1
|
Lovell JP, Duque C, Rousseau S, Bhalodia A, Bermea K, Cohen CD, Adamo L. B cell-mediated antigen presentation promotes adverse cardiac remodeling in chronic heart failure. RESEARCH SQUARE 2024:rs.3.rs-4536350. [PMID: 38978561 PMCID: PMC11230502 DOI: 10.21203/rs.3.rs-4536350/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide. A primary driver of cardiovascular mortality is ischemic heart failure, a form of cardiac dysfunction that can develop in patients who survive myocardial infarction. Acute cardiac damage triggers robust changes in the spleen with rapid migration of immune cells from the spleen to the heart. Activating this "cardio-splenic" axis contributes to progressive cardiac dysfunction. The cardio-splenic axis has, therefore, been identified as a promising therapeutic target to prevent or treat heart failure. However, our understanding of the precise mechanisms by which specific immune cells contribute to adverse cardiac remodeling within the cardio-splenic axis remains limited. Here, we show that splenic B cells contribute to the development of heart failure via MHC II-mediated antigen presentation. We found that the adoptive transfer of splenic B cells from mice with ischemic heart failure promoted adverse cardiac remodeling and splenic inflammatory changes in naïve recipient mice. Based on single-cell RNA sequencing analysis of splenic B cells from mice with ischemic heart failure, we hypothesized that B cells contributed to adverse cardiac remodeling through antigen presentation by MHC II molecules. This mechanism was confirmed using transgenic mice with B cell-specific MHC II deletion, and by analyzing circulating B cells from humans who experienced myocardial infarction. Our results broaden our understanding of B lymphocyte biology, reshape current models of immune activation in response to myocardial injury, and point towards MHC II-mediated signaling in B cells as a novel and specific therapeutic target in chronic heart failure.
Collapse
Affiliation(s)
- Jana P. Lovell
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carolina Duque
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Sylvie Rousseau
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Aashik Bhalodia
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kevin Bermea
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Charles D. Cohen
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Luigi Adamo
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Lovell JP, Duque C, Rousseau S, Bhalodia A, Bermea K, Cohen CD, Adamo L. B cell-mediated antigen presentation promotes adverse cardiac remodeling in chronic heart failure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593153. [PMID: 38766182 PMCID: PMC11100706 DOI: 10.1101/2024.05.08.593153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Cardiovascular disease remains the leading cause of death worldwide. A primary driver of cardiovascular mortality is ischemic heart failure, a form of cardiac dysfunction that can develop in patients who survive myocardial infarction. Acute cardiac damage triggers robust changes in the spleen with rapid migration of immune cells from the spleen to the heart. Activating this "cardio-splenic" axis contributes to progressive cardiac dysfunction. The cardio-splenic axis has, therefore, been identified as a promising therapeutic target to prevent or treat heart failure. However, our understanding of the precise mechanisms by which specific immune cells contribute to adverse cardiac remodeling within the cardio-splenic axis remains limited. Here, we show that splenic B cells contribute to the development of heart failure via MHC II-mediated antigen presentation. We found that the adoptive transfer of splenic B cells from mice with ischemic heart failure promoted adverse cardiac remodeling and splenic inflammatory changes in naïve recipient mice. Based on single-cell RNA sequencing analysis of splenic B cells from mice with ischemic heart failure, we hypothesized that B cells contributed to adverse cardiac remodeling through antigen presentation by MHC II molecules. This mechanism was confirmed using transgenic mice with B cell-specific MHC II deletion, and by analyzing circulating B cells from humans who experienced myocardial infarction. Our results broaden our understanding of B lymphocyte biology, reshape current models of immune activation in response to myocardial injury, and point towards MHC II-mediated signaling in B cells as a novel and specific therapeutic target in chronic heart failure.
Collapse
|
3
|
Mehdipour M, Park S, Huang GN. Unlocking cardiomyocyte renewal potential for myocardial regeneration therapy. J Mol Cell Cardiol 2023; 177:9-20. [PMID: 36801396 PMCID: PMC10699255 DOI: 10.1016/j.yjmcc.2023.02.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/28/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Cardiovascular disease remains the leading cause of mortality worldwide. Cardiomyocytes are irreversibly lost due to cardiac ischemia secondary to disease. This leads to increased cardiac fibrosis, poor contractility, cardiac hypertrophy, and subsequent life-threatening heart failure. Adult mammalian hearts exhibit notoriously low regenerative potential, further compounding the calamities described above. Neonatal mammalian hearts, on the other hand, display robust regenerative capacities. Lower vertebrates such as zebrafish and salamanders retain the ability to replenish lost cardiomyocytes throughout life. It is critical to understand the varying mechanisms that are responsible for these differences in cardiac regeneration across phylogeny and ontogeny. Adult mammalian cardiomyocyte cell cycle arrest and polyploidization have been proposed as major barriers to heart regeneration. Here we review current models about why adult mammalian cardiac regenerative potential is lost including changes in environmental oxygen levels, acquisition of endothermy, complex immune system development, and possible cancer risk tradeoffs. We also discuss recent progress and highlight conflicting reports pertaining to extrinsic and intrinsic signaling pathways that control cardiomyocyte proliferation and polyploidization in growth and regeneration. Uncovering the physiological brakes of cardiac regeneration could illuminate novel molecular targets and offer promising therapeutic strategies to treat heart failure.
