1
|
Mora P, Laisné M, Bourguignon C, Rouault P, Jaspard-Vinassa B, Maître M, Gadeau AP, Renault MA, Horng S, Couffinhal T, Chapouly C. Astrocytic DLL4-NOTCH1 signaling pathway promotes neuroinflammation via the IL-6-STAT3 axis. J Neuroinflammation 2024; 21:258. [PMID: 39390606 PMCID: PMC11468415 DOI: 10.1186/s12974-024-03246-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: 07/04/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Under neuroinflammatory conditions, astrocytes acquire a reactive phenotype that drives acute inflammatory injury as well as chronic neurodegeneration. We hypothesized that astrocytic Delta-like 4 (DLL4) may interact with its receptor NOTCH1 on neighboring astrocytes to regulate astrocyte reactivity via downstream juxtacrine signaling pathways. Here we investigated the role of astrocytic DLL4 on neurovascular unit homeostasis under neuroinflammatory conditions. We probed for downstream effectors of the DLL4-NOTCH1 axis and targeted these for therapy in two models of CNS inflammatory disease. We first demonstrated that astrocytic DLL4 is upregulated during neuroinflammation, both in mice and humans, driving astrocyte reactivity and subsequent blood-brain barrier permeability and inflammatory infiltration. We then showed that the DLL4-mediated NOTCH1 signaling in astrocytes directly drives IL-6 levels, induces STAT3 phosphorylation promoting upregulation of astrocyte reactivity markers, pro-permeability factor secretion and consequent blood-brain barrier destabilization. Finally we revealed that blocking DLL4 with antibodies improves experimental autoimmune encephalomyelitis symptoms in mice, identifying a potential novel therapeutic strategy for CNS autoimmune demyelinating disease. As a general conclusion, this study demonstrates that DLL4-NOTCH1 signaling is not only a key pathway in vascular development and angiogenesis, but also in the control of astrocyte reactivity during neuroinflammation.
Collapse
Affiliation(s)
- Pierre Mora
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Margaux Laisné
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Célia Bourguignon
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Paul Rouault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Béatrice Jaspard-Vinassa
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marlène Maître
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, F-33000, France
| | - Alain-Pierre Gadeau
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marie-Ange Renault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Sam Horng
- Department of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Thierry Couffinhal
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Candice Chapouly
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France.
| |
Collapse
|
2
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
3
|
Guindi C, Khan FU, Cloutier A, Khongorzul P, Raki AA, Gaudreau S, McDonald PP, Gris D, Amrani A. Inhibition of PI3K/C/EBPβ axis in tolerogenic bone marrow-derived dendritic cells of NOD mice promotes Th17 differentiation and diabetes development. Transl Res 2022; 255:37-49. [PMID: 36400308 DOI: 10.1016/j.trsl.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Dendritic cells (DCs) are key regulators of the adaptive immune response. Tolerogenic dendritic cells play a crucial role in inducing and maintaining immune tolerance in autoimmune diseases such as type 1 diabetes in humans as well as in the NOD mouse model. We previously reported that bone marrow-derived DCs (BM.DCs) from NOD mice, generated with a low dose of GM-CSF (GM/DCs), induce Treg differentiation and are able to protect NOD mice from diabetes. We had also found that the p38 MAPK/C/EBPβ axis is involved in regulating the phenotype, as well as the production of IL-10 and IL-12p70, by tolerogenic GM/DCs. Here, we report that the inhibition of the PI3K signaling switched the cytokine profile of GM/DCs toward Th17-promoting cytokines without affecting their phenotype. PI3K inhibition abrogated the production of IL-10 by GM/DCs, whereas it enhanced their production of IL-23 and TGFβ. Inhibition of PI3K signaling in tolerogenic GM/DCs also induced naive CD4+ T cells differentiation toward Th17 cells. Mechanistically, PI3K inhibition increased the DNA-binding activity of C/EBPβ through a GSK3-dependent pathway, which is important to maintain the semimature phenotype of tolerogenic GM/DCs. Furthermore, analysis of C/EBPβ-/- GM/DCs demonstrated that C/EBPβ is required for IL-23 production. Of physiological relevance, the level of protection from diabetes following transfusion of GM/DCs into young NOD mice was significantly reduced when NOD mice were transfused with GM/DCs pretreated with a PI3K inhibitor. Our data suggest that PI3K/C/EBPβ signaling is important in controlling tolerogenic function of GM/DCs by limiting their Th17-promoting cytokines.
