, Birmingham, UK
| | - Amy Pointon
- Safety Sciences, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| |
Collapse
|
49
|
Lee SC, Huang CH, Oyang YJ, Huang HC, Juan HF. Macrophages as determinants and regulators of systemic sclerosis-related interstitial lung disease. J Transl Med 2024; 22:600. [PMID: 38937794 PMCID: PMC11212242 DOI: 10.1186/s12967-024-05403-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Interstitial lung disease (ILD) is the primary cause of mortality in systemic sclerosis (SSc), an autoimmune disease characterized by tissue fibrosis. SSc-related ILD (SSc-ILD) occurs more frequently in females aged 30-55 years, whereas idiopathic pulmonary fibrosis (IPF) is more prevalent in males aged 60-75 years. SSc-ILD occurs earlier than IPF and progresses rapidly. FCN1, FABP4, and SPP1 macrophages are involved in the pathogenesis of lung fibrosis; SPP1 macrophages demonstrate upregulated expression in both SSc-ILD and IPF. To identify the differences between SSc-ILD and IPF using single-cell analysis, clarify their distinct pathogeneses, and propose directions for prevention and treatment. METHODS We performed single-cell RNA sequencing on NCBI Gene Expression Omnibus (GEO) databases GSE159354 and GSE212109, and analyzed lung tissue samples across healthy controls, IPF, and SSc-ILD. The primary measures were the filtered genes integrated with batch correction and annotated cell types for distinguishing patients with SSc-ILD from healthy controls. We proposed an SSc-ILD pathogenesis using cell-cell interaction inferences, and predicted transcription factors regulating target genes using SCENIC. Drug target prediction of the TF gene was performed using Drug Bank Online. RESULTS A subset of macrophages activates the MAPK signaling pathway under oxidative stress. Owing to the lack of inhibitory feedback from ANNEXIN and the autoimmune characteristics, this leads to an earlier onset of lung fibrosis compared to IPF. During initial lung injury, fibroblasts begin to activate the IL6 pathway under the influence of SPP1 alveolar macrophages, but IL6 appears unrelated to other inflammatory and immune cells. This may explain why tocilizumab (an anti-IL6-receptor antibody) only preserves lung function in patients with early SSc-ILD. Finally, we identified BCLAF1 and NFE2L2 as influencers of MAPK activation in macrophages. Metformin downregulates NFE2L2 and could serve as a repurposed drug candidate. CONCLUSIONS SPP1 alveolar macrophages play a role in the profibrotic activity of IPF and SSc-ILD. However, SSc-ILD is influenced by autoimmunity and oxidative stress, leading to the continuous activation of MAPK in macrophages. This may result in an earlier onset of lung fibrosis than in IPF. Such differences could serve as potential research directions for early prevention and treatment.
Collapse
Affiliation(s)
- Shih-Ching Lee
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan
- Division of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan
| | - Chen-Hao Huang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan
| | - Yen-Jen Oyang
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan.
| | - Hsueh-Fen Juan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, 10617, Taiwan.
- Department of Life Science, National Taiwan University, Taipei, 106, Taiwan.
- Center for Computational and Systems Biology, National Taiwan University, Taipei, 106, Taiwan.
| |
Collapse
|
50
|
Patrick R, Janbandhu V, Tallapragada V, Tan SSM, McKinna EE, Contreras O, Ghazanfar S, Humphreys DT, Murray NJ, Tran YTH, Hume RD, Chong JJH, Harvey RP. Integration mapping of cardiac fibroblast single-cell transcriptomes elucidates cellular principles of fibrosis in diverse pathologies. SCIENCE ADVANCES 2024; 10:eadk8501. [PMID: 38905342 PMCID: PMC11192082 DOI: 10.1126/sciadv.adk8501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Single-cell technology has allowed researchers to probe tissue complexity and dynamics at unprecedented depth in health and disease. However, the generation of high-dimensionality single-cell atlases and virtual three-dimensional tissues requires integrated reference maps that harmonize disparate experimental designs, analytical pipelines, and taxonomies. Here, we present a comprehensive single-cell transcriptome integration map of cardiac fibrosis, which underpins pathophysiology in most cardiovascular diseases. Our findings reveal similarity between cardiac fibroblast (CF) identities and dynamics in ischemic versus pressure overload models of cardiomyopathy. We also describe timelines for commitment of activated CFs to proliferation and myofibrogenesis, profibrotic and antifibrotic polarization of myofibroblasts and matrifibrocytes, and CF conservation across mouse and human healthy and diseased hearts. These insights have the potential to inform knowledge-based therapies.
Collapse
Affiliation(s)
- Ralph Patrick
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Vaibhao Janbandhu
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | | | - Shannon S. M. Tan
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Emily E. McKinna
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
| | - Osvaldo Contreras
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David T. Humphreys
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Nicholas J. Murray
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Yen T. H. Tran
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
| | - Robert D. Hume
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- School of Medical Science, The University of Sydney, Camperdown, NSW 2006, Australia
- Centre for Heart Failure and Diseases of the Aorta, The Baird Institute, Sydney, NSW 2042, Australia
| | - James J. H. Chong
- Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
| | - Richard P. Harvey
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia
- School of Biotechnology and Biomolecular Science, UNSW Sydney, Kensington, NSW 2052, Australia
| |
Collapse
|