Collapse
Affiliation(s)
- Melod Mehdipour
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sangsoon Park
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94158, USA; Bakar Aging Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
4
|
Shao X, Liu X, Qi H. A role for Hes1 in constraining germinal center B cell formation. CELL INSIGHT 2023; 2:100078. [PMID: 37193067 PMCID: PMC10134198 DOI: 10.1016/j.cellin.2023.100078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/17/2023] [Accepted: 01/17/2023] [Indexed: 05/18/2023]
Abstract
Germinal center is a transient lymphoid tissue structure in which B cells undergo affinity maturation and differentiate into memory B cells and plasma cells. GC formation depends on B cell expression of BCL6, a master transcription regulator of the GC state. Bcl6 expression is under elaborate control by external signals. HES1 plays important roles in T-cell lineage commitment, although little is known about its potential roles in GC formation. Here we report that B-cell-specific HES1 deletion causes a significant increase in GC formation, leading to increased production of plasma cells. We further provide evidence that HES1 inhibits BCL6 expression in a bHLH domain-dependent manner. Our study suggests a new layer of regulation of GC initiation mediated by HES1 and, by inference, Notch signals in vivo.
Collapse
Affiliation(s)
- Xingxing Shao
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xin Liu
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Hai Qi
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Frontiers Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
5
|
Notch Signaling in Acute Inflammation and Sepsis. Int J Mol Sci 2023; 24:ijms24043458. [PMID: 36834869 PMCID: PMC9967996 DOI: 10.3390/ijms24043458] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Notch signaling, a highly conserved pathway in mammals, is crucial for differentiation and homeostasis of immune cells. Besides, this pathway is also directly involved in the transmission of immune signals. Notch signaling per se does not have a clear pro- or anti-inflammatory effect, but rather its impact is highly dependent on the immune cell type and the cellular environment, modulating several inflammatory conditions including sepsis, and therefore significantly impacts the course of disease. In this review, we will discuss the contribution of Notch signaling on the clinical picture of systemic inflammatory diseases, especially sepsis. Specifically, we will review its role during immune cell development and its contribution to the modulation of organ-specific immune responses. Finally, we will evaluate to what extent manipulation of the Notch signaling pathway could be a future therapeutic strategy.
Collapse
|
6
|
Quotti Tubi L, Mandato E, Canovas Nunes S, Arjomand A, Zaffino F, Manni S, Casellato A, Macaccaro P, Vitulo N, Zumerle S, Filhol O, Boldyreff B, Siebel CW, Viola A, Valle G, Mainoldi F, Casola S, Cancila V, Gulino A, Tripodo C, Pizzi M, Dei Tos AP, Trentin L, Semenzato G, Piazza F. CK2β-regulated signaling controls B cell differentiation and function. Front Immunol 2023; 13:959138. [PMID: 36713383 PMCID: PMC9874936 DOI: 10.3389/fimmu.2022.959138] [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: 06/01/2022] [Accepted: 12/08/2022] [Indexed: 01/13/2023] Open
Abstract
Serine-Threonine kinase CK2 supports malignant B-lymphocyte growth but its role in B-cell development and activation is largely unknown. Here, we describe the first B-cell specific knockout (KO) mouse model of the β regulatory subunit of CK2. CK2βKO mice present an increase in marginal zone (MZ) and a reduction in follicular B cells, suggesting a role for CK2 in the regulation of the B cell receptor (BCR) and NOTCH2 signaling pathways. Biochemical analyses demonstrate an increased activation of the NOTCH2 pathway in CK2βKO animals, which sustains MZ B-cell development. Transcriptomic analyses indicate alterations in biological processes involved in immune response and B-cell activation. Upon sheep red blood cells (SRBC) immunization CK2βKO mice exhibit enlarged germinal centers (GCs) but display a limited capacity to generate class-switched GC B cells and immunoglobulins. In vitro assays highlight that B cells lacking CK2β have an impaired signaling downstream of BCR, Toll-like receptor, CD40, and IL-4R all crucial for B-cell activation and antigen presenting efficiency. Somatic hypermutations analysis upon 4-Hydroxy-3-nitrophenylacetyl hapten conjugated to Chicken Gamma Globulin (NP-CGG) evidences a reduced NP-specific W33L mutation frequency in CK2βKO mice suggesting the importance of the β subunit in sustaining antibody affinity maturation. Lastly, since diffuse large B cell lymphoma (DLBCL) cells derive from GC or post-GC B cells and rely on CK2 for their survival, we sought to investigate the consequences of CK2 inhibition on B cell signaling in DLBCL cells. In line with the observations in our murine model, CK2 inactivation leads to signaling defects in pathways that are essential for malignant B-lymphocyte activation.
