1
|
Musai J, Mammen AL, Pinal-Fernandez I. Recent Updates on the Pathogenesis of Inflammatory Myopathies. Curr Rheumatol Rep 2024; 26:421-430. [PMID: 39316320 PMCID: PMC11527972 DOI: 10.1007/s11926-024-01164-7] [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] [Accepted: 09/04/2024] [Indexed: 09/25/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide a comprehensive and updated overview of autoimmune myopathies, with a special focus on the latest advancements in understanding the role of autoantibodies. We will begin by examining the risk factors and triggers associated with myositis. Next, we will delve into recent research on how autoantibodies contribute to disease pathogenesis. Finally, we will explore the latest innovations in treatment strategies and their implications for our understanding of myositis pathogenesis. RECENT FINDINGS Recent research has revealed that myositis-specific autoantibodies can infiltrate muscle cells and disrupt the function of their target autoantigens, playing a crucial role in disease pathogenesis. Significant advances in treatment include CD19 CAR-T cell therapy, JAK-STAT inhibitors, and novel strategies targeting the type 1 interferon pathway in dermatomyositis. Additionally, the ineffectiveness of complement inhibitors in treating immune-mediated necrotizing myositis has challenged established views on disease mechanisms. Autoimmune myopathies are a collection of disorders significantly influenced by specific autoantibodies that drive disease pathogenesis. This review highlights the critical role of autoantibody research in deepening our understanding of these conditions and discusses recent therapeutic advancements targeting key pathogenic pathways.
Collapse
Affiliation(s)
- Jon Musai
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Room 1141, Building 50, MSC 8024, Bethesda, MD, 20892, USA
| | - Andrew L Mammen
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Room 1141, Building 50, MSC 8024, Bethesda, MD, 20892, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Iago Pinal-Fernandez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Drive, Room 1141, Building 50, MSC 8024, Bethesda, MD, 20892, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Pinal-Fernandez I, Muñoz-Braceras S, Casal-Dominguez M, Pak K, Torres-Ruiz J, Musai J, Dell'Orso S, Naz F, Islam S, Gutierrez-Cruz G, Cano MD, Matas-Garcia A, Padrosa J, Tobias-Baraja E, Garrabou G, Aldecoa I, Espinosa G, Simeon-Aznar CP, Guillen-Del-Castillo A, Gil-Vila A, Trallero-Araguás E, Christopher-Stine L, Lloyd TE, Liewluck T, Naddaf E, Stenzel W, Greenberg SA, Grau JM, Selva-O'Callaghan A, Milisenda JC, Mammen AL. Pathological autoantibody internalisation in myositis. Ann Rheum Dis 2024; 83:1549-1560. [PMID: 38902010 PMCID: PMC11493519 DOI: 10.1136/ard-2024-225773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVES Autoantibodies targeting intracellular proteins are common in various autoimmune diseases. In the context of myositis, the pathologic significance of these autoantibodies has been questioned due to the assumption that autoantibodies cannot enter living muscle cells. This study aims to investigate the validity of this assumption. METHODS Confocal immunofluorescence microscopy was employed to localise antibodies and other proteins of interest in myositis muscle biopsies. Bulk RNA sequencing was used to examine the transcriptomic profiles of 669 samples, including those from patients with myositis, disease controls and healthy controls. Additionally, antibodies from myositis patients were introduced into cultured myoblasts through electroporation, and their transcriptomic profiles were analysed using RNA sequencing. RESULTS In patients with myositis autoantibodies, antibodies accumulated inside myofibres in the same subcellular compartment as the autoantigen. Bulk RNA sequencing revealed that muscle biopsies from patients with autoantibodies targeting transcriptional regulators exhibited transcriptomic patterns consistent with dysfunction of the autoantigen. For instance, in muscle biopsies from patients with anti-PM/Scl autoantibodies recognising components of the nuclear RNA exosome complex, an accumulation of divergent transcripts and long non-coding RNAs was observed; these RNA forms are typically degraded by the nuclear RNA exosome complex. Introducing patient antibodies into cultured muscle cells recapitulated the transcriptomic effects observed in human disease. Further supporting evidence suggested that myositis autoantibodies recognising other autoantigens may also disrupt the function of their targets. CONCLUSIONS This study demonstrates that, in myositis, autoantibodies are internalised into living cells, causing biological effects consistent with the disrupted function of their autoantigen.
Collapse
Affiliation(s)
- Iago Pinal-Fernandez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sandra Muñoz-Braceras
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Casal-Dominguez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Katherine Pak
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jiram Torres-Ruiz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jon Musai
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Stefania Dell'Orso
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Faiza Naz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Shamima Islam
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Gustavo Gutierrez-Cruz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maria Dolores Cano
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
| | - Ana Matas-Garcia
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | | | - Ester Tobias-Baraja
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Gloria Garrabou
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Iban Aldecoa
- Pathology, Neurological Tissue Bank, Hospital Clinic of Barcelona-CDB-IDIBAPS/FCRB-University of Barcelona, Barcelona, Spain
| | - Gerard Espinosa
- Barcelona University, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERN-ReCONNET, Hospital Clinic, Barcelona, Spain
| | - Carmen Pilar Simeon-Aznar
- Systemic Autoimmune Disease Section, Vall d'Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Alfredo Guillen-Del-Castillo
- Systemic Autoimmune Disease Section, Vall d'Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Albert Gil-Vila
- Systemic Autoimmune Disease Section, Vall d'Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Ernesto Trallero-Araguás
- Systemic Autoimmune Disease Section, Vall d'Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
- Rheumatology Department, Vall d'Hebron Hospital, Barcelona, Spain
| | - Lisa Christopher-Stine
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas E Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Teerin Liewluck
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elie Naddaf
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Steven A Greenberg
- Department of Neurology, Brigham and Women's Hospital and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Josep Maria Grau
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Albert Selva-O'Callaghan
- Systemic Autoimmune Disease Section, Vall d'Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Jose Cesar Milisenda
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Andrew Lee Mammen
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Hanieh H, Alfwuaires MA, Abduh MS, Abdrabu A, Qinna NA, Alzahrani AM. Protective Effects of a Dihydrodiazepine Against Endotoxin Shock Through Suppression of TLR4/NF-κB/IRF3 Signaling Pathways. Inflammation 2024:10.1007/s10753-024-02160-w. [PMID: 39400777 DOI: 10.1007/s10753-024-02160-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/15/2024]
Abstract
Sepsis and septic shock are life-threatening systemic inflammatory conditions and among the most frequent causes of morbidity and mortality globally. Preclinical evidence has identified a number of diazepine-based compounds with therapeutic potential in inflammatory diseases. However, the potential anti-inflammatory properties of diazepines in the overwhelming immune response during sepsis have been rarely examined. Thus, the current study aimed to identify a new diazepine compound with therapeutic potential in sepsis. Assessing the inflammatory response of macrophages to Lipopolysaccharides (LPS) in vitro identified 2-[7-(trifluoromethyl)-2,3-dihydro-1H-1,4-diazepin-5-yl]phenol (2-TDDP) as a potential anti-inflammatory agent. It reduced secretion of Interleukin-1β (IL-1β), IL-6, IL-12p70, IL-18, Tumor necrosis factor-α (TNF-α), Interferon-γ (IFN-γ), IFN-β, and increased the secretion of IL-10. In a mouse model of LPS-induced endotoxin shock, 2-TDDP reduced mortality and attenuated inflammation-induced tissue injury in the spleen, liver, kidney, and lung. This was accompanied by reduced serum levels of IL-1β, IL-6, IL-12p70, TNF-α, IFN-γ, IFN-β, and increased levels of IL-10. Importantly, 2-TDDP suppressed the Toll-like receptor 4 (TLR4)/Nuclear factor-κB (NF-κB) and TLR4/Interferon regulatory factor 3 (IRF3) signaling pathways through a reduction in the expression of TLR4, Myeloid differentiation primary response 88 (MyD88), P65, and TNF receptor-associated factor 3 (Traf3). Moreover, 2-TDDP suppressed the expression of CD86, Programmed death-ligand 1 (PD-L1) and C5a receptor (C5aR), but not Major histocompatibility complex II (MHCII). Analysis of splenic lymphocyte populations revealed a decrease in the number of CD4+, CD8+, and B cells. Collectively, these findings introduced the dihydrodiazepine 2-TDDP as a new anti-inflammatory agent with potent therapeutic potential in endotoxin shock, paving an avenue for future clinical application.
Collapse
Affiliation(s)
- Hamza Hanieh
- Basic Medical Sciences Department, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba, 77110, Jordan.
| | - Manal A Alfwuaires
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofuf, Saudi Arabia
| | - Maisa S Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, 21589, Jeddah, Saudi Arabia
| | - Alyaa Abdrabu
- Basic Medical Sciences Department, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba, 77110, Jordan
| | - Nidal A Qinna
- University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Abdullah M Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, 31982, Hofuf, Saudi Arabia
| |
Collapse
|
4
|
Sakai M, Masuda Y, Tarumoto Y, Aihara N, Tsunoda Y, Iwata M, Kamiya Y, Komorizono R, Noda T, Yusa K, Tomonaga K, Makino A. Genome-scale CRISPR-Cas9 screen identifies host factors as potential therapeutic targets for SARS-CoV-2 infection. iScience 2024; 27:110475. [PMID: 39100693 PMCID: PMC11295705 DOI: 10.1016/j.isci.2024.110475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 05/01/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Although many host factors important for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have been reported, the mechanisms by which the virus interacts with host cells remain elusive. Here, we identified tripartite motif containing (TRIM) 28, TRIM33, euchromatic histone lysine methyltransferase (EHMT) 1, and EHMT2 as proviral factors involved in SARS-CoV-2 infection by CRISPR-Cas9 screening. Our result suggested that TRIM28 may play a role in viral particle formation and that TRIM33, EHMT1, and EHMT2 may be involved in viral transcription and replication. UNC0642, a compound that specifically inhibits the methyltransferase activity of EHMT1/2, strikingly suppressed SARS-CoV-2 growth in cultured cells and reduced disease severity in a hamster infection model. This study suggests that EHMT1/2 may be a therapeutic target for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Madoka Sakai
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Yoshie Masuda
- Laboratory of Stem Cell Genetics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Yusuke Tarumoto
- Laboratory of Stem Cell Genetics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Naoyuki Aihara
- Laboratory of Veterinary Pathology, Azabu University, Kanagawa 2520206, Japan
| | - Yugo Tsunoda
- Laboratory of Ultrastructural Virology, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto 6068507, Japan
- CREST, Japan Science and Technology Agency, Saitama 1020076, Japan
| | - Michiko Iwata
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Yumiko Kamiya
- Laboratory of Veterinary Pathology, Azabu University, Kanagawa 2520206, Japan
| | - Ryo Komorizono
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
- Laboratory of Ultrastructural Virology, Graduate School of Biostudies, Kyoto University, Kyoto 6068507, Japan
- CREST, Japan Science and Technology Agency, Saitama 1020076, Japan
| | - Kosuke Yusa
- Laboratory of Stem Cell Genetics, Department of Biosystems Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
| | - Keizo Tomonaga
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
- Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto 6068507, Japan
- Department of Molecular Virology, Graduate School of Medicine, Kyoto University, Kyoto 6068507, Japan
| | - Akiko Makino
- Laboratory of RNA Viruses, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto 6068507, Japan
- Laboratory of RNA Viruses, Department of Mammalian Regulatory Network, Graduate School of Biostudies, Kyoto University, Kyoto 6068507, Japan
| |
Collapse
|
5
|
Shen X, Li X, Wu T, Guo T, Lv J, He Z, Luo M, Zhu X, Tian Y, Lai W, Dong C, Hu X, Wu L. TRIM33 plays a critical role in regulating dendritic cell differentiation and homeostasis by modulating Irf8 and Bcl2l11 transcription. Cell Mol Immunol 2024; 21:752-769. [PMID: 38822080 PMCID: PMC11214632 DOI: 10.1038/s41423-024-01179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
The development of distinct dendritic cell (DC) subsets, namely, plasmacytoid DCs (pDCs) and conventional DC subsets (cDC1s and cDC2s), is controlled by specific transcription factors. IRF8 is essential for the fate specification of cDC1s. However, how the expression of Irf8 is regulated is not fully understood. In this study, we identified TRIM33 as a critical regulator of DC differentiation and maintenance. TRIM33 deletion in Trim33fl/fl Cre-ERT2 mice significantly impaired DC differentiation from hematopoietic progenitors at different developmental stages. TRIM33 deficiency downregulated the expression of multiple genes associated with DC differentiation in these progenitors. TRIM33 promoted the transcription of Irf8 to facilitate the differentiation of cDC1s by maintaining adequate CDK9 and Ser2 phosphorylated RNA polymerase II (S2 Pol II) levels at Irf8 gene sites. Moreover, TRIM33 prevented the apoptosis of DCs and progenitors by directly suppressing the PU.1-mediated transcription of Bcl2l11, thereby maintaining DC homeostasis. Taken together, our findings identified TRIM33 as a novel and crucial regulator of DC differentiation and maintenance through the modulation of Irf8 and Bcl2l11 expression. The finding that TRIM33 functions as a critical regulator of both DC differentiation and survival provides potential benefits for devising DC-based immune interventions and therapies.