Collapse
Affiliation(s)
- Chantal Guindi
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Farhan Ullah Khan
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Alexandre Cloutier
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Puregmaa Khongorzul
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Ahmed Aziz Raki
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Simon Gaudreau
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Patrick P McDonald
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Denis Gris
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada
| | - Abdelaziz Amrani
- Department of Pediatrics, Immunology Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke and Centre de Recherche du CHUS, Sherbrooke, Quebec, Canada.
| |
Collapse
|
4
|
Croft CA, Thaller A, Marie S, Doisne JM, Surace L, Yang R, Puel A, Bustamante J, Casanova JL, Di Santo JP. Notch, RORC and IL-23 signals cooperate to promote multi-lineage human innate lymphoid cell differentiation. Nat Commun 2022; 13:4344. [PMID: 35896601 PMCID: PMC9329340 DOI: 10.1038/s41467-022-32089-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 07/13/2022] [Indexed: 11/09/2022] Open
Abstract
Innate lymphoid cells (ILCs) include cytotoxic natural killer cells and distinct groups of cytokine-producing innate helper cells which participate in immune defense and promote tissue homeostasis. Circulating human ILC precursors (ILCP) able to generate all canonical ILC subsets via multi-potent or uni-potent intermediates according to our previous work. Here we show potential cooperative roles for the Notch and IL-23 signaling pathways for human ILC differentiation from blood ILCP using single cell cloning analyses and validate these findings in patient samples with rare genetic deficiencies in IL12RB1 and RORC. Mechanistically, Notch signaling promotes upregulation of the transcription factor RORC, enabling acquisition of Group 1 (IFN-γ) and Group 3 (IL-17A, IL-22) effector functions in multi-potent and uni-potent ILCP. Interfering with RORC or signaling through its target IL-23R compromises ILC3 effector functions but also generally suppresses ILC production from multi-potent ILCP. Our results identify a Notch->RORC- > IL-23R pathway which operates during human ILC differentiation. These observations may help guide protocols to expand functional ILC subsets in vitro with an aim towards novel ILC therapies for human disease.
Collapse
Affiliation(s)
- Carys A Croft
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Anna Thaller
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Solenne Marie
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Jean-Marc Doisne
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Laura Surace
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France
| | - Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, UMR 1163, Paris, France.,Imagine Institute, Université Paris Cité, Paris, France.,Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, UMR 1163, Paris, France.,Imagine Institute, Université Paris Cité, Paris, France.,Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM, UMR 1163, Paris, France.,Imagine Institute, Université Paris Cité, Paris, France.,Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France.,Howard Hughes Medical Institute, New York, NY, USA
| | - James P Di Santo
- Institut Pasteur, Université Paris Cité, Inserm U1223, Innate Immunity Unit, Paris, France.
| |
Collapse
|
5
|
Allen F, Maillard I. Therapeutic Targeting of Notch Signaling: From Cancer to Inflammatory Disorders. Front Cell Dev Biol 2021; 9:649205. [PMID: 34124039 PMCID: PMC8194077 DOI: 10.3389/fcell.2021.649205] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
Over the past two decades, the Notch signaling pathway has been investigated as a therapeutic target for the treatment of cancers, and more recently in the context of immune and inflammatory disorders. Notch is an evolutionary conserved pathway found in all metazoans that is critical for proper embryonic development and for the postnatal maintenance of selected tissues. Through cell-to-cell contacts, Notch orchestrates cell fate decisions and differentiation in non-hematopoietic and hematopoietic cell types, regulates immune cell development, and is integral to shaping the amplitude as well as the quality of different types of immune responses. Depriving some cancer types of Notch signals has been shown in preclinical studies to stunt tumor growth, consistent with an oncogenic function of Notch signaling. In addition, therapeutically antagonizing Notch signals showed preclinical potential to prevent or reverse inflammatory disorders, including autoimmune diseases, allergic inflammation and immune complications of life-saving procedures such allogeneic bone marrow and solid organ transplantation (graft-versus-host disease and graft rejection). In this review, we discuss some of these unique approaches, along with the successes and challenges encountered so far to target Notch signaling in preclinical and early clinical studies. Our goal is to emphasize lessons learned to provide guidance about emerging strategies of Notch-based therapeutics that could be deployed safely and efficiently in patients with immune and inflammatory disorders.
