1
|
Köhler P, Ribeiro A, Honarpisheh M, von Rauchhaupt E, Lorenz G, Li C, Martin L, Steiger S, Lindenmeyer M, Schmaderer C, Anders HJ, Thomasova D, Lech M. Podocyte A20/TNFAIP3 Controls Glomerulonephritis Severity via the Regulation of Inflammatory Responses and Effects on the Cytoskeleton. Cells 2025; 14:381. [PMID: 40072109 PMCID: PMC11898495 DOI: 10.3390/cells14050381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/22/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
A20/Tnfaip3, an early NF-κB response gene and key negative regulator of NF-κB signaling, suppresses proinflammatory responses. Its ubiquitinase and deubiquitinase activities mediate proteasomal degradation within the NF-κB pathway. This study investigated the involvement of A20 signaling alterations in podocytes in the development of kidney injury. The phenotypes of A20Δpodocyte (podocyte-specific knockout of A20) mice were compared with those of control mice at 6 months of age to identify spontaneous changes in kidney function. A20Δpodocyte mice presented elevated serum urea nitrogen and creatinine levels, along with increased accumulation of inflammatory cells-neutrophils and macrophages-within the glomeruli. Additionally, A20Δpodocyte mice displayed significant podocyte loss. Ultrastructural analysis of A20 podocyte-knockout mouse glomeruli revealed hypocellularity of the glomerular tuft, expansion of the extracellular matrix, podocytopenia associated with foot process effacement, karyopyknosis, micronuclei, and podocyte detachment. In addition to podocyte death, we also observed damage to intracapillary endothelial cells with vacuolation of the cytoplasm and condensation of nuclear chromatin. A20 expression downregulation and CRISPR-Cas9 genome editing targeting A20 in a podocyte cell line confirmed these findings in vitro, highlighting the significant contribution of A20 activity in podocytes to glomerular injury pathogenesis. Finally, we analyzed TNFAIP3 transcription levels alongside genes involved in apoptosis, anoikis, NF-κB regulation, and cell attachment in glomerular and tubular compartments of kidney biopsies of patients with various renal diseases.
Collapse
Affiliation(s)
- Paulina Köhler
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Andrea Ribeiro
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Mohsen Honarpisheh
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Ekaterina von Rauchhaupt
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Georg Lorenz
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Chenyu Li
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Lucas Martin
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Maja Lindenmeyer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Christoph Schmaderer
- Klinikum Rechts der Isar, Department of Nephrology, Technical University Munich (TUM), 80333 München, Germany
| | - Hans-Joachim Anders
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Dana Thomasova
- Institute of Biology and Medical Genetics, 2nd Faculty of Medicine, Charles University Prague and University Hospital Motol, 15006 Prague, Czech Republic
| | - Maciej Lech
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University (LMU) Hospital, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| |
Collapse
|
2
|
Layzell S, Barbarulo A, van Loo G, Beyaert R, Seddon B. NF-κB regulated expression of A20 controls IKK dependent repression of RIPK1 induced cell death in activated T cells. Cell Death Differ 2025; 32:256-270. [PMID: 39327505 PMCID: PMC11802744 DOI: 10.1038/s41418-024-01383-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
IKK signalling is essential for survival of thymocytes by repressing RIPK1 induced cell death rather than its canonical function of activating NF-κB. The role of IKK signalling in activated T cells is unclear. To investigate this, we analysed activation of IKK2 deficient T cells. While TCR triggering was normal, proliferation and expansion was profoundly impaired. This was not due to defective cell cycle progression, rather dividing T cells became sensitised to TNF induced cell death, since inhibition of RIPK1 kinase activity rescued cell survival. Gene expression analysis of activated IKK2 deficient T cells revealed defective expression of Tnfaip3, that encodes A20, a negative regulator of NF-κB. To test whether A20 expression was required to protect IKK2 deficient T cells from cell death, we generated mice with T cells lacking both A20 and IKK2. Doing this resulted in near complete loss of peripheral T cells, in contrast to mice lacking one or other gene. Strikingly, this phenotype was completely reversed by inactivation of RIPK1 kinase activity in vivo. Together, our data show that IKK signalling in activated T cells protects against RIPK1 dependent death, both by direct phosphorylation of RIPK1 and through NF-κB mediated induction of A20, that we identify for the first time as a key modulator of RIPK1 activity in T cells.