Collapse
Affiliation(s)
- Xiangyi Shen
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Xiaoguang Li
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Tao Wu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Tingting Guo
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jiaoyan Lv
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zhimin He
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Maocai Luo
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Xinyi Zhu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yujie Tian
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Wenlong Lai
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Chen Dong
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China
- Westlake University School of Medicine, Hangzhou, 310024, China
| | - Xiaoyu Hu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China
| | - Li Wu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China.
| |
Collapse
|
6
|
Tiniakou I, Hsu PF, Lopez-Zepeda LS, Garipler G, Esteva E, Adams NM, Jang G, Soni C, Lau CM, Liu F, Khodadadi-Jamayran A, Rodrick TC, Jones D, Tsirigos A, Ohler U, Bedford MT, Nimer SD, Kaartinen V, Mazzoni EO, Reizis B. Genome-wide screening identifies Trim33 as an essential regulator of dendritic cell differentiation. Sci Immunol 2024; 9:eadi1023. [PMID: 38608038 PMCID: PMC11182672 DOI: 10.1126/sciimmunol.adi1023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 03/21/2024] [Indexed: 04/14/2024]
Abstract
The development of dendritic cells (DCs), including antigen-presenting conventional DCs (cDCs) and cytokine-producing plasmacytoid DCs (pDCs), is controlled by the growth factor Flt3 ligand (Flt3L) and its receptor Flt3. We genetically dissected Flt3L-driven DC differentiation using CRISPR-Cas9-based screening. Genome-wide screening identified multiple regulators of DC differentiation including subunits of TSC and GATOR1 complexes, which restricted progenitor growth but enabled DC differentiation by inhibiting mTOR signaling. An orthogonal screen identified the transcriptional repressor Trim33 (TIF-1γ) as a regulator of DC differentiation. Conditional targeting in vivo revealed an essential role of Trim33 in the development of all DCs, but not of monocytes or granulocytes. In particular, deletion of Trim33 caused rapid loss of DC progenitors, pDCs, and the cross-presenting cDC1 subset. Trim33-deficient Flt3+ progenitors up-regulated pro-inflammatory and macrophage-specific genes but failed to induce the DC differentiation program. Collectively, these data elucidate mechanisms that control Flt3L-driven differentiation of the entire DC lineage and identify Trim33 as its essential regulator.
Collapse
Affiliation(s)
- Ioanna Tiniakou
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Pei-Feng Hsu
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Lorena S. Lopez-Zepeda
- Department of Biology, Humboldt Universität zu Berlin; Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine; Berlin, Germany
| | - Görkem Garipler
- Department of Biology, New York University; New York, NY, USA
| | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Nicholas M. Adams
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Geunhyo Jang
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Chetna Soni
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| | - Colleen M. Lau
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine; Ithaca, NY, USA
| | - Fan Liu
- Department of Biochemistry and Molecular Biology, Department of Medicine and Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine; Miami, FL, USA
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine; New York, NY, USA
| | - Tori C. Rodrick
- Metabolomics Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine; New York, NY, USA
| | - Drew Jones
- Metabolomics Laboratory, Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine; New York, NY, USA
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine; New York, NY, USA
| | - Uwe Ohler
- Department of Biology, Humboldt Universität zu Berlin; Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine; Berlin, Germany
| | - Mark T. Bedford
- Department of Epigenetics & Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center; Houston, TX, USA
| | - Stephen D. Nimer
- Department of Biochemistry and Molecular Biology, Department of Medicine and Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine; Miami, FL, USA
| | - Vesa Kaartinen
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry; Ann Arbor, MI, USA
| | | | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine; New York, NY, USA
| |
Collapse
|
7
|
Pinal-Fernandez I, Muñoz-Braceras S, Casal-Dominguez M, Pak K, Torres-Ruiz J, Musai J, Dell’Orso S, Naz F, Islam S, Gutierrez-Cruz G, Cano MD, Matas-Garcia A, Padrosa J, Tobías-Baraja E, Garrabou G, Aldecoa I, Espinosa G, Simeon-Aznar CP, Guillen-Del-Castillo A, Gil-Vila A, Trallero-Araguas E, Christopher-Stine L, Lloyd TE, Liewluck T, Naddaf E, Stenzel W, Greenberg SA, Grau JM, Selva-O’Callaghan A, Milisenda JC, Mammen AL. Pathogenic autoantibody internalization in myositis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.01.15.24301339. [PMID: 38313303 PMCID: PMC10836124 DOI: 10.1101/2024.01.15.24301339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Objectives Myositis is a heterogeneous family of autoimmune muscle diseases. As myositis autoantibodies recognize intracellular proteins, their role in disease pathogenesis has been unclear. This study aimed to determine whether myositis autoantibodies reach their autoantigen targets within muscle cells and disrupt the normal function of these proteins. Methods Confocal immunofluorescence microscopy was used to localize antibodies and other proteins of interest in myositis muscle biopsies. Bulk RNA sequencing was used to study the transcriptomic profiles of 668 samples from patients with myositis, disease controls, and healthy controls. Antibodies from myositis patients were introduced into cultured myoblasts by electroporation and the transcriptomic profiles of the treated myoblasts were studied by bulk RNA sequencing. Results In patients with myositis autoantibodies, antibodies accumulated inside myofibers in the same subcellular compartment as the autoantigen. Each autoantibody was associated with effects consistent with dysfunction of its autoantigen, such as the derepression of genes normally repressed by Mi2/NuRD in patients with anti-Mi2 autoantibodies, the accumulation of RNAs degraded by the nuclear RNA exosome complex in patients with anti-PM/Scl autoantibodies targeting this complex, and the accumulation of lipids within myofibers of anti-HMGCR-positive patients. Internalization of patient immunoglobulin into cultured myoblasts recapitulated the transcriptomic phenotypes observed in human disease, including the derepression of Mi2/NuRD-regulated genes in anti-Mi2-positive dermatomyositis and the increased expression of genes normally degraded by the nuclear RNA exosome complex in anti-PM/Scl-positive myositis. Conclusions In myositis, autoantibodies are internalized into muscle fibers, disrupt the biological function of their autoantigen, and mediate the pathophysiology of the disease.
Collapse
Affiliation(s)
- Iago Pinal-Fernandez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sandra Muñoz-Braceras
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Casal-Dominguez
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Katherine Pak
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jiram Torres-Ruiz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jon Musai
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stefania Dell’Orso
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Faiza Naz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shamima Islam
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Gustavo Gutierrez-Cruz
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Maria Dolores Cano
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
| | - Ana Matas-Garcia
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | | | - Esther Tobías-Baraja
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Gloria Garrabou
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Iban Aldecoa
- Pathology, Neurological Tissue Bank. Hospital Clinic of Barcelona-CDB-IDIBAPS/FCRB-University of Barcelona, Barcelona, Spain
| | - Gerard Espinosa
- Barcelona University, Barcelona, Spain
- Department of Autoimmune Diseases, Reference Centre for Systemic Autoimmune Diseases (UEC/CSUR) of the Catalan and Spanish Health Systems-Member of ERN-ReCONNET, Hospital Clinic, Barcelona, Spain
| | - Carmen Pilar Simeon-Aznar
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Alfredo Guillen-Del-Castillo
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Albert Gil-Vila
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Ernesto Trallero-Araguas
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Lisa Christopher-Stine
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas E. Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teerin Liewluck
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Elie Naddaf
- Division of Neuromuscular Medicine, Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Steven A. Greenberg
- Department of Neurology, Brigham and Women’s Hospital and Boston Children’s Hospital, Harvard Medical School, MA, USA
| | - Josep Maria Grau
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Albert Selva-O’Callaghan
- Systemic Autoimmune Disease Section, Vall d’Hebron Institute of Research, Barcelona, Spain
- Autonomous University of Barcelona, Barcelona, Spain
| | - Jose C. Milisenda
- Muscle Research Unit, Internal Medicine Service, Hospital Clinic, Barcelona, Spain
- Barcelona University, Barcelona, Spain
- CIBERER and IDIBAPS, Barcelona, Spain
| | - Andrew L. Mammen
- Muscle Disease Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
8
|
Hui Z, Fu Y, Chen Y, Yin J, Fang H, Tu Y, Gu Y, Zhang J. Loss of TRIM24 promotes IL-10 expression via CBP/p300-dependent IFNβ1 transcription during macrophage activation. Inflamm Res 2023:10.1007/s00011-023-01751-x. [PMID: 37326695 DOI: 10.1007/s00011-023-01751-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/25/2023] [Accepted: 05/03/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND As an anti-inflammatory cytokine, interleukin 10 (IL-10) plays a vital role in preventing inflammatory and autoimmune pathologies while also maintaining immune homeostasis. IL-10 production in macrophages is tightly regulated by multiple pathways. TRIM24, a member of the Transcriptional Intermediary Factor 1 (TIF1) family, contributes to antiviral immunity and macrophage M2 polarization. However, the role of TRIM24 in regulating IL-10 expression and its involvement in endotoxic shock remains unclear. METHODS In vitro, bone marrow derived macrophages cultured with GM-CSF or M-CSF were stimulated with LPS (100ng/ml). Murine models of endotoxic shock were established by challenging the mice with different dose of LPS (i.p). RTPCR, RNA sequencing, ELISA and hematoxylin and eosin staining were performed to elucidate the role and mechanisms of TRIM24 in endotoxic shock. RESULTS The expression of TRIM24 is downregulated in LPS-stimulated bone marrow-derived macrophages (BMDMs). Loss of TRIM24 boosted IL-10 expression during the late stage of LPS-stimulation in macrophages. RNA-seq analysis revealed the upregulation of IFNβ1, an upstream regulator of IL-10, in TRIM24 knockout macrophages. Treatment with C646, a CBP/p300 inhibitor, diminished the difference in both IFNβ1 and IL-10 expression between TRIM24 knockout and control macrophages. Loss of TRIM24 provided protection against LPS-induced endotoxic shock in mice. CONCLUSION Our results demonstrated that inhibiting TRIM24 promoted the expression of IFNβ1 and IL-10 during macrophage activation, therefore protecting mice from endotoxic shock. This study offers novel insights into the regulatory role of TRIM24 in IL-10 expression, making it a potentially attractive therapeutic target for inflammatory diseases.
Collapse
Affiliation(s)
- Zhaoyuan Hui
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Pathogenic Biology and Medical Immunology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, Yinchuan, 750004, China
| | - Yuanzheng Fu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yunyun Chen
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jie Yin
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Hui Fang
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Yifan Tu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ying Gu
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
- Department of Genetics, Institute of Genetics, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab of Genetic and Developmental Disorder, Hangzhou, 310058, Zhejiang, China.