Collapse
Affiliation(s)
- Laura Quotti Tubi
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Elisa Mandato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Sara Canovas Nunes
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Arash Arjomand
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Fortunato Zaffino
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Sabrina Manni
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Alessandro Casellato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Paolo Macaccaro
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Nicola Vitulo
- Department of Biology, Interdepartmental Research Center for Biotechnologies (CRIBI) Biotechnology Center, University of Padova, Padova, Italy
| | - Sara Zumerle
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Odile Filhol
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1036, Institute de Recherches en Technologies et Sciences pour le Vivant/Biologie du Cancer et de l’Infection, Grenoble, France
| | | | - Christian W. Siebel
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA, United States
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgio Valle
- Department of Biology, Interdepartmental Research Center for Biotechnologies (CRIBI) Biotechnology Center, University of Padova, Padova, Italy
| | | | - Stefano Casola
- IFOM-ETS-The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Valeria Cancila
- Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | | | - Claudio Tripodo
- IFOM-ETS-The AIRC Institute of Molecular Oncology, Milan, Italy,Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | - Marco Pizzi
- Department of Medicine, Cytopathology and Surgical Pathology Unit, University of Padova, Padova, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine, Cytopathology and Surgical Pathology Unit, University of Padova, Padova, Italy
| | - Livio Trentin
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Gianpietro Semenzato
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy
| | - Francesco Piazza
- Department of Medicine, Division of Hematology, University of Padova, Padova, Italy,Unit of Normal and Malignant Hematopoiesis, Laboratory of Myeloma and Lymphoma Pathobiology, Veneto of Molecular Medicine (VIMM), Padova, Italy,*Correspondence: Francesco Piazza,
| |
Collapse
|
7
|
Leung HW, Foo G, VanDongen A. Arc Regulates Transcription of Genes for Plasticity, Excitability and Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081946. [PMID: 36009494 PMCID: PMC9405677 DOI: 10.3390/biomedicines10081946] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/21/2022] [Accepted: 07/25/2022] [Indexed: 02/06/2023] Open
Abstract
The immediate early gene Arc is a master regulator of synaptic function and a critical determinant of memory consolidation. Here, we show that Arc interacts with dynamic chromatin and closely associates with histone markers for active enhancers and transcription in cultured rat hippocampal neurons. Both these histone modifications, H3K27Ac and H3K9Ac, have recently been shown to be upregulated in late-onset Alzheimer’s disease (AD). When Arc induction by pharmacological network activation was prevented using a short hairpin RNA, the expression profile was altered for over 1900 genes, which included genes associated with synaptic function, neuronal plasticity, intrinsic excitability, and signalling pathways. Interestingly, about 100 Arc-dependent genes are associated with the pathophysiology of AD. When endogenous Arc expression was induced in HEK293T cells, the transcription of many neuronal genes was increased, suggesting that Arc can control expression in the absence of activated signalling pathways. Taken together, these data establish Arc as a master regulator of neuronal activity-dependent gene expression and suggest that it plays a significant role in the pathophysiology of AD.
Collapse
Affiliation(s)
| | - Gabriel Foo
- Duke-NUS Medical School, Singapore 169857, Singapore
| | - Antonius VanDongen
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC 27710, USA
- Correspondence:
| |
Collapse
|
8
|
Bayer M, Boller S, Ramamoothy S, Zolotarev N, Cauchy P, Iwanami N, Mittler G, Boehm T, Grosschedl R. Tnpo3 enables EBF1 function in conditions of antagonistic Notch signaling. Genes Dev 2022; 36:901-915. [PMID: 36167471 PMCID: PMC9575695 DOI: 10.1101/gad.349696.122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 09/02/2022] [Indexed: 02/03/2023]
Abstract
Transcription factor EBF1 (early B cell factor 1) acts as a key regulator of B cell specification. The transcriptional network in which EBF1 operates has been extensively studied; however, the regulation of EBF1 function remains poorly defined. By mass spectrometric analysis of proteins associated with endogenous EBF1 in pro-B cells, we identified the nuclear import receptor Transportin-3 (Tnpo3) and found that it interacts with the immunoglobulin-like fold domain of EBF1. We delineated glutamic acid 271 of EBF1 as a critical residue for the association with Tnpo3. EBF1E271A showed normal nuclear localization; however, it had an impaired B cell programming ability in conditions of Notch signaling, as determined by retroviral transduction of Ebf1 -/- progenitors. By RNA-seq analysis of EBF1E271A-expressing progenitors, we found an up-regulation of T lineage determinants and down-regulation of early B genes, although similar chromatin binding of EBF1E271A and EBF1wt was detected in pro-B cells expressing activated Notch1. B lineage-specific inactivation of Tnpo3 in mice resulted in a block of early B cell differentiation, accompanied by a down-regulation of B lineage genes and up-regulation of T and NK lineage genes. Taken together, our observations suggest that Tnpo3 ensures B cell programming by EBF1 in nonpermissive conditions.