Collapse
Affiliation(s)
- Scott Layzell
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK
| | - Alessandro Barbarulo
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK
| | - Geert van Loo
- VIB-UGent Center for Inflammation Research, UGent Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Gent, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, UGent Department for Biomedical Molecular Biology, Unit of Molecular Signal Transduction in Inflammation, Gent, Belgium
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, The Pears Building, Hampstead, London, UK.
| |
Collapse
|
3
|
Diehl C, Soberón V, Baygün S, Chu Y, Mandelbaum J, Kraus L, Engleitner T, Rudelius M, Fangazio M, Daniel C, Bortoluzzi S, Helmrath S, Singroul P, Gölling V, Osorio Barrios F, Seyhan G, Oßwald L, Kober-Hasslacher M, Zeng T, Öllinger R, Afzali AM, Korn T, Honarpisheh M, Lech M, Ul Ain Q, Pircher J, Imširović V, Jelenčić V, Wensveen FM, Passerini V, Bärthel S, Bhagat G, Dominguez-Sola D, Saur D, Steiger K, Rad R, Pasqualucci L, Weigert O, Schmidt-Supprian M. Hyperreactive B cells instruct their elimination by T cells to curb autoinflammation and lymphomagenesis. Immunity 2025; 58:124-142.e15. [PMID: 39729992 DOI: 10.1016/j.immuni.2024.11.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/14/2024] [Accepted: 11/26/2024] [Indexed: 12/29/2024]
Abstract
B cell immunity carries the inherent risk of deviating into autoimmunity and malignancy, which are both strongly associated with genetic variants or alterations that increase immune signaling. Here, we investigated the interplay of autoimmunity and lymphoma risk factors centered around the archetypal negative immune regulator TNFAIP3/A20 in mice. Counterintuitively, B cells with moderately elevated sensitivity to stimulation caused fatal autoimmune pathology, while those with high sensitivity did not. We resolved this apparent paradox by identifying a rheostat-like cytotoxic T cell checkpoint. Cytotoxicity was instructed by and directed against B cells with high intrinsic hyperresponsiveness, while less reactive cells were spared. Removing T cell control restored a linear relationship between intrinsic B cell reactivity and lethal lymphoproliferation, lymphomagenesis, and autoinflammation. We thus identify powerful T cell-mediated negative feedback control of inherited and acquired B cell pathogenicity and define a permissive window for autoimmunity to emerge.
Collapse
Affiliation(s)
- Carina Diehl
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Valeria Soberón
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Seren Baygün
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Yuanyuan Chu
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Jonathan Mandelbaum
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Laura Kraus
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Engleitner
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University, 80337 Munich, Germany
| | - Marco Fangazio
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Christoph Daniel
- Department of Nephropathology, Faculty of Medicine, Friedrich-Alexander University (FAU) Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Sabrina Bortoluzzi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Sabine Helmrath
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Pankaj Singroul
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Vanessa Gölling
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Francisco Osorio Barrios
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Gönül Seyhan
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Lena Oßwald
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; Department of Medicine III, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Maike Kober-Hasslacher
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany
| | - Theodor Zeng
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Rupert Öllinger
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Ali M Afzali
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Department of Neurology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Mohsen Honarpisheh
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Maciej Lech
- Renal Division, Department of Medicine IV, Faculty of Medicine, Ludwig-Maximilians-University, 80336 Munich, Germany
| | - Qurrat Ul Ain
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Joachim Pircher
- Department of Medicine I, Faculty of Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany; Partner site Munich Heart Alliance, DZHK (German Centre for Cardiovascular Research), 80802 Munich, Germany
| | - Vanna Imširović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Vedrana Jelenčić
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Felix M Wensveen
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Verena Passerini
- Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Stefanie Bärthel
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Govind Bhagat
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - David Dominguez-Sola
- Departments of Oncological Sciences and Pathology, Tisch Cancer Institute, Lipschultz Precision Immunology Institute, Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dieter Saur
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Chair of Translational Cancer Research and Institute of Experimental Cancer Therapy, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Katja Steiger
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY 10032, USA
| | - Oliver Weigert
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Laboratory for Experimental Leukemia and Lymphoma Research (ELLF), Faculty of Medicine, Department of Medicine III, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Marc Schmidt-Supprian
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675 Munich, Germany; Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany; Max-Planck Institute of Biochemistry, 82152 Planegg, Germany.