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China.
| | - Jiawei Zhang
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
9
|
Fan J, Li Q, Liang J, Chen Z, Chen L, Lai J, Chen Q. Regulation of IFNβ expression: focusing on the role of its promoter and transcription regulators. Front Microbiol 2023; 14:1158777. [PMID: 37396372 PMCID: PMC10309559 DOI: 10.3389/fmicb.2023.1158777] [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: 02/07/2023] [Accepted: 05/23/2023] [Indexed: 07/04/2023] Open
Abstract
IFNβ is a single-copy gene without an intron. Under normal circumstances, it shows low or no expression in cells. It is upregulated only when the body needs it or is stimulated. Stimuli bind to the pattern recognition receptors (PRRs) and pass via various signaling pathways to several basic transcriptional regulators, such as IRFs, NF-кB, and AP-1. Subsequently, the transcriptional regulators enter the nucleus and bind to regulatory elements of the IFNβ promoter. After various modifications, the position of the nucleosome is altered and the complex is assembled to activate the IFNβ expression. However, IFNβ regulation involves a complex network. For the study of immunity and diseases, it is important to understand how transcription factors bind to regulatory elements through specific forms, which elements in cells are involved in regulation, what regulation occurs during the assembly of enhancers and transcription complexes, and the possible regulatory mechanisms after transcription. Thus, this review focuses on the various regulatory mechanisms and elements involved in the activation of IFNβ expression. In addition, we discuss the impact of this regulation in biology.
Collapse
Affiliation(s)
- Jiqiang Fan
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Qiumei Li
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Jiadi Liang
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Zhirong Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Linqin Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| | - Junzhong Lai
- The Cancer Center, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qi Chen
- Fujian Key Laboratory of Innate Immune Biology, Biomedical Research Center of South China, Fujian Normal University, Fuzhou, China
| |
Collapse
|
10
|
Li M, Zhang Y, Zhang W, Sun J, Liu R, Pan Z, Zhang P, Liu S. Type 1 interferon signature in peripheral blood mononuclear cells and monocytes of idiopathic inflammatory myopathy patients with different myositis-specific autoantibodies. Front Immunol 2023; 14:1169057. [PMID: 37228600 PMCID: PMC10203462 DOI: 10.3389/fimmu.2023.1169057] [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: 02/18/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Background Myositis-specific autoantibodies (MSAs) are clinically used to diagnose and define idiopathic inflammatory myopathy (IIM) subsets. However, the underlying pathogenic mechanisms of patients with different MSAs remain unclear. Methods A total of 158 Chinese patients with IIM and 167 gender- and age-matched healthy controls (HCs) were enrolled. Transcriptome sequencing (RNA-Seq) was performed with peripheral blood mononuclear cells (PBMCs), followed by the identification of differentially expressed genes (DEGs) and analysis of gene set enrichment analysis, immune cell infiltration, and WGCNA. Monocyte subsets and related cytokines/chemokines were quantified. The expressions of interferon (IFN)-related genes were validated using qRT-PCR and Western blot in both PBMCs and monocytes. We also performed correlation analysis and ROC analysis to explore the potential clinical significance of the IFN-related genes. Results There were 1,364 genes altered in patients with IIM, including 952 upregulated and 412 downregulated genes. The type I interferon (IFN-I) pathway was remarkably activated in patients with IIM. Compared with patients with other MSAs, IFN-I signatures were significantly activated in patients with anti-melanoma differentiation-associated gene 5 (MDA5) antibodies. In total, 1,288 hub genes associated with IIM onset were identified using WGCNA, including 29 key DEGs associated with IFN signaling. The patients had more CD14brightCD16- classical, CD14brightCD16+ intermediate, and fewer CD14dimCD16+ non-classical monocyte subsets. Plasma cytokines like IL-6 and TNF and chemokines including CCL3 and MCPs increased. The validation of IFN-I-related gene expressions was consistent with the findings from RNA-Seq. The IFN-related genes were correlated with laboratory parameters and helpful for IIM diagnosis. Conclusion Gene expressions were remarkably altered in the PBMCs of IIM patients. Anti-MDA5+ IIM patients had a more pronounced activated IFN signature than others. Monocytes exhibited a proinflammatory feature and contributed to the IFN signature of IIM patients.
Collapse
Affiliation(s)
- Mengdi Li
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yusheng Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhe Zhang
- Department of Radiology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinlei Sun
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhou Pan
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Panpan Zhang
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shengyun Liu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Du L, Liu W, Rosen ST, Chen Y. Mechanism of SUMOylation-Mediated Regulation of Type I IFN Expression. J Mol Biol 2023; 435:167968. [PMID: 36681180 DOI: 10.1016/j.jmb.2023.167968] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 01/07/2023] [Accepted: 01/11/2023] [Indexed: 01/20/2023]
Abstract
Type I interferons (IFN) are cytokines that bridge the innate and adaptive immune response, and thus play central roles in human health, including vaccine efficacy, immune response to cancer and pathogen infection, and autoimmune disorders. Post-translational protein modifications by the small ubiquitin-like modifiers (SUMO) have recently emerged as an important regulator of type I IFN expression as shown by studies using murine and cellular models and recent human clinical trials. However, the mechanism regarding how SUMOylation regulates type I IFN expression remains poorly understood. In this study, we show that SUMOylation inhibition does not activate IFNB1 gene promoter that is regulated by known canonical pathways including cytosolic DNA. Instead, we identified a binding site for the chromatin modification enzyme, the SET Domain Bifurcated Histone Lysine Methyltransferase 1 (SETDB1), located between the IFNB1 promoter and a previously identified enhancer. We found that SETDB1 regulates IFNB1 expression and SUMOylation of SETDB1 is required for its binding and enhancing the H3K9me3 heterochromatin signal in this region. Heterochromatin, a tightly packed form of DNA, has been documented to suppress gene expression through suppressing enhancer function. Taken together, our study identified a novel mechanism of regulation of type I IFN expression, at least in part, through SUMOylation of a chromatin modification enzyme.
Collapse
Affiliation(s)
- Li Du
- Toni Stephenson Lymphoma Center, Beckman Research Institute of City of Hope, Duarte, CA, USA; Judy and Bernard Briskin Center for Multiple Myeloma Research, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Wei Liu
- Toni Stephenson Lymphoma Center, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Steven T Rosen
- Toni Stephenson Lymphoma Center, Beckman Research Institute of City of Hope, Duarte, CA, USA; Judy and Bernard Briskin Center for Multiple Myeloma Research, Beckman Research Institute of City of Hope, Duarte, CA, USA; Department of Hematology and Hematopoietic Cell Transplantation, Beckman Research Institute of City of Hope, Duarte, CA, USA; City of Hope Comprehensive Cancer Center, City of Hope National Medical Center, Duarte, CA, USA.
| | - Yuan Chen
- Division of Surgical Sciences, Department of Surgery and Moores Cancer Center, UC San Diego Health, San Diego, CA, USA.
| |
Collapse
|
12
|
Lou Demy D, Touret AL, Lancino M, Tauzin M, Capuana L, Pierre C, Herbomel P. Trim33 conditions the lifespan of primitive macrophages and onset of definitive macrophage production. Development 2022; 149:276505. [DOI: 10.1242/dev.200835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022]
Abstract
ABSTRACT
Trim33 (Tif1γ) is a transcriptional regulator that is notably involved in several aspects of hematopoiesis. It is essential for the production of erythrocytes in zebrafish, and for the proper functioning and aging of hematopoietic stem and progenitor cells (HSPCs) in mice. Here, we have found that, in zebrafish development, Trim33 is essential cell-autonomously for the lifespan of the yolk sac-derived primitive macrophages, as well as for the initial production of definitive (HSPC-derived) macrophages in the first niche of definitive hematopoiesis, the caudal hematopoietic tissue. Moreover, Trim33 deficiency leads to an excess production of definitive neutrophils and thrombocytes. Our data indicate that Trim33 radically conditions the differentiation output of aorta-derived HSPCs in all four erythro-myeloid cell types, in a niche-specific manner.
Collapse
Affiliation(s)
- Doris Lou Demy
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Anne-Lou Touret
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Mylène Lancino
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Muriel Tauzin
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Lavinia Capuana
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Constance Pierre
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| | - Philippe Herbomel
- Institut Pasteur 1 , Department of Developmental and Stem Cell Biology, 75015 Paris , France
- CNRS, UMR3738 2 , 75015 Paris , France
| |
Collapse
|
13
|
Chen M, Lingadahalli S, Narwade N, Lei KMK, Liu S, Zhao Z, Zheng Y, Lu Q, Tang AHN, Poon TCW, Cheung E. TRIM33 drives prostate tumor growth by stabilizing androgen receptor from Skp2-mediated degradation. EMBO Rep 2022; 23:e53468. [PMID: 35785414 DOI: 10.15252/embr.202153468] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 05/13/2022] [Accepted: 06/07/2022] [Indexed: 12/23/2022] Open
Abstract
Androgen receptor (AR) is a master transcription factor that drives prostate cancer (PCa) development and progression. Alterations in the expression or activity of AR coregulators significantly impact the outcome of the disease. Using a proteomics approach, we identified the tripartite motif-containing 33 (TRIM33) as a novel transcriptional coactivator of AR. We demonstrate that TRIM33 facilitates AR chromatin binding to directly regulate a transcription program that promotes PCa progression. TRIM33 further stabilizes AR by protecting it from Skp2-mediated ubiquitination and proteasomal degradation. We also show that TRIM33 is essential for PCa tumor growth by avoiding cell-cycle arrest and apoptosis, and TRIM33 knockdown sensitizes PCa cells to AR antagonists. In clinical analyses, we find TRIM33 upregulated in multiple PCa patient cohorts. Finally, we uncover an AR-TRIM33-coactivated gene signature highly expressed in PCa tumors and predict disease recurrence. Overall, our results reveal that TRIM33 is an oncogenic AR coactivator in PCa and a potential therapeutic target for PCa treatment.
Collapse
Affiliation(s)
- Mi Chen
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Shreyas Lingadahalli
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Nitin Narwade
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Kate Man Kei Lei
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Pilot Laboratory, University of Macau, Taipa, Macau SAR.,Institute of Translational Medicine, University of Macau, Taipa, Macau SAR
| | | | - Zuxianglan Zhao
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Yimin Zheng
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| | - Qian Lu
- Xuzhou Medical University, Xuzhou, China
| | | | - Terence Chuen Wai Poon
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR.,Pilot Laboratory, University of Macau, Taipa, Macau SAR.,Institute of Translational Medicine, University of Macau, Taipa, Macau SAR
| | - Edwin Cheung
- Cancer Centre, University of Macau, Taipa, Macau SAR.,Centre for Precision Medicine Research and Training, University of Macau, Taipa, Macau SAR.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR.,Faculty of Health Sciences, University of Macau, Taipa, Macau SAR
| |
Collapse
|
14
|
Turnier JL, Kahlenberg JM. Using autoantibody signatures to define cancer risk in dermatomyositis. J Clin Invest 2022; 132:e156025. [PMID: 35040442 PMCID: PMC8759773 DOI: 10.1172/jci156025] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dermatomyositis is an idiopathic inflammatory myopathy with a highly heterogeneous disease course. Although there is a known increase in cancer risk surrounding the time of dermatomyositis diagnosis, the mechanisms driving this increased risk are not well understood. Further, there are no current standardized cancer screening guidelines for dermatomyositis patients. In this issue of the JCI, Fiorentino, Mecoli, et al. discovered additional autoantibodies in patients with dermatomyositis and anti-TIF1-γ autoantibodies, a known risk factor for malignancy. They observed a decreased cancer risk with an increasing number of autoantibodies. Importantly, these findings indicate that more detailed autoantibody phenotyping at diagnosis might better predict cancer risk and also suggest that diversity and kinetics of the host immune response might influence cancer development.