Collapse
Affiliation(s)
- Marc Bayer
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Sören Boller
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Senthilkumar Ramamoothy
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Nikolay Zolotarev
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Pierre Cauchy
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Norimasa Iwanami
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Gerhard Mittler
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Thomas Boehm
- Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
- Faculty of Medicine, University of Freiburg, 79108 Freiburg, Germany
| | - Rudolf Grosschedl
- Laboratory of Cellular and Molecular Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| |
Collapse
|
9
|
Garis M, Garrett-Sinha LA. Notch Signaling in B Cell Immune Responses. Front Immunol 2021; 11:609324. [PMID: 33613531 PMCID: PMC7892449 DOI: 10.3389/fimmu.2020.609324] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/23/2020] [Indexed: 12/22/2022] Open
Abstract
The Notch signaling pathway is highly evolutionarily conserved, dictating cell fate decisions and influencing the survival and growth of progenitor cells that give rise to the cells of the immune system. The roles of Notch signaling in hematopoietic stem cell maintenance and in specification of T lineage cells have been well-described. Notch signaling also plays important roles in B cells. In particular, it is required for specification of marginal zone type B cells, but Notch signaling is also important in other stages of B cell development and activation. This review will focus on established and new roles of Notch signaling during B lymphocyte lineage commitment and describe the function of Notch within mature B cells involved in immune responses.
Collapse
Affiliation(s)
- Matthew Garis
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| | - Lee Ann Garrett-Sinha
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
10
|
Ng HL, Taylor RL, Cheng J, Abraham LJ, Quail E, Cruickshank MN, Ulgiati D. Notch signaling induces a transcriptionally permissive state at the Complement C3d Receptor 2 (CR2) promoter in a pre-B cell model. Mol Immunol 2020; 128:150-164. [PMID: 33129017 DOI: 10.1016/j.molimm.2020.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/11/2020] [Accepted: 10/02/2020] [Indexed: 01/08/2023]
Abstract
During mammalian lymphoid development, Notch signaling is necessary at multiple stages of T lymphopoiesis, including lineage commitment, and later stages of T cell effector differentiation. In contrast, outside of a defined role in the development of splenic marginal zone B cells, there is conflicting evidence regarding whether Notch signaling plays functional roles in other B cell sub-populations. Complement receptor 2 (CR2) modulates BCR-signaling and is tightly regulated throughout differentiation. During B lymphopoiesis, CR2 is detected on immature and mature B cells with high surface expression on marginal zone B cells. Here, we have explored the possibility that Notch regulates human CR2 transcriptional activity using in vitro models including a co-culture system, co-transfection gene reporters and chromatin accessibility assays. We provide evidence that Notch signaling regulates CR2 promoter activity in a mature B cell line, as well as the induction of endogenous CR2 mRNA in a non-expressing pre-B cell line. The dynamics of endogenous gene activation suggests additional unidentified factors are required to mediate surface CR2 expression on immature and mature B lineage cells.
Collapse
Affiliation(s)
- Han Leng Ng
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia
| | - Rhonda L Taylor
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia
| | - Jessica Cheng
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia
| | - Lawrence J Abraham
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia
| | - Elizabeth Quail
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia; School of Molecular Sciences, Faculty of Science, The University of Western Australia, Australia
| | - Mark N Cruickshank
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia
| | - Daniela Ulgiati
- School of Biomedical Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, Australia.
| |
Collapse
|
11
|
Burgener AV, Bantug GR, Meyer BJ, Higgins R, Ghosh A, Bignucolo O, Ma EH, Loeliger J, Unterstab G, Geigges M, Steiner R, Enamorado M, Ivanek R, Hunziker D, Schmidt A, Müller-Durovic B, Grählert J, Epple R, Dimeloe S, Lötscher J, Sauder U, Ebnöther M, Burger B, Heijnen I, Martínez-Cano S, Cantoni N, Brücker R, Kahlert CR, Sancho D, Jones RG, Navarini A, Recher M, Hess C. SDHA gain-of-function engages inflammatory mitochondrial retrograde signaling via KEAP1-Nrf2. Nat Immunol 2019; 20:1311-1321. [PMID: 31527833 DOI: 10.1038/s41590-019-0482-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 07/31/2019] [Indexed: 12/15/2022]
Abstract
Whether screening the metabolic activity of immune cells facilitates discovery of molecular pathology remains unknown. Here we prospectively screened the extracellular acidification rate as a measure of glycolysis and the oxygen consumption rate as a measure of mitochondrial respiration in B cells from patients with primary antibody deficiency. The highest oxygen consumption rate values were detected in three study participants with persistent polyclonal B cell lymphocytosis (PPBL). Exome sequencing identified germline mutations in SDHA, which encodes succinate dehydrogenase subunit A, in all three patients with PPBL. SDHA gain-of-function led to an accumulation of fumarate in PPBL B cells, which engaged the KEAP1-Nrf2 system to drive the transcription of genes encoding inflammatory cytokines. In a single patient trial, blocking the activity of the cytokine interleukin-6 in vivo prevented systemic inflammation and ameliorated clinical disease. Overall, our study has identified pathological mitochondrial retrograde signaling as a disease modifier in primary antibody deficiency.