| |
Collapse
|
4
|
Schnizler S, Naumann M, Vieth M. Differential expression of the ubiquitin-editing enzyme A20 in gastric biopsies indicates the severity of disease. Histochem Cell Biol 2024; 163:22. [PMID: 39738689 PMCID: PMC11978676 DOI: 10.1007/s00418-024-02345-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2024] [Indexed: 01/02/2025]
Abstract
A20, an ubiquitin-editing enzyme, plays a pivotal role in regulating cell signaling and immune responses. Dysregulated A20 expression has been associated with various pathological conditions, including inflammatory diseases and malignancies, where its expression levels often correlate with differing prognoses in solid tumors. This study aimed to explore the expression and cellular localization of A20 in both nonpathological and diseased human gastric tissues to gain deeper insights into its involvement in gastric pathologies. We analyzed paraffin-embedded gastric tissue samples from 326 patients. A20 expression was assessed using immunohistochemistry (IHC) with results categorized according to the Remmele and Stegner immunoreactive score (IRS). The study compared A20 expression across a spectrum of gastric pathologies, including Helicobacter pylori (HP) gastritis, autoimmune gastritis (A-gastritis), reactive gastropathy (C-gastritis), Ex-HP-gastritis, adenomas, and adenocarcinomas, with nonpathological gastric mucosa serving as a baseline. Our findings demonstrate a significant increase in A20 expression in HP-gastritis (p = 0.019), A-gastritis (p = 0.001), adenomas (p < 0.001), and adenocarcinomas (p < 0.001). Conversely, no significant differences in A20 expression were observed in C-gastritis or Ex-HP-gastritis cases.
Collapse
Affiliation(s)
- Stephan Schnizler
- Institute of Pathology, Klinikum Bayreuth, 95445, Bayreuth, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| | - Michael Naumann
- Institute of Pathology, Klinikum Bayreuth, 95445, Bayreuth, Germany.
- Institute of Experimental Internal Medicine, Otto-Von-Guericke-Universität Magdeburg, 39120, Magdeburg, Germany.
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, 95445, Bayreuth, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054, Erlangen, Germany
| |
Collapse
|
5
|
Liang J, Wang N, Yao Y, Wang Y, An X, Wang H, Liu H, Jiang Y, Li H, Cheng X, Xu J, Liang X, Lou J, Xin Z, Zhang T, Wang X, Lin W. NEDD4L mediates intestinal epithelial cell ferroptosis to restrict inflammatory bowel diseases and colorectal tumorigenesis. J Clin Invest 2024; 135:e173994. [PMID: 39688910 PMCID: PMC11785928 DOI: 10.1172/jci173994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/10/2024] [Indexed: 12/18/2024] Open
Abstract
Various factors play key roles in maintaining intestine homeostasis. Disruption of the balance may lead to inflammatory bowel diseases and even colorectal cancer (CRC). Loss or gain of function of many key proteins can result in dysregulated intestinal homeostasis. Our research demonstrated that neural precursor cells expressed developmentally downregulated 4-like protein (NEDD4L, or NEDD4-2), a type of HECT family E3 ubiquitin ligase, played an important role in maintaining intestinal homeostasis. NEDD4L expression was significantly inhibited in intestinal epithelial cells (IECs) of patients with Crohn's disease, ulcerative colitis, and CRC. Global KO of NEDD4L or its deficiency in IECs exacerbated colitis induced by dextran sulfate sodium (DSS) and 2,4,6-trinitrobenzene sulfonic acid (TNBS) and CRC induced by azoxymethane and DSS. Mechanistically, NEDD4L deficiency in IECs inhibited expression of the key ferroptosis regulator glutathione peroxidase 4 (GPX4) by reducing the protein expression of solute carrier family 3 member 2 (SLC3A2) without affecting its gene expression, ultimately promoting DSS-induced IEC ferroptosis. Importantly, ferroptosis inhibitors reduced the susceptibility of NEDD4L-deficient mice to colitis and colitis-associated CRC. Thus, NEDD4L is an important regulator in IEC ferroptosis, maintaining intestinal homeostasis, making it a potential clinical target for diagnosing and treating IBDs.