Collapse
Affiliation(s)
| | - J. Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
15
|
Thomsen EA, Andersen S, Marqvorsen MHS, Skipper KA, Paludan SR, Mikkelsen JG. Single-Cell Monitoring of Activated Innate Immune Signaling by a d2eGFP-Based Reporter Mimicking Time-Restricted Activation of IFNB1 Expression. Front Cell Infect Microbiol 2022; 11:784762. [PMID: 35118008 PMCID: PMC8803904 DOI: 10.3389/fcimb.2021.784762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/21/2021] [Indexed: 01/19/2023] Open
Abstract
The innate immune system represents a balanced first line of defense against infection. Type I interferons (IFNs) are key regulators of the response to viral infections with an essential early wave of IFN-β expression, which is conditional, time-restricted, and stochastic in its nature. The possibility to precisely monitor individual cells with active IFNB1 transcription during innate signaling requires a robust reporter system that mimics the endogenous IFN-β signal. Here, we present a reporter system based on expression of a destabilized version of eGFP (d2eGFP) from a stably integrated reporter cassette containing the IFNB1 promoter and 3'-untranslated region, enabling both spatial and temporal detection of regulated IFNB1 expression. Specifically, this reporter permits detection, quantification, and isolation of cells actively producing d2eGFP in a manner that fully mimics IFN-β production allowing tracking of IFNB1 gene activation and repression in monocytic cells and keratinocytes. Using induced d2eGFP expression as a readout for activated immune signaling at the single-cell level, we demonstrate the application of the reporter for FACS-based selection of cells with genotypes supporting cGAS-STING signaling. Our studies provide a novel approach for monitoring on/off-switching of innate immune signaling and form the basis for investigating genotypes affecting immune regulation at the single-cell level.
Collapse
|
16
|
Wei W, Li G, Liu Z, Yang H, Liu S, Zou X, Jiao Y. Feibi decoction-medicated serum inhibits lipopolysaccharide-induced inflammation in RAW264.7 cells and BMDMs. Exp Ther Med 2022; 23:110. [PMID: 34976152 DOI: 10.3892/etm.2021.11033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 07/06/2021] [Indexed: 02/05/2023] Open
Abstract
Feibi decoction (FBD) is a traditional Chinese herbal medicine and has been clinically used in the treatment of pulmonary fibrosis (PF), which is characterized by diffuse interstitial inflammation and exaggerated collagen accumulation. However, the potential mechanisms remain to be elucidated. The present study aimed to investigate the effect of FBD-medicated serum (FBDS) on lipopolysaccharide (LPS)-induced inflammation in macrophages. In RAW264.7 macrophages and bone marrow-derived macrophages (BMDMs), FBDS treatment significantly inhibited the production of pro-inflammatory cytokines induced by LPS. In addition, it was indicated that FBDS treatment suppressed the activation of NF-κB and Smad2/Smad3 following LPS treatment. Furthermore, FBDS treatment decreased the expression of transforming growth factor-β1 and chitinase-3-like protein 1. In conclusion, the results demonstrated that treatment with FBDS inhibited LPS-induced inflammation in RAW264.7 and BMDM cells. These data may improve understanding of the effect of FBD on anti-inflammation and help determine the mechanisms underlying the alleviation of PF via FBD.
Collapse
Affiliation(s)
- Wan Wei
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China.,Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| | - Guodong Li
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Zhaoheng Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Haojie Yang
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Shuo Liu
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Xinxin Zou
- Graduate School, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
| | - Yang Jiao
- Dongfang Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100078, P.R. China
| |
Collapse
|
17
|
Gaidt MM, Morrow A, Fairgrieve MR, Karr JP, Yosef N, Vance RE. Self-guarding of MORC3 enables virulence factor-triggered immunity. Nature 2021; 600:138-142. [PMID: 34759314 PMCID: PMC9045311 DOI: 10.1038/s41586-021-04054-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 09/23/2021] [Indexed: 01/08/2023]
Abstract
Pathogens use virulence factors to inhibit the immune system1. The guard hypothesis2,3 postulates that hosts monitor (or 'guard') critical innate immune pathways such that their disruption by virulence factors provokes a secondary immune response1. Here we describe a 'self-guarded' immune pathway in human monocytes, in which guarding and guarded functions are combined in one protein. We find that this pathway is triggered by ICP0, a key virulence factor of herpes simplex virus type 1, resulting in robust induction of anti-viral type I interferon (IFN). Notably, induction of IFN by ICP0 is independent of canonical immune pathways and the IRF3 and IRF7 transcription factors. A CRISPR screen identified the ICP0 target MORC34 as an essential negative regulator of IFN. Loss of MORC3 recapitulates the IRF3- and IRF7-independent IFN response induced by ICP0. Mechanistically, ICP0 degrades MORC3, which leads to de-repression of a MORC3-regulated DNA element (MRE) adjacent to the IFNB1 locus. The MRE is required in cis for IFNB1 induction by the MORC3 pathway, but is not required for canonical IFN-inducing pathways. As well as repressing the MRE to regulate IFNB1, MORC3 is also a direct restriction factor of HSV-15. Our results thus suggest a model in which the primary anti-viral function of MORC3 is self-guarded by its secondary IFN-repressing function-thus, a virus that degrades MORC3 to avoid its primary anti-viral function will unleash the secondary anti-viral IFN response.
Collapse
Affiliation(s)
- Moritz M Gaidt
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| | - Alyssa Morrow
- Electrical Engineering and Computer Science Department, University of California, Berkeley, CA, USA
| | - Marian R Fairgrieve
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Jonathan P Karr
- Division of Genetics, Genomics and Development, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Nir Yosef
- Electrical Engineering and Computer Science Department, University of California, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, CA, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Boston, MA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Russell E Vance
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Cancer Research Laboratory and the Immunotherapeutics and Vaccine Research Initiative, University of California, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
18
|
Li Q, Qin Y, Wang W, Jia M, Zhao W, Zhao C. KAP1-Mediated Epigenetic Suppression in Anti-RNA Viral Responses by Direct Targeting RIG-I and MDA5. THE JOURNAL OF IMMUNOLOGY 2021; 207:1903-1910. [PMID: 34497149 DOI: 10.4049/jimmunol.2100342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/26/2021] [Indexed: 11/19/2022]
Abstract
Retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), including RIG-I (encoded by Ddx58) and melanoma differentiation-associated gene 5 (MDA5) (encoded by Ifih1), are crucial for initiating antiviral responses. Endogenous retroviral elements (ERVs) are transposable elements derived from exogenous retroviruses that are integrated into the genome. KRAB-associated protein 1 (KAP1) is a key epigenetic suppressor of ERVs that protects cells from detrimental genome instability. Increased ERV transcripts are sensed by RLRs and trigger innate immune signaling. However, whether KAP1 directly controls RLRs activity remains unclear. In this study, we show that KAP1 attenuates RNA viral infection-induced type I IFNs and facilitates viral replication by inhibiting RIG-I/MDA5 expression in primary peritoneal macrophages (PMs) of C57BL/6J mice. Kap1 deficiency increases IFN-β expression and inhibits vesicular stomatitis virus replication in C57BL/6J mice in vivo. Mechanistically, KAP1 binds to the promoter regions of Ddx58 and Ifih1 and promotes the establishment of repressive histone marks in primary PMs of C57BL/6J mice. Concordantly, KAP1 suppresses the expression of RIG-I and MDA5 at the transcriptional level in primary PMs of C57BL/6J mice. Our results establish that KAP1 epigenetically suppresses host antiviral responses by directly targeting RIG-1 and MDA5, thus facilitating the immune escape of RNA viruses.
Collapse
Affiliation(s)
- Qi Li
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; and
| | - Ying Qin
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; and
| | - Wenwen Wang
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; and
| | - Mutian Jia
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; and
| | - Wei Zhao
- Department of Pathogenic Biology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; and
| | - Chunyuan Zhao
- Department of Cell Biology, School of Basic Medical Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
19
|
Dubey AR, Jagtap YA, Kumar P, Patwa SM, Kinger S, Kumar A, Singh S, Prasad A, Jana NR, Mishra A. Biochemical strategies of E3 ubiquitin ligases target viruses in critical diseases. J Cell Biochem 2021; 123:161-182. [PMID: 34520596 DOI: 10.1002/jcb.30143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/23/2021] [Accepted: 08/24/2021] [Indexed: 12/15/2022]
Abstract
Viruses are known to cause various diseases in human and also infect other species such as animal plants, fungi, and bacteria. Replication of viruses depends upon their interaction with hosts. Human cells are prone to such unwanted viral infections. Disintegration and reconstitution require host machinery and various macromolecules like DNA, RNA, and proteins are invaded by viral particles. E3 ubiquitin ligases are known for their specific function, that is, recognition of their respective substrates for intracellular degradation. Still, we do not understand how ubiquitin proteasome system-based enzymes E3 ubiquitin ligases do their functional interaction with different viruses. Whether E3 ubiquitin ligases help in the elimination of viral components or viruses utilize their molecular capabilities in their intracellular propagation is not clear. The first time our current article comprehends fundamental concepts and new insights on the different viruses and their interaction with various E3 Ubiquitin Ligases. In this review, we highlight the molecular pathomechanism of viruses linked with E3 Ubiquitin Ligases dependent mechanisms. An enhanced understanding of E3 Ubiquitin Ligase-mediated removal of viral proteins may open new therapeutic strategies against viral infections.
Collapse
Affiliation(s)
- Ankur R Dubey
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Yuvraj A Jagtap
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Prashant Kumar
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Som M Patwa
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sumit Kinger
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Amit Kumar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Sarika Singh
- Department of Neuroscience and Ageing Biology, Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India
| | - Amit Prasad
- School of Basic Sciences, Indian Institute of Technology Mandi, Mandi, India
| | - Nihar R Jana
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Amit Mishra
- Department of Bioscience and Bioengineering, Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
20
|
Pavlova AV, Kubareva EA, Monakhova MV, Zvereva MI, Dolinnaya NG. Impact of G-Quadruplexes on the Regulation of Genome Integrity, DNA Damage and Repair. Biomolecules 2021; 11:1284. [PMID: 34572497 PMCID: PMC8472537 DOI: 10.3390/biom11091284] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/19/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022] Open
Abstract
DNA G-quadruplexes (G4s) are known to be an integral part of the complex regulatory systems in both normal and pathological cells. At the same time, the ability of G4s to impede DNA replication plays a critical role in genome integrity. This review summarizes the results of recent studies of G4-mediated genomic and epigenomic instability, together with associated DNA damage and repair processes. Although the underlying mechanisms remain to be elucidated, it is known that, among the proteins that recognize G4 structures, many are linked to DNA repair. We analyzed the possible role of G4s in promoting double-strand DNA breaks, one of the most deleterious DNA lesions, and their repair via error-prone mechanisms. The patterns of G4 damage, with a focus on the introduction of oxidative guanine lesions, as well as their removal from G4 structures by canonical repair pathways, were also discussed together with the effects of G4s on the repair machinery. According to recent findings, there must be a delicate balance between G4-induced genome instability and G4-promoted repair processes. A broad overview of the factors that modulate the stability of G4 structures in vitro and in vivo is also provided here.
Collapse
Affiliation(s)
- Anzhela V. Pavlova
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia; (M.I.Z.); (N.G.D.)
| | - Elena A. Kubareva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia; (E.A.K.); (M.V.M.)
| | - Mayya V. Monakhova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia; (E.A.K.); (M.V.M.)
| | - Maria I. Zvereva
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia; (M.I.Z.); (N.G.D.)
| | - Nina G. Dolinnaya
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1, 119991 Moscow, Russia; (M.I.Z.); (N.G.D.)
| |
Collapse
|
21
|
Davis EE, Balasubramanian R, Kupchinsky ZA, Keefe DL, Plummer L, Khan K, Meczekalski B, Heath KE, Lopez-Gonzalez V, Ballesta-Martinez MJ, Margabanthu G, Price S, Greening J, Brauner R, Valenzuela I, Cusco I, Fernandez-Alvarez P, Wierman ME, Li T, Lage K, Barroso PS, Chan YM, Crowley WF, Katsanis N. TCF12 haploinsufficiency causes autosomal dominant Kallmann syndrome and reveals network-level interactions between causal loci. Hum Mol Genet 2021; 29:2435-2450. [PMID: 32620954 DOI: 10.1093/hmg/ddaa120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/27/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
Dysfunction of the gonadotropin-releasing hormone (GnRH) axis causes a range of reproductive phenotypes resulting from defects in the specification, migration and/or function of GnRH neurons. To identify additional molecular components of this system, we initiated a systematic genetic interrogation of families with isolated GnRH deficiency (IGD). Here, we report 13 families (12 autosomal dominant and one autosomal recessive) with an anosmic form of IGD (Kallmann syndrome) with loss-of-function mutations in TCF12, a locus also known to cause syndromic and non-syndromic craniosynostosis. We show that loss of tcf12 in zebrafish larvae perturbs GnRH neuronal patterning with concomitant attenuation of the orthologous expression of tcf3a/b, encoding a binding partner of TCF12, and stub1, a gene that is both mutated in other syndromic forms of IGD and maps to a TCF12 affinity network. Finally, we report that restored STUB1 mRNA rescues loss of tcf12 in vivo. Our data extend the mutational landscape of IGD, highlight the genetic links between craniofacial patterning and GnRH dysfunction and begin to assemble the functional network that regulates the development of the GnRH axis.