Collapse
Affiliation(s)
- Frederick Allen
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Ivan Maillard
- Division of Hematology and Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
6
|
Deng Y, Chen S, Song S, Huang Y, Chen R, Tao A. Anti-DLL4 ameliorates toluene diisocyanate-induced experimental asthma by inhibiting Th17 response. Int Immunopharmacol 2021; 94:107444. [PMID: 33578263 DOI: 10.1016/j.intimp.2021.107444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 11/22/2022]
Abstract
Toluene diisocyanate (TDI) exhibits an ability to induce steroid insensitive asthma with the involvement of Th17 cells. And emerging evidence has indicated that DLL4 signaling promotes Th17 differentiation through directly upregulating Rorc and IL-17 transcription. Thus, we sought to evaluate the effects of DLL4 blocking antibody on TDI-induced asthma model. Female BALB/c mice were sensitized and challenged with TDI to generate an asthma model. TDI-exposed mice were intraperitoneally injected with anti-DLL4 antibody and then analyzed for various parameters of the airway inflammatory responses. Increased expression of DLL4 in spleen and lung was detected in TDI-exposed mice. Furthermore, anti-DLL4 treatment alleviated TDI-induced airway hyperreactivity (AHR), airway inflammation, airway epithelial injury and airway smooth muscle (ASM) thickening. In the meantime, neutralizing DLL4 also blunted Th17 response via downregulation of ROR-γt expression, while had no effect on Th2 cells and regulatory T (Treg) cells. Overall, anti-DLL4 ameliorated TDI-induced experimental asthma by inhibiting Th17 response, implying the feasibility of targeting DLL4 for therapy of Th17-predominant severe asthma.
Collapse
Affiliation(s)
- Yao Deng
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China
| | - Shuyu Chen
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China; Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Shijie Song
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China
| | - Yin Huang
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China
| | - Rongchang Chen
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen 518020, China
| | - Ailin Tao
- The Second Affiliated Hospital, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 510260, China.
| |
Collapse
|
7
|
Salazar JL, Yang SA, Yamamoto S. Post-Developmental Roles of Notch Signaling in the Nervous System. Biomolecules 2020; 10:biom10070985. [PMID: 32630239 PMCID: PMC7408554 DOI: 10.3390/biom10070985] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/25/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Since its discovery in Drosophila, the Notch signaling pathway has been studied in numerous developmental contexts in diverse multicellular organisms. The role of Notch signaling in nervous system development has been extensively investigated by numerous scientists, partially because many of the core Notch signaling components were initially identified through their dramatic ‘neurogenic’ phenotype of developing fruit fly embryos. Components of the Notch signaling pathway continue to be expressed in mature neurons and glia cells, which is suggestive of a role in the post-developmental nervous system. The Notch pathway has been, so far, implicated in learning and memory, social behavior, addiction, and other complex behaviors using genetic model organisms including Drosophila and mice. Additionally, Notch signaling has been shown to play a modulatory role in several neurodegenerative disease model animals and in mediating neural toxicity of several environmental factors. In this paper, we summarize the knowledge pertaining to the post-developmental roles of Notch signaling in the nervous system with a focus on discoveries made using the fruit fly as a model system as well as relevant studies in C elegans, mouse, rat, and cellular models. Since components of this pathway have been implicated in the pathogenesis of numerous psychiatric and neurodegenerative disorders in human, understanding the role of Notch signaling in the mature brain using model organisms will likely provide novel insights into the mechanisms underlying these diseases.
Collapse
Affiliation(s)
- Jose L. Salazar
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Sheng-An Yang
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, TX 77030, USA; (J.L.S.); (S.-A.Y.)