Collapse
Affiliation(s)
- Anne-Valérie Burgener
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Glenn R Bantug
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland.,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Benedikt J Meyer
- Immunodeficiency Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Rebecca Higgins
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Adhideb Ghosh
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland.,Competence Center for Personalized Medicine University of Zürich/Eidgenössische Technische Hochschule, Zürich, Switzerland
| | - Olivier Bignucolo
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland
| | - Eric H Ma
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Jordan Loeliger
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Gunhild Unterstab
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Marco Geigges
- Epigenomics Group, D-BSSE, Eidgenössische Technische Hochschule, Basel, Switzerland
| | - Rebekah Steiner
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Michel Enamorado
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain.,Mucosal Immunology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Washington DC, USA
| | - Robert Ivanek
- Bioinformatics Facility, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Danielle Hunziker
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Bojana Müller-Durovic
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Jasmin Grählert
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Raja Epple
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Sarah Dimeloe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Jonas Lötscher
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Ursula Sauder
- Electron Microscopy Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Monika Ebnöther
- Division of Hematology and Oncology, Claraspital, Basel, Switzerland
| | - Bettina Burger
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Ingmar Heijnen
- Division Medical Immunology, Laboratory Medicine, University Hospital Basel, Basel, Switzerland
| | - Sarai Martínez-Cano
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Nathan Cantoni
- Division of Hematology, Cantonal Hospital of Aarau, Aargau, Switzerland
| | - Rolf Brücker
- Division of Internal Medicine and Rheumatology, Hospital St. Anna, Luzern, Switzerland
| | - Christian R Kahlert
- Division of Infectious Diseases, Children's Hospital of St. Gallen, St. Gallen, Switzerland
| | - David Sancho
- Immunobiology Laboratory, entro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Russell G Jones
- Center for Cancer and Cell Biology, Van Andel Institute, Grand Rapids, MI, USA.,Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.,Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Alexander Navarini
- Division of Dermatology and Dermatology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Mike Recher
- Immunodeficiency Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland
| | - Christoph Hess
- Immunobiology Laboratory, Department of Biomedicine, University and University Hospital of Basel, Basel, Switzerland. .,Cambridge Institute of Therapeutic Immunology & Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
12
|
B cells with aberrant activation of Notch1 signaling promote Treg and Th2 cell-dominant T-cell responses via IL-33. Blood Adv 2019; 2:2282-2295. [PMID: 30213787 DOI: 10.1182/bloodadvances.2018019919] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/19/2018] [Indexed: 01/14/2023] Open
Abstract
The Notch-signaling pathway in a variety of mature B-cell neoplasms is often activated by gene alterations, but its role remains unclear. Here, we show that B cells harboring dysregulated activation of Notch1 signaling have an immunomodulatory effect on T cells by amplifying regulatory T (Treg) and T helper 2 (Th2) cell responses in an interleukin-33 (IL-33)-dependent manner. A conditional mouse model, in which constitutive expression of an active form of Notch1 is induced in B cells by Aicda gene promoter-driven Cre recombinase, revealed no obvious phenotypic changes in B cells; however, mice demonstrated an expansion of Treg and Th2 cell subsets and a decrease in cytokine production by Th1 and CD8+ T cells. The mice were susceptible to soft tissue sarcoma and defective production of CD8+ T cells specific for inoculated tumor cells, suggesting impaired antitumor T-cell activity. Gene-expression microarray revealed that altered T-cell responses were due to increased IL-33 production by Notch1-activated B cells. Knockout of IL33 or blockade of IL-33 by a receptor-blocking antibody abrogated the Treg and Th2 cell-dominant T-cell response triggered by B cells. Gene-expression data derived from human diffuse large B-cell lymphoma (DLBCL) samples showed that an activated Notch-signaling signature correlates positively with IL33 expression and Treg cell-rich gene-expression signatures. These findings indicate that B cells harboring dysregulated Notch signaling alter T-cell responses via IL-33, and suggest that aberrant activation of Notch signaling plays a role in fostering immune privilege in mature B-cell neoplasms.
Collapse
|
13
|
Wu H, Shao Q. The role of inhibitor of binding or differentiation 2 in the development and differentiation of immune cells. Immunobiology 2018; 224:142-146. [PMID: 30340915 DOI: 10.1016/j.imbio.2018.09.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/05/2018] [Accepted: 09/19/2018] [Indexed: 12/24/2022]
Abstract
Inhibitor of binding or differentiation 2 (Id2), a member of helix-loop-helix (HLH) transcriptional factors, is recently reported as an important regulator of the development or differentiation of immune cells. It has been demonstrated that Id2 plays a critical role in the early lymphopoiesis. However, it has been discovered recently that Id2 displays new functions in different immune cells. In the adaptive immune cells, Id2 is required for determining T-cell subsets and B cells. In addition, Id2 is also involved in the development of innate immune cells, including dendritic cells (DCs), natural killer (NK) cells, and other innate lymphoid cells (ILCs). Here, we review the current reports about the role of Id2 in the development or differentiation of main immune cells.