Collapse
Affiliation(s)
- Erica E Davis
- Center for Human Disease Modeling, Duke University, Durham, NC 27701, USA.,Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ravikumar Balasubramanian
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA.,Harvard Medical School, Boston, MA 02115, USA
| | | | - David L Keefe
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA
| | - Lacey Plummer
- Harvard Reproductive Endocrine Science Center, Massachusetts General Hospital (MGH), Boston, MA 02114, USA
| | - Kamal Khan
- Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Blazej Meczekalski
- Department of Gynecological Endocrinology, Poznan University of Medical Sciences, 60-512 Poznan, Poland
| | - Karen E Heath
- Institute of Medical and Molecular Genetics (INGEMM) Hospital Universitario La Paz, Universidad Autonoma de Madrid, IdiPAZ, Madrid, Spain and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28046 Madrid, Spain
| | - Vanesa Lopez-Gonzalez
- Medical Genetics Unit, Department of Pediatrics, Hospital Clinico, Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain and CIBERER, ISCIII, 28046 Madrid, Spain
| | - Mary J Ballesta-Martinez
- Medical Genetics Unit, Department of Pediatrics, Hospital Clinico, Universitario Virgen de la Arrixaca, IMIB-Arrixaca, Murcia, Spain and CIBERER, ISCIII, 28046 Madrid, Spain
| | | | - Susan Price
- Northampton General Hospital, Northampton NN1 5BD, UK
| | - James Greening
- University Hospitals of Leicester, Leicester LE3 9QP, UK
| | - Raja Brauner
- Pediatric Endocrinology Unit, Fondation Ophtalmologique Adolphe de Rothschild and Université Paris Descartes, 75019 Paris, France
| | - Irene Valenzuela
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ivon Cusco
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Paula Fernandez-Alvarez
- Department of Clinical and Molecular Genetics, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain.,Medicine Genetics Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Margaret E Wierman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taibo Li
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.,Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Kasper Lage
- Harvard Medical School, Boston, MA 02115, USA.,Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Priscila Sales Barroso
- Divisao de Endocrinologia e Metabologia, Hospital das Clinicas da Faculdade de Medicina da Universidade de Sao Paulo, Sao Paulo, 05403-900 Brazil
| | - Yee-Ming Chan
- Division of Endocrinology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - William F Crowley
- Harvard Medical School, Boston, MA 02115, USA.,MGH Center for Human Genetics & The Endocrine Unit, Department of Medicine, Massachusetts General Hospital, Boston MA 02114, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University, Durham, NC 27701, USA.,Advanced Center for Translational and Genetic Medicine (ACT-GeM), Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.,Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
22
|
Valentino SA, Chézeau L, Seidel C, Sébillaud S, Lorcin M, Chalansonnet M, Cosnier F, Gaté L. Exposure to TiO 2 Nanostructured Aerosol Induces Specific Gene Expression Profile Modifications in the Lungs of Young and Elderly Rats. NANOMATERIALS 2021; 11:nano11061466. [PMID: 34206090 PMCID: PMC8230065 DOI: 10.3390/nano11061466] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022]
Abstract
Although aging is associated with a higher risk of developing respiratory pathologies, very few studies have assessed the impact of age on the adverse effects of inhaled nanoparticles. Using conventional and transcriptomic approaches, this study aimed to compare in young (12–13-week-old) and elderly (19-month-old) fisher F344 rats the pulmonary toxicity of an inhaled nanostructured aerosol of titanium dioxide (TiO2). Animals were nose-only exposed to this aerosol at a concentration of 10 mg/m3 for 6 h per day, 5 days per week for 4 weeks. Tissues were collected immediately (D0), and 28 days after exposure (D28). A pulmonary influx of neutrophilic granulocytes was observed in exposed rats at D0, but diminished with time while remaining significant until D28. Similarly, an increased expression of several genes involved in inflammation at the two post-exposure time-points was seen. Apart from an age-specific pulmonary influx of lymphocyte, only slight differences in physio-pathological responses following TiO2 exposure between young and elderly animals were noticed. Conversely, marked age-related differences in gene expression profiles were observed making possible to establish lists of genes specific to each age group and post-exposure times. These results highlight different signaling pathways that were disrupted in rats according to their age.
Collapse
|
23
|
An unconventional role of an ASB family protein in NF-κB activation and inflammatory response during microbial infection and colitis. Proc Natl Acad Sci U S A 2021; 118:2015416118. [PMID: 33431678 DOI: 10.1073/pnas.2015416118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor κB (NF-κB)-mediated signaling pathway plays a crucial role in the regulation of inflammatory process, innate and adaptive immune responses. The hyperactivation of inflammatory response causes host cell death, tissue damage, and autoinflammatory disorders, such as sepsis and inflammatory bowel disease. However, how these processes are precisely controlled is still poorly understood. In this study, we demonstrated that ankyrin repeat and suppressor of cytokine signaling box containing 1 (ASB1) is involved in the positive regulation of inflammatory responses by enhancing the stability of TAB2 and its downstream signaling pathways, including NF-κB and mitogen-activated protein kinase pathways. Mechanistically, unlike other members of the ASB family that induce ubiquitination-mediated degradation of their target proteins, ASB1 associates with TAB2 to inhibit K48-linked polyubiquitination and thereby promote the stability of TAB2 upon stimulation of cytokines and lipopolysaccharide (LPS), which indicates that ASB1 plays a noncanonical role to further stabilize the target protein rather than induce its degradation. The deficiency of Asb1 protects mice from Salmonella typhimurium- or LPS-induced septic shock and increases the survival of mice. Moreover, Asb1-deficient mice exhibited less severe colitis and intestinal inflammation induced by dextran sodium sulfate. Given the crucial role of ASB proteins in inflammatory signaling pathways, our study offers insights into the immune regulation in pathogen infection and inflammatory disorders with therapeutic implications.
Collapse
|
24
|
Bolko L, Jiang W, Tawara N, Landon‐Cardinal O, Anquetil C, Benveniste O, Allenbach Y. The role of interferons type I, II and III in myositis: A review. Brain Pathol 2021; 31:e12955. [PMID: 34043262 PMCID: PMC8412069 DOI: 10.1111/bpa.12955] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 03/19/2021] [Indexed: 12/27/2022] Open
Abstract
The classification of idiopathic inflammatory myopathies (IIM) is based on clinical, serological and histological criteria. The identification of myositis-specific antibodies has helped to define more homogeneous groups of myositis into four dominant subsets: dermatomyositis (DM), antisynthetase syndrome (ASyS), sporadic inclusion body myositis (sIBM) and immune-mediated necrotising myopathy (IMNM). sIBM and IMNM patients present predominantly with muscle involvement, whereas DM and ASyS patients present additionally with other extramuscular features, such as skin, lung and joints manifestations. Moreover, the pathophysiological mechanisms are distinct between each myositis subsets. Recently, interferon (IFN) pathways have been identified as key players implicated in the pathophysiology of myositis. In DM, the key role of IFN, especially type I IFN, has been supported by the identification of an IFN signature in muscle, blood and skin of DM patients. In addition, DM-specific antibodies are targeting antigens involved in the IFN signalling pathways. The pathogenicity of type I IFN has been demonstrated by the identification of mutations in the IFN pathways leading to genetic diseases, the monogenic interferonopathies. This constitutive activation of IFN signalling pathways induces systemic manifestations such as interstitial lung disease, myositis and skin rashes. Since DM patients share similar features in the context of an acquired activation of the IFN signalling pathways, we may extend underlying concepts of monogenic diseases to acquired interferonopathy such as DM. Conversely, in ASyS, available data suggest a role of type II IFN in blood, muscle and lung. Indeed, transcriptomic analyses highlighted a type II IFN gene expression in ASyS muscle tissue. In sIBM, type II IFN appears to be an important cytokine involved in muscle inflammation mechanisms and potentially linked to myodegenerative features. For IMNM, currently published data are scarce, suggesting a minor implication of type II IFN. This review highlights the involvement of different IFN subtypes and their specific molecular mechanisms in each myositis subset.
Collapse
Affiliation(s)
- Loïs Bolko
- Division of RheumatologyHopital Maison BlancheReimsFrance
| | - Wei Jiang
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Nozomu Tawara
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Océane Landon‐Cardinal
- Division of RheumatologyCentre hospitalier de l'Université de Montréal (CHUM)CHUM Research CenterMontréalQCCanada
- Department of MedicineUniversité de MontréalMontréalQCCanada
| | - Céline Anquetil
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| | - Yves Allenbach
- Department of Internal Medicine and Clinical ImmunlogySorbonne UniversitéPitié‐Salpêtrière University HospitalParisFrance
- Centre de Recherche en MyologieUMRS974Institut National de la Santé et de la Recherche MédicaleAssociation Institut de MyologieSorbonne UniversitéParisFrance
| |
Collapse
|
25
|
Liu H, Zhang J, Xu X, Lu S, Yang D, Xie C, Jia M, Zhang W, Jin L, Wang X, Shen X, Li F, Wang W, Bao X, Li S, Zhu M, Wang W, Wang Y, Huang Z, Teng H. SARM1 promotes neuroinflammation and inhibits neural regeneration after spinal cord injury through NF-κB signaling. Am J Cancer Res 2021; 11:4187-4206. [PMID: 33754056 PMCID: PMC7977471 DOI: 10.7150/thno.49054] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/17/2021] [Indexed: 12/18/2022] Open
Abstract
Axonal degeneration is a common pathological feature in many acute and chronic neurological diseases such as spinal cord injury (SCI). SARM1 (sterile alpha and TIR motif-containing 1), the fifth TLR (Toll-like receptor) adaptor, has diverse functions in the immune and nervous systems, and recently has been identified as a key mediator of Wallerian degeneration (WD). However, the detailed functions of SARM1 after SCI still remain unclear. Methods: Modified Allen's method was used to establish a contusion model of SCI in mice. Furthermore, to address the function of SARM1 after SCI, conditional knockout (CKO) mice in the central nervous system (CNS), SARM1Nestin-CKO mice, and SARM1GFAP-CKO mice were successfully generated by Nestin-Cre and GFAP-Cre transgenic mice crossed with SARM1flox/flox mice, respectively. Immunostaining, Hematoxylin-Eosin (HE) staining, Nissl staining and behavioral test assays such as footprint and Basso Mouse Scale (BMS) scoring were used to examine the roles of SARM1 pathway in SCI based on these conditional knockout mice. Drugs such as FK866, an inhibitor of SARM1, and apoptozole, an inhibitor of heat shock protein 70 (HSP70), were used to further explore the molecular mechanism of SARM1 in neural regeneration after SCI. Results: We found that SARM1 was upregulated in neurons and astrocytes at early stage after SCI. SARM1Nestin-CKO and SARM1GFAP-CKO mice displayed normal development of the spinal cords and motor function. Interestingly, conditional deletion of SARM1 in neurons and astrocytes promoted the functional recovery of behavior performance after SCI. Mechanistically, conditional deletion of SARM1 in neurons and astrocytes promoted neuronal regeneration at intermediate phase after SCI, and reduced neuroinflammation at SCI early phase through downregulation of NF-κB signaling after SCI, which may be due to upregulation of HSP70. Finally, FK866, an inhibitor of SARM1, reduced the neuroinflammation and promoted the neuronal regeneration after SCI. Conclusion: Our results indicate that SARM1-mediated prodegenerative pathway and neuroinflammation promotes the pathological progress of SCI and anti-SARM1 therapeutics are viable and promising approaches for preserving neuronal function after SCI.