- Department of Neuroscience, BCM, Houston, TX 77030, USA
- Program in Developmental Biology, BCM, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, BCM, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-832-824-8119
| |
Collapse
|
8
|
Cummings M, Arumanayagam ACS, Zhao P, Kannanganat S, Stuve O, Karandikar NJ, Eagar TN. Presenilin1 regulates Th1 and Th17 effector responses but is not required for experimental autoimmune encephalomyelitis. PLoS One 2018; 13:e0200752. [PMID: 30089166 PMCID: PMC6082653 DOI: 10.1371/journal.pone.0200752] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/02/2018] [Indexed: 02/02/2023] Open
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) where pathology is thought to be regulated by autoreactive T cells of the Th1 and Th17 phenotype. In this study we sought to understand the functions of Presenilin 1 (PSEN1) in regulating T cell effector responses in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. PSEN1 is the catalytic subunit of γ-secretase a multimolecular protease that mediates intramembranous proteolysis. γ-secretase is known to regulate several pathways of immune importance. Here we examine the effects of disrupting PSEN1 functions on EAE and T effector differentiation using small molecule inhibitors of γ-secretase (GSI) and T cell-specific conditional knockout mice (PSEN1 cKO). Surprisingly, blocking PSEN1 function by GSI treatment or PSEN1 cKO had little effect on the development or course of MOG35-55-induced EAE. In vivo GSI administration reduced the number of myelin antigen-specific T cells and suppressed Th1 and Th17 differentiation following immunization. In vitro, GSI treatment inhibited Th1 differentiation in neutral but not IL-12 polarizing conditions. Th17 differentiation was also suppressed by the presence of GSI in all conditions and GSI-treated Th17 T cells failed to induce EAE following adoptive transfer. PSEN cKO T cells showed reduced Th1 and Th17 differentiation. We conclude that γ-secretase and PSEN1-dependent signals are involved in T effector responses in vivo and potently regulate T effector differentiation in vitro, however, they are dispensable for EAE.
Collapse
MESH Headings
- Amyloid Precursor Protein Secretases/antagonists & inhibitors
- Amyloid Precursor Protein Secretases/metabolism
- Animals
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Dibenzazepines/pharmacology
- Dibenzazepines/therapeutic use
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/metabolism
- Interleukin-17/metabolism
- Interleukin-2/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Presenilin-1/deficiency
- Presenilin-1/genetics
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
Collapse
Affiliation(s)
- Matthew Cummings
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States of America
| | | | - Picheng Zhao
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital Research Institute, Houston, TX, United States of America
| | - Sunil Kannanganat
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital Research Institute, Houston, TX, United States of America
| | - Olaf Stuve
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, United States of America
| | - Nitin J. Karandikar
- Department of Pathology, University of Iowa, Iowa City, IA, United States of America
| | - Todd N. Eagar
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital Research Institute, Houston, TX, United States of America
| |
Collapse
|
9
|
Anti-Dll4 Antibody Inhibits the Differentiation of Th17 Cells in Asthmatic Mice. Inflammation 2018; 40:1975-1982. [PMID: 28812183 DOI: 10.1007/s10753-017-0638-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
T helper 17 (Th17) cells play an important role in allergic asthma, and the Notch ligand Delta-like ligand (Dll)4 has been reported to direct the differentiation of Th17 cells. In this study, experimental animals were divided into five groups (control group, asthma group, physiological saline group, anti-Dll4 antibody group, and immunoglobulin G group). The study aimed to explore the effect of anti-Dll4 antibody on the differentiation of Th17 cell in asthmatic mice. Dll4 protein expressions were performed by immunohistochemical imaging. The proportion of Th17 cells in mouse spleen-isolated CD4+ T cells were measured by flow cytometry. The protein expression of Th17 transcription factor retinoid-related orphan nuclear receptor (RORγt) was detected by Western blotting. Interleukin (IL)-17 levels in serum were measured by enzyme-linked immunosorbent assay (ELISA). The study found that the expression of Dll4 in lung tissue from the asthma group significantly increased compared with the anti-Dll4 antibody group. The ratio of Th17 cells in CD4+ T cells was significantly downregulated, and the protein expression of RORγt in spleen significantly reduced in the anti-Dll4 antibody group compared with the asthma group. Moreover, the IL-17 level in serum from the anti-Dll4 antibody group significantly reduced compared with the asthma group. These results suggested that anti-Dll4 antibody could inhibit the differentiation of Th17 cells in asthmatic mice.