Collapse
Affiliation(s)
- Haojie Wu
- Reproductive Sciences Institute of Jiangsu University, Zhenjiang 212001, Jiangsu, P.R. China; Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China
| | - Qixiang Shao
- Reproductive Sciences Institute of Jiangsu University, Zhenjiang 212001, Jiangsu, P.R. China; Department of Immunology, Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang 212013, Jiangsu, P.R. China.
| |
Collapse
|
14
|
Yang G, Li S, Yang Y, Yuan L, Wang P, Zhao H, Ho CT, Lin CC. Nobiletin and 5-Hydroxy-6,7,8,3',4'-pentamethoxyflavone Ameliorate 12- O-Tetradecanoylphorbol-13-acetate-Induced Psoriasis-Like Mouse Skin Lesions by Regulating the Expression of Ki-67 and Proliferating Cell Nuclear Antigen and the Differentiation of CD4 + T Cells through Mitogen-Activated Protein Kinase Signaling Pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:8299-8306. [PMID: 30058806 DOI: 10.1021/acs.jafc.8b02524] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Psoriasis is a chronic and benign proliferative skin disease. Flavonoids in chenpi (aged tangerine peel) from tangerine ( Citrus reticulate Blanco), such as nobiletin (Nob), tangeretin, and 5-hydroxy-6,7,8,3',4'-pentamethoxyflavone (5-HPMF), possess anti-inflammation and regulation of immune activity among others. In this study, psoriasis-like skin lesions were induced by 12- O-tetradecanoylphorbol-13-acetate (TPA), and the preventive effect of Nob and 5-HPMF on psoriasis-like skin lesions was evaluated. Results showed that skin lesions were dramatically reduced by Nob and 5-HPMF. Levels of cytokines, including interleukin (IL)-1β, IL-17, IL-4, IL-6, tumor necrosis factor-α, and interferon-γ, were also reduced after Nob and 5-HPMF treatment. The expression levels of p-ERK1/2 and p-p38 mitogen-activated protein kinase (MAPK) in the TPA group were 5.3, 4.8, and 5.7 but downregulated to 2.7, 2.9, and 2.3 in the Nob group and 2.4, 2.7, and 1.2 in the 5-HPMF group, respectively ( p ≤ 0.05). The expression of transcription factors Ki-67 and proliferating cell nuclear antigen (PCNA) and the differentiation of CD4+ T cells were reduced by downregulating the expression of the MAPK signaling pathways. The expression levels in TPA, Nob, and 5-HPMF groups were 0.649 ± 0.094, 0.218 ± 0.034, and 0.193 ± 0.042 for Ki-67 and 0.753 ± 0.114, 0.315 ± 0.094, and 0.294 ± 0.035 for PCNA, respectively. Moreover, 5-HPMF showed stronger reduction activity in the prevention of psoriasis than Nob, indicating that the 5-hydroxyl group facilitated the suppression of psoriasis.
Collapse
Affiliation(s)
- Guliang Yang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources , Huanggang Normal University , Huanggang , Hubei 438000 , People's Republic of China
- Key Laboratory of Cultivation and Protection for Non-wood Forest Trees, Ministry of Education , Central South University of Forestry and Technology , Changsha , Hunan 410004 , People's Republic of China
| | - Shiming Li
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources , Huanggang Normal University , Huanggang , Hubei 438000 , People's Republic of China
- Key Laboratory of Cultivation and Protection for Non-wood Forest Trees, Ministry of Education , Central South University of Forestry and Technology , Changsha , Hunan 410004 , People's Republic of China
| | - Yiwen Yang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources , Huanggang Normal University , Huanggang , Hubei 438000 , People's Republic of China
| | - Li Yuan
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources , Huanggang Normal University , Huanggang , Hubei 438000 , People's Republic of China
| | - Peilei Wang
- Hubei Key Laboratory of Economic Forest Germplasm Improvement and Resources , Huanggang Normal University , Huanggang , Hubei 438000 , People's Republic of China
| | - Hui Zhao
- Tianjin Key Laboratory of Food and Biotechnology, School of Biotechnology and Food Science , Tianjin University of Commerce , Tianjin 300134 , People's Republic of China
| | - Chi-Tang Ho
- Department of Food Science , Rutgers, The State University of New Jersey , New Brunswick , New Jersey 08901 , United States
| | - Chi-Chen Lin
- Institute of Biomedical Science , National Chung-Hsing University , Taichung 402 , Taiwan
- Department of Medical Research , China Medical University Hospital , Taichung , Taiwan
| |
Collapse
|
15
|
Hwang IY, Boularan C, Harrison K, Kehrl JH. Gα i Signaling Promotes Marginal Zone B Cell Development by Enabling Transitional B Cell ADAM10 Expression. Front Immunol 2018; 9:687. [PMID: 29696016 PMCID: PMC5904254 DOI: 10.3389/fimmu.2018.00687] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
The follicular (FO) versus marginal zone (MZ) B cell fate decision in the spleen depends upon BCR, BAFF, and Notch2 signaling. Whether or how Gi signaling affects this fate decision is unknown. Here, we show that direct contact with Notch ligand expressing stromal cells (OP9-Delta-like 1) cannot promote normal MZ B cell development when progenitor B cells lack Gαi proteins, or if Gi signaling is disabled. Consistent with faulty ADAM10-dependent Notch2 processing, Gαi-deficient transitional B cells had low ADAM10 membrane expression levels and reduced Notch2 target gene expression. Immunoblotting Gαi-deficient B cell lysates revealed a reduction in mature, processed ADAM10. Suggesting that Gαi signaling promotes ADAM10 membrane expression, stimulating normal transitional B cells with CXCL12 raised it, while inhibiting Gαi nucleotide exchange blocked its upregulation. Surprisingly, inhibiting Gαi nucleotide exchange in transitional B cells also impaired the upregulation of ADAM10 that occurs following antigen receptor crosslinking. These results indicate that Gαi signaling supports ADAM10 maturation and activity in transitional B cells, and ultimately Notch2 signaling to promote MZ B cell development.