Collapse
|
26
|
Hou P, Yang K, Jia P, Liu L, Lin Y, Li Z, Li J, Chen S, Guo S, Pan J, Wu J, Peng H, Zeng W, Li C, Liu Y, Guo D. A novel selective autophagy receptor, CCDC50, delivers K63 polyubiquitination-activated RIG-I/MDA5 for degradation during viral infection. Cell Res 2021; 31:62-79. [PMID: 32612200 PMCID: PMC7852694 DOI: 10.1038/s41422-020-0362-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Autophagy is a conserved process that delivers cytosolic substances to the lysosome for degradation, but its direct role in the regulation of antiviral innate immunity remains poorly understood. Here, through high-throughput screening, we discovered that CCDC50 functions as a previously unknown autophagy receptor that negatively regulates the type I interferon (IFN) signaling pathway initiated by RIG-I-like receptors (RLRs), the sensors for RNA viruses. The expression of CCDC50 is enhanced by viral infection, and CCDC50 specifically recognizes K63-polyubiquitinated RLRs, thus delivering the activated RIG-I/MDA5 for autophagic degradation. The association of CCDC50 with phagophore membrane protein LC3 is confirmed by crystal structure analysis. In contrast to other known autophagic cargo receptors that associate with either the LIR-docking site (LDS) or the UIM-docking site (UDS) of LC3, CCDC50 can bind to both LDS and UDS, representing a new type of cargo receptor. In mouse models with RNA virus infection, CCDC50 deficiency reduces the autophagic degradation of RIG-I/MDA5 and promotes type I IFN responses, resulting in enhanced viral resistance and improved survival rates. These results reveal a new link between autophagy and antiviral innate immune responses and provide additional insights into the regulatory mechanisms of RLR-mediated antiviral signaling.
Collapse
Affiliation(s)
- Panpan Hou
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Kongxiang Yang
- Modern Virology Research Centre, College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Penghui Jia
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Lan Liu
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yuxin Lin
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Zibo Li
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Jun Li
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Shuliang Chen
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Shuting Guo
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Ji'An Pan
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Junyu Wu
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Hong Peng
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Weijie Zeng
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Chunmei Li
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Yingfang Liu
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China
| | - Deyin Guo
- MOE Key Laboratory of Tropical Disease Control, Centre for Infection and Immunity Studies (CIIS), Seventh Affiliated Hospital, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
27
|
Assouvie A, Rotival M, Hamroune J, Busso D, Romeo PH, Quintana-Murci L, Rousselet G. A genetic variant controls interferon-β gene expression in human myeloid cells by preventing C/EBP-β binding on a conserved enhancer. PLoS Genet 2020; 16:e1009090. [PMID: 33147208 PMCID: PMC7641354 DOI: 10.1371/journal.pgen.1009090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 08/31/2020] [Indexed: 12/01/2022] Open
Abstract
Interferon β (IFN-β) is a cytokine that induces a global antiviral proteome, and regulates the adaptive immune response to infections and tumors. Its effects strongly depend on its level and timing of expression. Therefore, the transcription of its coding gene IFNB1 is strictly controlled. We have previously shown that in mice, the TRIM33 protein restrains Ifnb1 transcription in activated myeloid cells through an upstream inhibitory sequence called ICE. Here, we show that the deregulation of Ifnb1 expression observed in murine Trim33-/- macrophages correlates with abnormal looping of both ICE and the Ifnb1 gene to a 100 kb downstream region overlapping the Ptplad2/Hacd4 gene. This region is a predicted myeloid super-enhancer in which we could characterize 3 myeloid-specific active enhancers, one of which (E5) increases the response of the Ifnb1 promoter to activation. In humans, the orthologous region contains several single nucleotide polymorphisms (SNPs) known to be associated with decreased expression of IFNB1 in activated monocytes, and loops to the IFNB1 gene. The strongest association is found for the rs12553564 SNP, located in the E5 orthologous region. The minor allele of rs12553564 disrupts a conserved C/EBP-β binding motif, prevents binding of C/EBP-β, and abolishes the activation-induced enhancer activity of E5. Altogether, these results establish a link between a genetic variant preventing binding of a transcription factor and a higher order phenotype, and suggest that the frequent minor allele (around 30% worldwide) might be associated with phenotypes regulated by IFN-β expression in myeloid cells. Genome-wide association studies identify multiple genetic variants associated with higher order phenotypes. Pinpointing the causative variant and understanding its molecular mode of action is a complex task. Using a murine model of interferon-β transcriptional deregulation, we characterize a super-enhancer controlling Ifnb1 expression in myeloid cells. The most active enhancer of this locus is conserved in humans, but presents a frequent variant found in around 30% of the population worldwide. This variant prevents binding of the C/EBP-β transcription factor, and is associated with decreased expression of IFNB1 in activated monocytes. When mimicked in the murine enhancer, it abolishes its inducible enhancer activity. Our results describe the molecular link between a point mutation and a cellular phenotype that could influence clinical situations.
Collapse
Affiliation(s)
- Anaïs Assouvie
- Laboratoire Réparation et Transcription dans les cellules Souches, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA/IRCM, Inserm U1274, Fontenay-aux-Roses, France
| | - Maxime Rotival
- Unit of Human Evolutionary Genetics, CNRS UMR2000, Institut Pasteur, Paris, France
| | - Juliette Hamroune
- Plate-forme Génomique, Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | - Didier Busso
- CIGEx, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA/IRCM, Inserm U1274, Fontenay-aux-Roses, France
| | - Paul-Henri Romeo
- Laboratoire Réparation et Transcription dans les cellules Souches, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA/IRCM, Inserm U1274, Fontenay-aux-Roses, France
| | - Lluis Quintana-Murci
- Unit of Human Evolutionary Genetics, CNRS UMR2000, Institut Pasteur, Paris, France
- Chair Human Genomics & Evolution, Collège de France, Paris, France
| | - Germain Rousselet
- Laboratoire Réparation et Transcription dans les cellules Souches, UMRE008 Stabilité Génétique Cellules Souches et Radiations, Université de Paris, Université Paris-Saclay, CEA/IRCM, Inserm U1274, Fontenay-aux-Roses, France
- * E-mail:
| |
Collapse
|
28
|
De Vooght J, Vulsteke JB, De Haes P, Bossuyt X, Lories R, De Langhe E. Anti-TIF1-γ autoantibodies: warning lights of a tumour autoantigen. Rheumatology (Oxford) 2020; 59:469-477. [PMID: 31883334 DOI: 10.1093/rheumatology/kez572] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/25/2019] [Indexed: 11/14/2022] Open
Abstract
Anti-transcription intermediary factor 1 (TIF1)-γ autoantibodies are robustly linked with cancer-associated DM in adults. This review aims to give an overview of the physiological context of TIF1-γ and to determine whether there is a pathophysiological link between anti-TIF1-γ autoantibodies and the occurrence of cancer. Detection of anti-TIF1-γ autoantibodies has a high sensitivity and specificity for cancer-associated DM in adults and is therefore useful for both diagnosis and cancer risk stratification. The function of the autoantigen, TIF1-γ, may provide insight into the mechanism behind this association. TIF1-γ is a ubiquitously present protein involved in various biological pathways, including TGF-β signalling. In cancer, it can act either as a tumour suppressor or promoter, depending on the cellular context and cancer stage. Evolving data provide pathophysiological insights, linking anti-TIF1-γ autoantibodies to both the anti-tumour response and to muscle and skin damage. TIF1-γ expression is increased in muscle and skin tissue of patients with DM. Mutations or loss-of-heterozygosity in TIF1-γ alleles in malignant tissue may result in the expression of tumour-specific neo-antigens stimulating autoantibody production. The newly formed autoantibodies are hypothesized to cross-react with antigens in muscle and skin, driving the development of DM. Based on the current evidence, anti-TIF1-γ autoantibodies should be considered warning lights of a potential tumour autoantigen and should alert the physician to the possibility of an underlying cancer.
Collapse
Affiliation(s)
| | - Jean-Baptiste Vulsteke
- Division of Rheumatology, University Hospitals Leuven, Belgium.,Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Belgium
| | - Petra De Haes
- Division of Dermatology, University Hospitals Leuven, Belgium
| | - Xavier Bossuyt
- Clinical and Diagnostic Immunology, Department of Microbiology and Immunology, KU Leuven, Belgium.,Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, KU, Leuven, Leuven, Belgium
| | - Rik Lories
- Division of Rheumatology, University Hospitals Leuven, Belgium.,Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Belgium
| | - Ellen De Langhe
- Division of Rheumatology, University Hospitals Leuven, Belgium.,Department of Development and Regeneration, Skeletal Biology and Engineering Research Centre, KU Leuven, Belgium
| |
Collapse
|
29
|
Boutanquoi PM, Burgy O, Beltramo G, Bellaye PS, Dondaine L, Marcion G, Pommerolle L, Vadel A, Spanjaard M, Demidov O, Mailleux A, Crestani B, Kolb M, Garrido C, Goirand F, Bonniaud P. TRIM33 prevents pulmonary fibrosis by impairing TGF-β1 signalling. Eur Respir J 2020; 55:13993003.01346-2019. [PMID: 32184320 DOI: 10.1183/13993003.01346-2019] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 03/02/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterised by myofibroblast proliferation and abnormal extracellular matrix accumulation in the lungs. Transforming growth factor (TGF)-β1 initiates key profibrotic signalling involving the SMAD pathway and the small heat shock protein B5 (HSPB5). Tripartite motif-containing 33 (TRIM33) has been reported to negatively regulate TGF-β/SMAD signalling, but its role in fibrogenesis remains unknown. The objective of this study was to elucidate the role of TRIM33 in IPF. METHODS TRIM33 expression was assessed in the lungs of IPF patients and rodent fibrosis models. Bone marrow-derived macrophages (BMDM), primary lung fibroblasts and 3D lung tissue slices were isolated from Trim33-floxed mice and cultured with TGF-β1 or bleomycin (BLM). Trim33 expression was then suppressed by adenovirus Cre recombinase (AdCre). Pulmonary fibrosis was evaluated in haematopoietic-specific Trim33 knockout mice and in Trim33-floxed mice that received AdCre and BLM intratracheally. RESULTS TRIM33 was overexpressed in alveolar macrophages and fibroblasts in IPF patients and rodent fibrotic lungs. Trim33 inhibition in BMDM increased TGF-β1 secretion upon BLM treatment. Haematopoietic-specific Trim33 knockout sensitised mice to BLM-induced fibrosis. In primary lung fibroblasts and 3D lung tissue slices, Trim33 deficiency increased expression of genes downstream of TGF-β1. In mice, AdCre-Trim33 inhibition worsened BLM-induced fibrosis. In vitro, HSPB5 was able to bind directly to TRIM33, thereby diminishing its protein level and TRIM33/SMAD4 interaction. CONCLUSION Our results demonstrate a key role of TRIM33 as a negative regulator of lung fibrosis. Since TRIM33 directly associates with HSPB5, which impairs its activity, inhibitors of TRIM33/HSPB5 interaction may be of interest in the treatment of IPF.