Collapse
|
10
|
Jiang S, Han S, Chen J, Li X, Che H. Inhibition effect of blunting Notch signaling on food allergy through improving T H1/T H2 balance in mice. Ann Allergy Asthma Immunol 2017; 118:94-102. [PMID: 28007091 DOI: 10.1016/j.anai.2016.10.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 10/20/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022]
Abstract
BACKGROUND Notch signaling regulates proliferation, differentiation, and function of dendritic cells, T cells, and mast cells, as well as many other immune cells, which act as important parts in food allergy, Notch signaling may play an important role in food allergy. OBJECTIVE To investigate the role of Notch signaling in IgE-mediated food allergy. METHODS An ovalbumin-induced food allergy mouse model was built (cholera toxin as adjuvant) and Notch signaling was blunted by FLI-06 and MW167, which inhibited Notch receptor-expressing phase and the γ-secretase-affecting phase, respectively. Then food allergy indicators, including levels of serum antibodies, cytokines, and degranulation, were examined. Meanwhile, clinical features, such as vascular permeability changes, intestinal permeability changes, body temperature changes, and symptoms, were also observed. RESULTS After blunting Notch signaling, the levels of serum ovalbumin specific IgE and IgG1 were decreased significantly, suggesting that blunting Notch signaling inhibited antibody responses. The levels of TH1 cytokines (interferon-γ) were increased significantly, whereas the levels of TH2 cytokines (interleukin-4, -5, and -13) were decreased significantly, suggesting TH2 polarization was suppressed after blunting Notch signaling. The expression of T-bet was significantly increased, whereas the expression of Gata-3 was significantly reduced in both messenger RNA and protein levels, indicating TH2 polarization was inhibited and TH1 polarization was enhanced after blunting Notch signaling. Moreover, allergic clinical features of mice were alleviated after blunting Notch signaling. CONCLUSION Food allergy was inhibited by blunting Notch signaling through suppressing TH2 polarization, enhancing TH1 cell differentiation and promoting TH1/TH2 balance in mice. Notch signaling plays a key role in IgE-mediated food allergy.
Collapse
Affiliation(s)
- Songsong Jiang
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Shiwen Han
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Jingyu Chen
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Xuejiao Li
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China
| | - Huilian Che
- College of Food Science and Nutrition Engineering, China Agricultural University, Beijing, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China.
| |
Collapse
|
11
|
Tindemans I, Peeters MJW, Hendriks RW. Notch Signaling in T Helper Cell Subsets: Instructor or Unbiased Amplifier? Front Immunol 2017; 8:419. [PMID: 28458667 PMCID: PMC5394483 DOI: 10.3389/fimmu.2017.00419] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/24/2017] [Indexed: 11/16/2022] Open
Abstract
For protection against pathogens, it is essential that naïve CD4+ T cells differentiate into specific effector T helper (Th) cell subsets following activation by antigen presented by dendritic cells (DCs). Next to T cell receptor and cytokine signals, membrane-bound Notch ligands have an important role in orchestrating Th cell differentiation. Several studies provided evidence that DC activation is accompanied by surface expression of Notch ligands. Intriguingly, DCs that express the delta-like or Jagged Notch ligands gain the capacity to instruct Th1 or Th2 cell polarization, respectively. However, in contrast to this model it has also been hypothesized that Notch signaling acts as a general amplifier of Th cell responses rather than an instructive director of specific T cell fates. In this alternative model, Notch enhances proliferation, cytokine production, and anti-apoptotic signals or promotes co-stimulatory signals in T cells. An instructive role for Notch ligand expressing DCs in the induction of Th cell differentiation is further challenged by evidence for the involvement of Notch signaling in differentiation of Th9, Th17, regulatory T cells, and follicular Th cells. In this review, we will discuss the two opposing models, referred to as the “instructive” and the “unbiased amplifier” model. We highlight both the function of different Notch receptors on CD4+ T cells and the impact of Notch ligands on antigen-presenting cells.