Collapse
Affiliation(s)
- Il-Young Hwang
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cedric Boularan
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States.,InvivoGen, Toulouse, France
| | - Kathleen Harrison
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - John H Kehrl
- B-Cell Molecular Immunology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
16
|
Mohammadi H, Sharafkandi N, Hemmatzadeh M, Azizi G, Karimi M, Jadidi-Niaragh F, Baradaran B, Babaloo Z. The role of innate lymphoid cells in health and disease. J Cell Physiol 2018; 233:4512-4529. [PMID: 29058773 DOI: 10.1002/jcp.26250] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/13/2017] [Indexed: 12/13/2022]
Abstract
Innate lymphoid cells (ILCs) are kind of innate immune cells which can be divided into three main subsets according to their cytokine release profile, transcription factors, and surface markers. ILCs affect the initial stages of immunity in response to microbes and participate in immunity, inflammation, and tissue repair. ILCs modulate immunity through resistance to the pathogens and regulation of autoimmune inflammation and metabolic homeostasis. Therefore dysregulation of ILCs may lead to chronic pathologies such as allergies (i.e., asthma), inflammation (i.e., inflammatory bowel disease), and autoimmunity (i.e., psoriasis, atopic dermatitis, rheumatoid arthritis, multiple sclerosis, and ankylosing spondylitis). Regarding the critical role of ILCs in the regulation of immune system, the elucidation of their function in different conditions makes an interesting target for improvement of novel therapeutic approach to modulate an immune response in different disease context.
Collapse
Affiliation(s)
- Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nadia Sharafkandi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Hemmatzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Laboratory Medicine, Imam Hassan Mojtaba Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Mohammad Karimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Siebel C, Lendahl U. Notch Signaling in Development, Tissue Homeostasis, and Disease. Physiol Rev 2017; 97:1235-1294. [PMID: 28794168 DOI: 10.1152/physrev.00005.2017] [Citation(s) in RCA: 657] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/19/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023] Open
Abstract
Notch signaling is an evolutionarily highly conserved signaling mechanism, but in contrast to signaling pathways such as Wnt, Sonic Hedgehog, and BMP/TGF-β, Notch signaling occurs via cell-cell communication, where transmembrane ligands on one cell activate transmembrane receptors on a juxtaposed cell. Originally discovered through mutations in Drosophila more than 100 yr ago, and with the first Notch gene cloned more than 30 yr ago, we are still gaining new insights into the broad effects of Notch signaling in organisms across the metazoan spectrum and its requirement for normal development of most organs in the body. In this review, we provide an overview of the Notch signaling mechanism at the molecular level and discuss how the pathway, which is architecturally quite simple, is able to engage in the control of cell fates in a broad variety of cell types. We discuss the current understanding of how Notch signaling can become derailed, either by direct mutations or by aberrant regulation, and the expanding spectrum of diseases and cancers that is a consequence of Notch dysregulation. Finally, we explore the emerging field of Notch in the control of tissue homeostasis, with examples from skin, liver, lung, intestine, and the vasculature.
Collapse
Affiliation(s)
- Chris Siebel
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Urban Lendahl
- Department of Discovery Oncology, Genentech Inc., DNA Way, South San Francisco, California; and Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
18
|
Eberl G, Colonna M, Di Santo JP, McKenzie ANJ. Innate lymphoid cells. Innate lymphoid cells: a new paradigm in immunology. Science 2015; 348:aaa6566. [PMID: 25999512 DOI: 10.1126/science.aaa6566] [Citation(s) in RCA: 622] [Impact Index Per Article: 62.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Innate lymphoid cells (ILCs) are a growing family of immune cells that mirror the phenotypes and functions of T cells. However, in contrast to T cells, ILCs do not express acquired antigen receptors or undergo clonal selection and expansion when stimulated. Instead, ILCs react promptly to signals from infected or injured tissues and produce an array of secreted proteins termed cytokines that direct the developing immune response into one that is adapted to the original insult. The complex cross-talk between microenvironment, ILCs, and adaptive immunity remains to be fully deciphered. Only by understanding these complex regulatory networks can the power of ILCs be controlled or unleashed in order to regulate or enhance immune responses in disease prevention and therapy.
Collapse
Affiliation(s)
- Gérard Eberl
- Institut Pasteur, Microenvironment and Immunity Unit, 75724 Paris, France.