Collapse
Affiliation(s)
- Pierre-Marie Boutanquoi
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Olivier Burgy
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,Division of Pulmonary Sciences and Critical Care Medicine, Dept of Medicine, University of Colorado Denver, Aurora, CO, USA
| | - Guillaume Beltramo
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,Dept of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, Dijon, France.,Reference Center for Rare Lung Diseases, University Hospital, Bourgogne-Franche Comté, Dijon, France
| | | | - Lucile Dondaine
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,Reference Center for Rare Lung Diseases, University Hospital, Bourgogne-Franche Comté, Dijon, France
| | - Guillaume Marcion
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Lenny Pommerolle
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Aurélie Vadel
- INSERM U1152, Faculty of Medicine, University of Bichat, Paris, France
| | - Maximilien Spanjaard
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,Dept of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, Dijon, France
| | - Oleg Demidov
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Arnaud Mailleux
- INSERM U1152, Faculty of Medicine, University of Bichat, Paris, France
| | - Bruno Crestani
- INSERM U1152, Faculty of Medicine, University of Bichat, Paris, France
| | | | - Carmen Garrido
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France
| | - Françoise Goirand
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France.,These authors codirected this work and contributed equally to this work
| | - Philippe Bonniaud
- INSERM U1231, Faculty of Medicine and Pharmacy, University of Bourgogne-Franche Comté, Dijon, France .,Dept of Pulmonary Medicine and Intensive Care Unit, University Hospital, Bourgogne-Franche Comté, Dijon, France.,Reference Center for Rare Lung Diseases, University Hospital, Bourgogne-Franche Comté, Dijon, France.,These authors codirected this work and contributed equally to this work
| |
Collapse
|
30
|
Jiang Y, Wang X, Dong C. Molecular mechanisms of T helper 17 cell differentiation: Emerging roles for transcription cofactors. Adv Immunol 2019; 144:121-153. [PMID: 31699215 DOI: 10.1016/bs.ai.2019.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T helper 17 (Th17) cells, characterized by secretion of IL-17 and IL-17F, are a specialized CD4+ effector T cell lineage that not only facilitates host defense against pathogen infection and maintenance of mucosal barrier, but also potently induces tissue inflammation and autoimmune diseases. Since its discovery in 2005, the developmental program of Th17 cells has been characterized, which involves a number of key cytokines, transcription factors and multiple layers of epigenetic modifications. However, how these mechanisms integrate into the complex regulatory network in Th17 cells has not been well defined. Emerging evidences have revealed essential roles of cofactors in controlling chromosome accessibilities and activities of Th17-specific transcription factors. Moreover, cofactors also act as critical signaling integrators to coordinate multiple signaling pathways and transcriptional programs. Deficiency or dysregulation of these cofactors results in defects in Th17 responses and induction of associated autoimmune diseases. Our lab has recently reported several important cofactors in Th17 cells. Here we summarize our findings regarding this new scenario of developmental regulation of Th17 cells. These findings may benefit the development of innovative strategies to treat autoimmune diseases.
Collapse
Affiliation(s)
- Yu Jiang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing, China.
| |
Collapse
|
31
|
Baloghova N, Lidak T, Cermak L. Ubiquitin Ligases Involved in the Regulation of Wnt, TGF-β, and Notch Signaling Pathways and Their Roles in Mouse Development and Homeostasis. Genes (Basel) 2019; 10:genes10100815. [PMID: 31623112 PMCID: PMC6826584 DOI: 10.3390/genes10100815] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/02/2019] [Accepted: 10/13/2019] [Indexed: 12/20/2022] Open
Abstract
The Wnt, TGF-β, and Notch signaling pathways are essential for the regulation of cellular polarity, differentiation, proliferation, and migration. Differential activation and mutual crosstalk of these pathways during animal development are crucial instructive forces in the initiation of the body axis and the development of organs and tissues. Due to the ability to initiate cell proliferation, these pathways are vulnerable to somatic mutations selectively producing cells, which ultimately slip through cellular and organismal checkpoints and develop into cancer. The architecture of the Wnt, TGF-β, and Notch signaling pathways is simple. The transmembrane receptor, activated by the extracellular stimulus, induces nuclear translocation of the transcription factor, which subsequently changes the expression of target genes. Nevertheless, these pathways are regulated by a myriad of factors involved in various feedback mechanisms or crosstalk. The most prominent group of regulators is the ubiquitin-proteasome system (UPS). To open the door to UPS-based therapeutic manipulations, a thorough understanding of these regulations at a molecular level and rigorous confirmation in vivo are required. In this quest, mouse models are exceptional and, thanks to the progress in genetic engineering, also an accessible tool. Here, we reviewed the current understanding of how the UPS regulates the Wnt, TGF-β, and Notch pathways and we summarized the knowledge gained from related mouse models.
Collapse
Affiliation(s)
- Nikol Baloghova
- Laboratory of Cancer Biology, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, 252 42 Vestec, Czech Republic.
| | - Tomas Lidak
- Laboratory of Cancer Biology, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, 252 42 Vestec, Czech Republic.
| | - Lukas Cermak
- Laboratory of Cancer Biology, Division BIOCEV, Institute of Molecular Genetics of the Czech Academy of Sciences, 252 42 Vestec, Czech Republic.
| |
Collapse
|
32
|
Yu C, Ding Z, Liang H, Zhang B, Chen X. The Roles of TIF1γ in Cancer. Front Oncol 2019; 9:979. [PMID: 31632911 PMCID: PMC6783507 DOI: 10.3389/fonc.2019.00979] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 09/13/2019] [Indexed: 12/22/2022] Open
Abstract
Transcriptional intermediary factor 1 γ (TIF1γ), also known as TRIM33, RFG7, PTC7, or Ectodermin, is an E3 ubiquitin-ligase family member with a ring-box-coiled-coil region. It can regulate TGF-β/Smad signaling in two different ways in different cellular contexts. On one hand, TIF1γ can monoubiquitinate Smad4 to inhibit the formation of Smad2/3/4 nuclear complexes. On the other hand, TIF1γ can function as a cofactor of phosphorylated (p)-Smad2/3, competing with Smad4 to inhibit the formation of the Smad2/3/4 complex. In addition, TIF1γ has been reported to play a role in transcription elongation, cellular differentiation, embryonic development, and mitosis. As transforming growth factor-β (TGF-β) superfamily signaling plays an important role in the occurrence and development of cancer, and TIF1γ was reported to be involved in the regulation of TGF-β superfamily signaling, studies on TIF1γ during the last decade have focused on its role in the development of cancer. However, TIF1γ can function either as a tumor suppressor or promoter in different cellular contexts, yet there are few reviews focusing on the roles of TIF1γ in cancer. Hence, in this paper we systematically review and discuss the roles of TIF1γ in cancer. Firstly, we review the biological features, the regulatory mechanisms and the related signaling pathways of TIF1γ. Next, we illustrate the roles of TIF1γ in different tumors. We then provide a tentative hypothesis that explains the dual roles of TIF1 γ in cancer. Finally, we provide our viewpoint regarding the future developments of cancer research focusing on TIF1γ, especially in relation to the effects of TIF1γ on tumoral immunity.
Collapse
Affiliation(s)
- Chengpeng Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Ferri F, Petit V, Barroca V, Romeo PH. Interplay between FACT subunit SPT16 and TRIM33 can remodel chromatin at macrophage distal regulatory elements. Epigenetics Chromatin 2019; 12:46. [PMID: 31331374 PMCID: PMC6647326 DOI: 10.1186/s13072-019-0288-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/07/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cell type-specific use of cis-acting regulatory elements is mediated by the combinatorial activity of transcription factors involved in lineage determination and maintenance of cell identity. In macrophages, specific transcriptional programs are dictated by the transcription factor PU.1 that primes distal regulatory elements for macrophage identities and makes chromatin competent for activity of stimuli-dependent transcription factors. Although the advances in genome-wide approaches have elucidated the functions of these macrophage-specific distal regulatory elements in transcriptional responses, chromatin structures associated with PU.1 priming and the underlying mechanisms of action of these cis-acting sequences are not characterized. RESULTS Here, we show that, in macrophages, FACT subunit SPT16 can bind to positioned nucleosomes directly flanking PU.1-bound sites at previously uncharacterized distal regulatory elements located near genes essential for macrophage development and functions. SPT16 can interact with the transcriptional co-regulator TRIM33 and binds to half of these sites in a TRIM33-dependent manner. Using the Atp1b3 locus as a model, we show that FACT binds to two positioned nucleosomes surrounding a TRIM33/PU.1-bound site in a region, located 35 kb upstream the Atp1b3 TSS, that interact with the Atp1b3 promoter. At this - 35 kb region, TRIM33 deficiency leads to FACT release, loss of the two positioned nucleosomes, RNA Pol II recruitment and bidirectional transcription. These modifications are associated with higher levels of FACT binding at the Atp1b3 promoter, an increase of RNA Pol II recruitment and an increased expression of Atp1b3 in Trim33-/- macrophages. CONCLUSIONS Thus, sequestering of SPT16/FACT by TRIM33 at PU.1-bound distal regions might represent a new regulatory mechanism for RNA Pol II recruitment and transcription output in macrophages.
Collapse
Affiliation(s)
- Federica Ferri
- CEA/DRF/IBFJ/iRCM/LRTS, 92265, Fontenay-aux-Roses Cedex, France. .,Inserm U967, 92265, Fontenay-aux-Roses Cedex, France. .,Université Paris-Diderot, Paris 7, France. .,Université Paris-Sud, Paris 11, France. .,Equipe labellisée Ligue contre le Cancer, Paris, France.
| | - Vanessa Petit
- CEA/DRF/IBFJ/iRCM/LRTS, 92265, Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265, Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Paris 7, France.,Université Paris-Sud, Paris 11, France.,Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Vilma Barroca
- CEA/DRF/IBFJ/iRCM/LRTS, 92265, Fontenay-aux-Roses Cedex, France.,Inserm U967, 92265, Fontenay-aux-Roses Cedex, France.,Université Paris-Diderot, Paris 7, France.,Université Paris-Sud, Paris 11, France.,Equipe labellisée Ligue contre le Cancer, Paris, France
| | - Paul-Henri Romeo
- CEA/DRF/IBFJ/iRCM/LRTS, 92265, Fontenay-aux-Roses Cedex, France. .,Inserm U967, 92265, Fontenay-aux-Roses Cedex, France. .,Université Paris-Diderot, Paris 7, France. .,Université Paris-Sud, Paris 11, France. .,Equipe labellisée Ligue contre le Cancer, Paris, France.
| |
Collapse
|
34
|
Petit V, Parcelier A, Mathé C, Barroca V, Torres C, Lewandowski D, Ferri F, Gallouët AS, Dalloz M, Dinet O, Boschetti G, Vozenin MC, Roméo PH. TRIM33 deficiency in monocytes and macrophages impairs resolution of colonic inflammation. EBioMedicine 2019; 44:60-70. [PMID: 31130476 PMCID: PMC6604767 DOI: 10.1016/j.ebiom.2019.05.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Mature myeloid cells play a crucial role in Crohn's disease (CD) but the molecular players that regulate their functions in CD are not fully characterized. We and others have shown that TRIM33 is involved in the innate immune response and in the inflammatory response but TRIM33 role in intestinal inflammation is not known. In this study, we investigated the role of TRIM33 in myeloid cells during dextran sulfate sodium (DSS)-induced colitis. METHODS We study the role of TRIM33 during DSS-induced colitis which mimics intestinal inflammation using mice deleted for Trim33 only in mature myeloid cells (Trim33-/- mice) FINDINGS: We first show that Trim33 mRNA level is decreased in CD patient's blood monocytes suggesting a role of TRIM33 in CD. Using Trim33-/- mice, we show that these mice display an impaired resolution of colonic inflammation with an increased number of blood and colon monocytes and a decreased number of colonic macrophages. Trim33-/- monocytes are less competent for recruitment and macrophage differentiation. Finally, during resolution of inflammation, Trim33-/- colonic macrophages display an impaired M1/M2 switch and express a low level of membrane-bound TNF that is associated with an increased number of colonic neutrophils. INTERPRETATION Our study shows an important role of TRIM33 in monocytes/macrophages during DSS-induced colitis and suggests that the decreased expression of TRIM33 in CD patient's blood monocytes might not be a consequence but might be involved in CD progression. FUND: La Ligue contre le Cancer (équipe labelisée), INSERM, CEA, Université Paris-Diderot, Université Paris-Sud.