Collapse
Affiliation(s)
- Irma Tindemans
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | | | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
12
|
Qin L, Zhou YC, Wu HJ, Zhuo Y, Wang YP, Si CY, Qin YM. Notch Signaling Modulates the Balance of Regulatory T Cells and T Helper 17 Cells in Patients with Chronic Hepatitis C. DNA Cell Biol 2017; 36:311-320. [PMID: 28157396 DOI: 10.1089/dna.2016.3609] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The imbalance of regulatory T cells (Tregs) and T helper 17 (Th17) cells contributes to the persistent hepatitis C virus (HCV) infection. However, modulatory factors associated with Tregs-Th17 balance were not fully elucidated. A recent study demonstrated an immunoregulatory strategy by inactivation of Notch signaling to reverse the disequilibrium of Tregs-Th17 cells in immune thrombocytopenia. Thus, the aim of this study was to assess the effect of Notch signaling in regulating the functions of Tregs and Th17 cells in chronic hepatitis C. A total of 46 patients with chronic hepatitis C and 17 normal controls (NCs) were enrolled. mRNA expressions of Notch1 and Notch2 were semiquantified by real-time reserve polymerase chain reaction. Percentages of Tregs-Th17, levels of key transcriptional factors, and cytokine productions were measured in response to treatment by DAPT, a γ-secretase inhibitor to suppress Notch signaling. We found that Notch1 and Notch2 mRNAs were significantly elevated in peripheral blood mononuclear cells from chronic hepatitis C patients compared with those from NCs. DAPT treatment reduced Th17 response by downregulation of RORγt expression and interleukin (IL)-17/IL-22 secretion. Tregs proportion, FoxP3 expression, and IL-10 production did not change significantly with DAPT treatment in chronic hepatitis C; however, blockage of Notch signaling inhibited the suppressive function of Tregs. Moreover, effective anti-HCV therapy not only reduced Notch1 and Notch2 expression but also decreased Tregs and Th17 proportions. The current data provided a novel mechanism underlying the modulation of Treg-Th17 balance. The link between Notch signaling and Th cells might lead to a new intervention for breaking immunotolerance of chronic HCV infection.
Collapse
Affiliation(s)
- Lei Qin
- 1 Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Yan-Cai Zhou
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Hong-Jie Wu
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Ya Zhuo
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Yan-Ping Wang
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Chang-Yun Si
- 2 Department of Infectious Diseases, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| | - Yong-Mei Qin
- 1 Department of Gastroenterology, The First Affiliated Hospital of Xinxiang Medical University , Weihui, Xinxiang, China
| |
Collapse
|
13
|
Shen C, Detry B, Lecocq M, Pilette C. A novel IgA/Delta-like 4/Notch axis induces immunosuppressive activity in human dendritic cells. Clin Immunol 2016; 168:37-46. [PMID: 27117596 DOI: 10.1016/j.clim.2016.04.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 04/20/2016] [Accepted: 04/22/2016] [Indexed: 01/17/2023]
Affiliation(s)
- Chong Shen
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Bruno Detry
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Marylène Lecocq
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium
| | - Charles Pilette
- Université catholique de Louvain (UCL), Institut de Recherche Expérimentale & Clinique (IREC), Pôle Pneumologie, ORL & dermatologie; Institute for Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Cliniques universitaires St-Luc, Brussels, Belgium.
| |
Collapse
|
14
|
Jiang Y, Zhao S, Yang X, Liu Y, Wang C. Dll4 in the DCs isolated from OVA-sensitized mice is involved in Th17 differentiation inhibition by 1,25-dihydroxyvitamin D3 in vitro. J Asthma 2015; 52:989-95. [PMID: 26333305 DOI: 10.3109/02770903.2015.1056349] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION T helper 17 cell (Th17) cells play an important role in neutrophilic asthma, and 1,25(OH)2D3 has been reported to modulate the proliferation and differentiation of T cells. In this study, we examined the effects of 1,25(OH)2D3 on the dendritic cell (DC)-mediated regulation of Th17differentiation from OVA-sensitized mice. METHODS DCs were isolated from ovalbumin-sensitized mouse spleens. Lipopolysaccharide (LPS) was administered to stimulate the DCs for 24 h, and dexamethasone or 1,25(OH)2D3 was applied simultaneously. The expression of Notch ligand delta-like ligand 4 (Dll4) in the DCs was detected in each group. All the groups of treated DCs were co-cultured with T cells, and Dll4 was inhibited in these groups. After 24 h, Th17 and Treg cell differentiation and the IL-17A levels were measured. RESULTS Dll4 expression was increased in LPS-treated DCs compared with the control group (p = 0.05), resulting in increased Th17 cell differentiation (p = 0.002). Treatment with 1,25(OH)2D3 inhibited the Dll4 expression(p = 0.04) and decreased Th17 cell differentiation (p = 0.001) in DCs that was induced by LPS. Directly inhibiting Dll4 reduced Th17 cell differentiation, and Th17 cell differentiation was not further inhibited by 1,25(OH)2D3 once Dll4 was blocked. CONCLUSIONS These result suggest that Dll4 in the DCs isolated from OVA-sensitized mice is involved in Th17 differentiation inhibition by 1,25(OH)2D3.