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James P Di Santo
- Institut Pasteur, Innate Immunity Unit, INSERM U668, 75724 Paris, France
| | - Andrew N J McKenzie
- Medical Research Council (MRC) Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| |
Collapse
|
19
|
Abstract
Endometriosis has been associated with aberrant methylation in the eutopic endometrium. Using a genome-wide methylation array, we identified differentially methylated genes in the endometrium from women with or without endometriosis. One hundred and twenty genes were significantly altered by >1.5-fold. In all, 59 genes were significantly hypermethylated and 61 genes were significantly hypomethylated. Changes in gene expression associated with the altered methylation status were validated using quantitative real-time polymerase chain reaction. A limited number of candidate genes are selectively methylated in the endometrium of women with endometriosis. Several genes not previously associated with endometriosis are aberrantly methylated and expressed. These include O-6-methylguanine-DNA methyltransferase, dual specificity phosphatase 22, cell division cycle associated 2, inhibitor of DNA binding 2, retinoblastoma binding protein 7, bone morphogenetic protein receptor, type 1B, tumor necrosis factor receptor 1B, zinc finger protein receptor 681, immunoglobulin superfamily, member 21, and tumor protein 73. Aberrant DNA methylation and gene expression of these genes may contribute to abnormal regulation of endometrial cell proliferation and function in women.
Collapse
Affiliation(s)
- Hanyia Naqvi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT, USA
| | - Ysabel Ilagan
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT, USA
| | - Graciela Krikun
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT, USA
| |
Collapse
|
20
|
Ratliff ML, Templeton TD, Ward JM, Webb CF. The Bright Side of Hematopoiesis: Regulatory Roles of ARID3a/Bright in Human and Mouse Hematopoiesis. Front Immunol 2014; 5:113. [PMID: 24678314 PMCID: PMC3958700 DOI: 10.3389/fimmu.2014.00113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/04/2014] [Indexed: 12/12/2022] Open
Abstract
ARID3a/Bright is a DNA-binding protein that was originally discovered for its ability to increase immunoglobulin transcription in antigen-activated B cells. It interacts with DNA as a dimer through its ARID, or A/T-rich interacting domain. In association with other proteins, ARID3a increased transcription of the immunoglobulin heavy chain and led to improved chromatin accessibility of the heavy chain enhancer. Constitutive expression of ARID3a in B lineage cells resulted in autoantibody production, suggesting its regulation is important. Abnormal ARID3a expression has also been associated with increased proliferative capacity and malignancy. Roles for ARID3a in addition to interactions with the immunoglobulin locus were suggested by transgenic and knockout mouse models. Over-expression of ARID3a resulted in skewing of mature B cell subsets and altered gene expression patterns of follicular B cells, whereas loss of function resulted in loss of B1 lineage B cells and defects in hematopoiesis. More recent studies showed that loss of ARID3a in adult somatic cells promoted developmental plasticity, alterations in gene expression patterns, and lineage fate decisions. Together, these data suggest new regulatory roles for ARID3a. The genes influenced by ARID3a are likely to play pivotal roles in lineage decisions, highlighting the importance of this understudied transcription factor.
Collapse
Affiliation(s)
- Michelle L Ratliff
- Immunobiology and Cancer Research, Oklahoma Medical Research Foundation , Oklahoma City, OK , USA
| | - Troy D Templeton
- Department of Cell Biology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| | - Julie M Ward
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| | - Carol F Webb
- Immunobiology and Cancer Research, Oklahoma Medical Research Foundation , Oklahoma City, OK , USA ; Department of Cell Biology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA ; Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center , Oklahoma City, OK , USA
| |
Collapse
|
21
|
Abstract
The family of inhibitor of differentiation (Id) proteins is a group of evolutionarily conserved molecules, which play important regulatory roles in organisms ranging from Drosophila to humans. Id proteins are small polypeptides harboring a helix-loop-helix (HLH) motif, which are best known to mediate dimerization with other basic HLH proteins, primarily E proteins. Because Id proteins do not possess the basic amino acids adjacent to the HLH motif necessary for DNA binding, Id proteins inhibit the function of E protein homodimers, as well as heterodimers between E proteins and tissue-specific bHLH proteins. However, Id proteins have also been shown to have E protein-independent functions. The Id genes are broadly but differentially expressed in a variety of cell types. Transcription of the Id genes is controlled by transcription factors such as C/EBPβ and Egr as well as by signaling pathways triggered by different stimuli, which include bone morphogenic proteins, cytokines, and ligands of T cell receptors. In general, Id proteins are capable of inhibiting the differentiation of progenitors of different cell types, promoting cell-cycle progression, delaying cellular senescence, and facilitating cell migration. These properties of Id proteins enable them to play significant roles in stem cell maintenance, vasculogenesis, tumorigenesis and metastasis, the development of the immune system, and energy metabolism. In this review, we intend to highlight the current understanding of the function of Id proteins and discuss gaps in our knowledge about the mechanisms whereby Id proteins exert their diverse effects in multiple cellular processes.
Collapse
Affiliation(s)
- Flora Ling
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Bin Kang
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Xiao-Hong Sun
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.
| |
Collapse
|