Collapse
Affiliation(s)
- Vanessa Petit
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France.
| | - Aude Parcelier
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France; Yposkei (Genethon), 91200 Corbeil-Essonnes, France
| | - Cecile Mathé
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France
| | - Vilma Barroca
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France
| | - Claire Torres
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France
| | - Daniel Lewandowski
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France
| | - Federica Ferri
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France
| | - Anne-Sophie Gallouët
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France; CEA/DRF/JACOB/IDMIT, 92265 Fontenay-aux-Roses Cedex, France
| | - Marion Dalloz
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France; Servier, Oncology, 78290 Croissy, France
| | - Océane Dinet
- Hospices Civils de Lyon, Department of Gastroenterology, Centre Hospitalier Lyon Sud, Pierre-Bénite, France; CIRI, International Center for Infectiology Research, INSERM U1111, Lyon, France
| | - Gilles Boschetti
- Hospices Civils de Lyon, Department of Gastroenterology, Centre Hospitalier Lyon Sud, Pierre-Bénite, France; CIRI, International Center for Infectiology Research, INSERM U1111, Lyon, France
| | - Marie-Catherine Vozenin
- Radio-Oncology Research Laboratory, Vaudois University Hospital Centre (CHUV), Epalinges, Switzerland
| | - Paul-Henri Roméo
- CEA/DRF/IBFJ/iRCM/LRTS, 92265 Fontenay-aux-Roses Cedex, France; Inserm U967, 92265 Fontenay-aux-Roses Cedex, France; Université Paris-Diderot, Paris 7, France; Université Paris-Sud, Paris 11, France; Equipe labellisée Ligue contre le Cancer, France.
| |
Collapse
|
35
|
Bai J, Li K, Tang W, Liang Z, Wang X, Feng W, Zhang S, Ren L, Wu S, Han H, Zhao Y. A high-throughput screen for genes essential for PRRSV infection using a piggyBac-based system. Virology 2019; 531:19-30. [DOI: 10.1016/j.virol.2019.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/02/2019] [Accepted: 03/02/2019] [Indexed: 01/11/2023]
|
36
|
Kim Y, Song KS, Sohn EH, Kang SW, Yoo IS, Shim SC, Yoo SJ, Kim J. Anti-TIF1γ antibody and the expression of TIF1γ in idiopathic inflammatory myopathies. Int J Rheum Dis 2018; 22:314-320. [DOI: 10.1111/1756-185x.13424] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/26/2018] [Accepted: 09/28/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Young Kim
- Department of Internal Medicine; Daejeon Veterans Hospital; Daejeon Korea
| | - Kyu Sang Song
- Department of Pathology; Chungnam National University College of Medicine; Daejeon Korea
| | - Eun Hee Sohn
- Department of Neurology; Chungnam National University College of Medicine; Daejeon Korea
| | - Seong Wook Kang
- Department of Internal Medicine; Chungnam National University College of Medicine; Daejeon Korea
| | - In Seol Yoo
- Department of Internal Medicine; Chungnam National University College of Medicine; Daejeon Korea
| | - Seung-Cheol Shim
- Department of Internal Medicine; Chungnam National University College of Medicine; Daejeon Korea
| | - Su-Jin Yoo
- Department of Internal Medicine; Chungnam National University College of Medicine; Daejeon Korea
| | - Jinhyun Kim
- Department of Internal Medicine; Chungnam National University College of Medicine; Daejeon Korea
| |
Collapse
|
37
|
Macrophage production and activation are dependent on TRIM33. Oncotarget 2018; 8:5111-5122. [PMID: 27974684 PMCID: PMC5354896 DOI: 10.18632/oncotarget.13872] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 11/12/2016] [Indexed: 11/25/2022] Open
Abstract
The tripartite motif (TRIM) family of proteins plays important roles in innate immunity and antimicrobial infection. None of these proteins has been shown to directly regulate transcription of genes in monocyte/macrophage except TRIM33 that we have recently shown to be a macrophage specific transcriptional inhibitor of Ifnb1. Using ChIP-seq analyses, we now report that TRIM33 is bound to two fold more genes in immature than in mature myeloid cell lines. When located near the same genes, TRIM33 is bound to different sequences in the two cell lines suggesting a role of TRIM33 in both immature and mature myeloid cells. Accordingly, expression of TRIM33 in immature myeloid cells is necessary for efficient production of small peritoneal macrophages, monocytes and bone marrow derived macrophage (BMDM) and TRIM33 targets a subset of genes involved in the inflammatory response only in mature myeloid cells. Functionally, this targeting is associated with impaired repression of pathways regulating the late phases of lipopolysaccharide (LPS) activation of BMDM and a high sensitivity to LPS in vivo when the trim33 gene is inactivated in mature myeloid cells. These findings pinpoint TRIM33 as an important transcriptional actor of monocyte/macrophage mediated inflammation.
Collapse
|
38
|
Abstract
Innate immunity is traditionally thought of as the first line of defense against pathogens that enter the body. It is typically characterized as a rather weak defense mechanism, designed to restrict pathogen replication until the adaptive immune response generates a tailored response and eliminates the infectious agent. However, intensive research in recent years has resulted in better understanding of innate immunity as well as the discovery of many effector proteins, revealing its numerous powerful mechanisms to defend the host. Furthermore, this research has demonstrated that it is simplistic to strictly separate adaptive and innate immune functions since these two systems often work synergistically rather than sequentially. Here, we provide a broad overview of innate pattern recognition receptors in antiviral defense, with a focus on the TRIM family, and discuss their signaling pathways and mechanisms of action with special emphasis on the intracellular antibody receptor TRIM21.
Collapse
Affiliation(s)
| | - Leo C James
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom.
| |
Collapse
|
39
|
Aussy A, Boyer O, Cordel N. Dermatomyositis and Immune-Mediated Necrotizing Myopathies: A Window on Autoimmunity and Cancer. Front Immunol 2017; 8:992. [PMID: 28871260 PMCID: PMC5566616 DOI: 10.3389/fimmu.2017.00992] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/03/2017] [Indexed: 12/21/2022] Open
Abstract
Autoimmune myopathies (myositides) are strongly associated with malignancy. The link between myositis and cancer, originally noticed by Bohan and Peter in their classification in 1975 (1), has been evidenced by large population-based cohort studies and a recent meta-analysis. The numerous reports of cases in which the clinical course of myositis reflects that of cancer and the short delay between myositis and cancer onset support the notion that myositis may be an authentic paraneoplastic disorder. Thus, cancer-associated myositis raises the question of cancer as a cause rather than a consequence of autoimmunity. Among myositides, dermatomyositis and more recently, although to a lesser extent, immune-mediated necrotizing myopathies are the most documented forms associated with cancer. Interestingly, the current diagnostic approach for myositis is based on the identification of specific antibodies where each antibody determines specific clinical features and outcomes. Recent findings have shown that the autoantibodies anti-TIF1γ, anti-NXP2 and anti-HMGCR are associated with cancers in the course of myositis. Herein, we highlight the fact that the targets of these three autoantibodies involve cellular pathways that intervene in tumor promotion and we discuss the role of cancer mutations as autoimmunity triggers in adult myositis.
Collapse
Affiliation(s)
- Audrey Aussy
- Normandie University, UNIROUEN, INSERM, U1234, Rouen University Hospital, Department of Immunology, Rouen, France
| | - Olivier Boyer
- Normandie University, UNIROUEN, INSERM, U1234, Rouen University Hospital, Department of Immunology, Rouen, France
| | - Nadège Cordel
- Normandie University, UNIROUEN, INSERM, U1234, Rouen University Hospital, Department of Immunology, Rouen, France.,Unit of Dermatology and Internal Medicine, Pointe-à-Pitre University Hospital, University of the French West Indies, Fouillole, Pointe-à-Pitre, Guadeloupe
| |
Collapse
|
40
|
Demy DL, Tauzin M, Lancino M, Le Cabec V, Redd M, Murayama E, Maridonneau-Parini I, Trede N, Herbomel P. Trim33 is essential for macrophage and neutrophil mobilization to developmental or inflammatory cues. J Cell Sci 2017; 130:2797-2807. [PMID: 28724755 DOI: 10.1242/jcs.203471] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 07/10/2017] [Indexed: 01/04/2023] Open
Abstract
Macrophages infiltrate and establish in developing organs from an early stage, often before these have become vascularized. Similarly, leukocytes, in general, can quickly migrate through tissues to any site of wounding. This unique capacity is rooted in their characteristic amoeboid motility, the genetic basis of which is poorly understood. Trim33 (also known as Tif1-γ), a nuclear protein that associates with specific DNA-binding transcription factors to modulate gene expression, has been found to be mainly involved in hematopoiesis and gene regulation mediated by TGF-β. Here, we have discovered that in Trim33-deficient zebrafish embryos, primitive macrophages are unable to colonize the central nervous system to become microglia. Moreover, both macrophages and neutrophils of Trim33-deficient embryos display a reduced basal mobility within interstitial tissues, and a profound lack of a response to inflammatory recruitment signals, including local bacterial infections. Correlatively, Trim33-deficient mouse bone marrow-derived macrophages display a strongly reduced three-dimensional amoeboid mobility in fibrous collagen gels. The transcriptional regulator Trim33 is thus revealed as being essential for the navigation of macrophages and neutrophils towards developmental or inflammatory cues within vertebrate tissues.
Collapse
Affiliation(s)
- Doris Lou Demy
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, 75015 Paris, France.,CNRS, UMR3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Muriel Tauzin
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, 75015 Paris, France.,CNRS, UMR3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Mylène Lancino
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, 75015 Paris, France.,CNRS, UMR3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Véronique Le Cabec
- CNRS UMR5089, IPBS (Institut de Pharmacologie et de Biologie Structurale), 205 route de Narbonne BP64182, 31077 Toulouse, France.,Université de Toulouse, UPS, IPBS, 31077 Toulouse, France
| | - Michael Redd
- University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT 94112, USA
| | - Emi Murayama
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, 75015 Paris, France.,CNRS, UMR3738, 25 rue du Dr Roux, 75015 Paris, France
| | - Isabelle Maridonneau-Parini
- CNRS UMR5089, IPBS (Institut de Pharmacologie et de Biologie Structurale), 205 route de Narbonne BP64182, 31077 Toulouse, France.,Université de Toulouse, UPS, IPBS, 31077 Toulouse, France
| | - Nikolaus Trede
- University of Utah, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT 94112, USA
| | - Philippe Herbomel
- Institut Pasteur, Department of Developmental & Stem Cell Biology, 25 rue du Dr Roux, 75015 Paris, France .,CNRS, UMR3738, 25 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
41
|
Developmental Control of NRAMP1 (SLC11A1) Expression in Professional Phagocytes. BIOLOGY 2017; 6:biology6020028. [PMID: 28467369 PMCID: PMC5485475 DOI: 10.3390/biology6020028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 12/11/2022]
Abstract
NRAMP1 (SLC11A1) is a professional phagocyte membrane importer of divalent metals that contributes to iron recycling at homeostasis and to nutritional immunity against infection. Analyses of data generated by several consortia and additional studies were integrated to hypothesize mechanisms restricting NRAMP1 expression to mature phagocytes. Results from various epigenetic and transcriptomic approaches were collected for mesodermal and hematopoietic cell types and compiled for combined analysis with results of genetic studies associating single nucleotide polymorphisms (SNPs) with variations in NRAMP1 expression (eQTLs). Analyses establish that NRAMP1 is part of an autonomous topologically associated domain delimited by ubiquitous CCCTC-binding factor (CTCF) sites. NRAMP1 locus contains five regulatory regions: a predicted super-enhancer (S-E) key to phagocyte-specific expression; the proximal promoter; two intronic areas, including 3' inhibitory elements that restrict expression during development; and a block of upstream sites possibly extending the S-E domain. Also the downstream region adjacent to the 3' CTCF locus boundary may regulate expression during hematopoiesis. Mobilization of the locus 14 predicted transcriptional regulatory elements occurs in three steps, beginning with hematopoiesis; at the onset of myelopoiesis and through myelo-monocytic differentiation. Basal expression level in mature phagocytes is further influenced by genetic variation, tissue environment, and in response to infections that induce various epigenetic memories depending on microorganism nature. Constitutively associated transcription factors (TFs) include CCAAT enhancer binding protein beta (C/EBPb), purine rich DNA binding protein (PU.1), early growth response 2 (EGR2) and signal transducer and activator of transcription 1 (STAT1) while hypoxia-inducible factors (HIFs) and interferon regulatory factor 1 (IRF1) may stimulate iron acquisition in pro-inflammatory conditions. Mouse orthologous locus is generally conserved; chromatin patterns typify a de novo myelo-monocytic gene whose expression is tightly controlled by TFs Pu.1, C/ebps and Irf8; Irf3 and nuclear factor NF-kappa-B p 65 subunit (RelA) regulate expression in inflammatory conditions. Functional differences in the determinants identified at these orthologous loci imply that species-specific mechanisms control gene expression.
Collapse
|
42
|
Trim33/Tif1γ is involved in late stages of granulomonopoiesis in mice. Exp Hematol 2016; 44:727-739.e6. [DOI: 10.1016/j.exphem.2016.04.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/14/2016] [Accepted: 04/16/2016] [Indexed: 12/30/2022]
|