Collapse
Affiliation(s)
- Yunqiu Jiang
- a Institute of Human Respiratory Disease, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Shengtao Zhao
- a Institute of Human Respiratory Disease, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Xu Yang
- a Institute of Human Respiratory Disease, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Yu Liu
- a Institute of Human Respiratory Disease, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Changzheng Wang
- a Institute of Human Respiratory Disease, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| |
Collapse
|
15
|
Juryńczyk M, Lewkowicz P, Domowicz M, Mycko MP, Selmaj KW. Heat shock protein 70 (Hsp70) interacts with the Notch1 intracellular domain and contributes to the activity of Notch signaling in myelin-reactive CD4 T cells. J Neuroimmunol 2015; 287:19-26. [PMID: 26439956 DOI: 10.1016/j.jneuroim.2015.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/28/2015] [Accepted: 08/04/2015] [Indexed: 10/23/2022]
Abstract
Notch receptors (Notch1-4) are involved in the differentiation of CD4 T cells and the development of autoimmunity. Mechanisms regulating Notch signaling in CD4 T cells are not fully elucidated. In this study we investigated potential crosstalk between Notch pathway molecules and heat shock protein 70 (Hsp70), the major intracellular chaperone involved in the protein transport during immune responses and other stress conditions. Using Hsp70(-/-) mice we found that Hsp70 is critical for up-regulation of NICD1 and induction of Notch target genes in Jagged1- and Delta-like1-stimulated CD4 T cells. Co-immunoprecipitation analysis of wild-type CD4 T cells stimulated with either Jagged1 or Delta-like1 showed a direct interaction between NICD1 and Hsp70. Both molecules co-localized within the nucleus of CD4 T cells stimulated with Notch ligands. Molecular interaction and nuclear colocalization of NICD1 and Hsp70 were also detected in CD4 T cells reactive against myelin oligodendrocyte glycoprotein (MOG)35-55, which showed Hsp70-dependent up-regulation of both NICD1 and Notch target genes. In conclusion, we demonstrate for the first time that Hsp70 interacts with NICD1 and contributes to the activity of Notch signaling in CD4 T cells. Interaction between Hsp70 and NICD1 may represent a novel mechanism regulating Notch signaling in activated CD4 T cells.
Collapse
Affiliation(s)
- Maciej Juryńczyk
- Department of Neurology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Przemysław Lewkowicz
- Department of Neurology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Małgorzata Domowicz
- Department of Neurology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Marcin P Mycko
- Department of Neurology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland
| | - Krzysztof W Selmaj
- Department of Neurology, Medical University of Lodz, Kopcińskiego 22, 90-153 Lodz, Poland.
| |
Collapse
|
16
|
Expression of Notch signaling components in cutaneous foreign body and sarcoidal granulomas and fusing macrophages. Am J Dermatopathol 2014; 36:409-13. [PMID: 24394305 DOI: 10.1097/dad.0b013e3182a730cb] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The evolutionarily conserved Notch signaling pathway affects tissue-specific cell differentiation, proliferation, and apoptosis. In the immune system, Notch has been implicated in the development and function of both adoptive and innate immune cells. Notch signaling is initiated by Notch receptor binding to cognate ligands, which results in the enzymatic cleavage and intranuclear translocation of the intracellular domain of Notch receptor (ICN). Recent murine models of chronic inflammation highlighted a critical role for a Notch ligand, Delta-like ligand (Dll)-4, in granuloma formation. In this study, we aimed to assess Notch-1 receptor activation and Dll4 expression in human cutaneous granulomas and in cultured human macrophages and multinucleated giant cells. ICN1 and Dll4 expression was evaluated by immunohistochemistry of cutaneous foreign body (n = 15) and sarcoidal (n = 19) granulomas. The results showed consistent intranuclear staining for ICN1 in foreign body but not in sarcoidal granulomas and strong cytoplasmic staining for Dll4 in mononuclear histiocytes and multinucleate giant cells in both types of granulomas. Additionally, immunofluorescence confocal microscopy showed ICN1 and Dll4 expression by cultured human macrophages undergoing fusion in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4. These findings indicate a potential role for the Notch-1-Dll4 signaling pathway in foreign body-induced granulomatous reactions and possibly distinct Notch pathway utilization in sarcoidal granulomas.
Collapse
|
17
|
Protective effect of tanreqing injection on axon myelin damage in the brain of mouse model for experimental autoimmune encephalomyelitis. J TRADIT CHIN MED 2014; 34:576-83. [PMID: 25417409 DOI: 10.1016/s0254-6272(15)30066-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To evaluate the effect of Tanreqing injection on axon myelin in the mouse brain of experimental autoimmune encephalomyelitis (EAE). METHODS An EAE model was established by myelin oligodendrocyte glycoprotein (MOG)35-55 immunization in C57BL/6 mice. Mice were randomly divided into the following groups: normal, model, prednisone acetate (PA) (6 mg/kg), Tanreqing high dose (5.14 mL/kg), Tanreqing low dose (2.57 mL/kg). On the day of immunization, both Tanreqing groups were treated by intraperitoneal injection, with the PA group treated by intragastrical perfusion after T cell response, and the other groups treated with saline. Changes in body weight, neurological deficit score, incidence rate, mortality rate, and course of disease were observed for all mice. Brain tissue was isolated and stained with hematoxylin-eosin, and pathological investigations performed to evaluate axon myelin damage by transmission electron microscopy (TEM). Myelin basic protein and microtubule associated protein-2 were analyzed by immunohistochemistry. RESULTS Tanreqing injection significantly prolonged EAE latency and decreased the neurological deficit score, alleviated infiltration of inflammatory cells in the focus area, up-regulated hippocampal MBP expression at the acute stage and the remission stage, and increased microtubule associated protein-2 expression in the EAE brain to varying degrees in the acute stage. TEM analysis indicated that Tanreqing injection alleviates myelin damage in the EAE mouse and maintains the integrity of circular layer structures and alleviates axon mitochondrial swelling. CONCLUSION Tanreqing injection alleviates EAE symptoms.
Collapse
|
18
|
Sandy AR, Stoolman J, Malott K, Pongtornpipat P, Segal BM, Maillard I. Notch signaling regulates T cell accumulation and function in the central nervous system during experimental autoimmune encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2013; 191:1606-13. [PMID: 23825310 DOI: 10.4049/jimmunol.1301116] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Systemic inhibition of Notch signaling was previously shown to attenuate experimental autoimmune encephalomyelitis (EAE), a disease model of multiple sclerosis in mice. Different studies attributed these effects to decreased T-bet and IFN-γ expression, enhanced regulatory T cell function, reduced T cell chemotaxis to the CNS, or impaired Th9 cell differentiation. Interpretation of these heterogeneous findings is difficult because past experimental strategies did not ensure complete Notch inhibition in T cells and because many cell populations could be affected by systemic Notch blockade. To resolve the role of Notch in T cells during EAE, we used the pan-Notch inhibitor dominant-negative form of Mastermind-like 1 (DNMAML), as well as several complementary loss-of-function approaches specifically in myelin-reactive T cells. Notch inhibition in T cells profoundly decreased EAE incidence and severity. Notch-deprived myelin-reactive T cells had preserved activation and effector differentiation in secondary lymphoid tissues. However, Notch-deprived T cells failed to accumulate in the CNS after immunization. Parking wild-type and DNMAML T cells together in bone marrow chimeras increased accumulation of Notch-deprived T cells in the CNS after immunization but did not prevent EAE, indicating the absence of dominant suppression by DNMAML T cells. Analysis of CNS-infiltrating DNMAML T cells revealed markedly defective IL-17A and IFN-γ production, despite preserved T-bet expression. Collectively, our findings capture the profound overall effects of Notch signaling in myelin-reactive T cells and demonstrate that Notch controls the accumulation and pathogenic functions of CD4(+) T cells within their target organ but not in lymphoid tissues during EAE.
Collapse
Affiliation(s)
- Ashley R Sandy
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|