Collapse
Affiliation(s)
- Jingjing Liang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ning Wang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yihan Yao
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi’an, China
| | - Xiang An
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Haofei Wang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Huan Liu
- Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Yu Jiang
- Department of Clinical Laboratory Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hui Li
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Zhejiang, China
| | | | | | - Xiaojing Liang
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lou
- Department of Respiratory and Critical Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zengfeng Xin
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Ting Zhang
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojian Wang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Wenlong Lin
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- School of Medicine, Hangzhou City University, Hangzhou, China
| |
Collapse
|
6
|
Faghih M, Moshiri M, Mazrouei Arani N, Ahmadzadeh F, Jafari N, Ghasemi M, Abediankenari S. Evaluation of TNF-α and IFN-γ primed conditioned medium of mesenchymal stem cell in acetic acid-induced mouse model of acute colitis. Cell Immunol 2024; 405-406:104876. [PMID: 39342814 DOI: 10.1016/j.cellimm.2024.104876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
IBD, an autoimmune-inflammatory disorder that affects people who are genetically prone to inflammation. There is a lot of interest in MSC-CM therapy, especially when primed with TNF-α + IFN-γ. Throughout the study, data were collected on the percentage of apoptotic cells, gene expression of ZO-1, Foxp3, GATA3, IDO-1, Muc2, T-bet, Notch1, TNFR2, and ROR-γt, colon weight and length, histopathological analysis, and DAI. TNF-α and IL-10 levels were assessed in addition to the NO level. The results suggest that primed MSC-CM improved DAI, mucosal deterioration, intestinal inflammation and NO concentration. The amount of TNF-α was decreased, but IL-10 and the colon's percentage of apoptotic cells was increased. The mRNA expression of ZO-1, Foxp3, GATA3, IDO-1, and Muc2 genes increased greatly in the treatment groups, while the expression of T-bet, Notch1, TNFR2, and ROR-γt genes has decreased. These studies suggest that primed MSC-CM may combine with common treatments to improve responsiveness.
Collapse
MESH Headings
- Animals
- Mice
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Colitis/chemically induced
- Colitis/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- Disease Models, Animal
- GATA3 Transcription Factor/metabolism
- GATA3 Transcription Factor/genetics
- Mesenchymal Stem Cells/metabolism
- Culture Media, Conditioned/pharmacology
- T-Box Domain Proteins/metabolism
- T-Box Domain Proteins/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Interleukin-10/metabolism
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Mucin-2/metabolism
- Mucin-2/genetics
- Zonula Occludens-1 Protein/metabolism
- Zonula Occludens-1 Protein/genetics
- Apoptosis/drug effects
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Male
- Colon/pathology
- Colon/metabolism
- Receptor, Notch1/metabolism
- Receptor, Notch1/genetics
- Acute Disease
Collapse
Affiliation(s)
- Manizhe Faghih
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, IRAN; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mona Moshiri
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, IRAN; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nader Mazrouei Arani
- Anatomical Research Center, Kashan University of Medical Sciences and Health Services, kashan, IRAN
| | - Fatemeh Ahmadzadeh
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, IRAN; Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narjes Jafari
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Maryam Ghasemi
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department of Pathology, School of Medicine, Mazandaran University of Medical Sciences, Sari, IRAN
| | - Saeid Abediankenari
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, IRAN.
| |
Collapse
|
7
|
Shen J, Lou L, Du X, Zhou B, Xu Y, Mei F, Wu L, Li J, Waisman A, Ruan J, Wang X. YOD1 sustains NOD2-mediated protective signaling in colitis by stabilizing RIPK2. EMBO Rep 2024; 25:4827-4845. [PMID: 39333628 PMCID: PMC11549337 DOI: 10.1038/s44319-024-00276-6] [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: 03/08/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a disorder causing chronic inflammation in the gastrointestinal tract, and its pathophysiological mechanisms are still under investigation. Here, we find that mice deficient of YOD1, a deubiquitinating enzyme, are highly susceptible to dextran sulfate sodium (DSS)-induced colitis. The bone marrow transplantation experiment reveals that YOD1 derived from hematopoietic cells inhibits DSS colitis. Moreover, YOD1 exerts its protective role by promoting nucleotide-binding oligomerization domain 2 (NOD2)-mediated physiological inflammation in macrophages. Mechanistically, YOD1 inhibits the proteasomal degradation of receptor-interacting serine/threonine kinase 2 (RIPK2) by reducing its K48 polyubiquitination, thereby increasing RIPK2 abundance to enhance NOD2 signaling. Consistently, the protective function of muramyldipeptide, a NOD2 ligand, in experimental colitis is abolished in mice deficient of YOD1. Importantly, YOD1 is upregulated in colon-infiltrating macrophages in patients with colitis. Collectively, this study identifies YOD1 as a novel regulator of colitis.
Collapse
Affiliation(s)
- Jiangyun Shen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Liyan Lou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Xue Du
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Bincheng Zhou
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Yanqi Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Fuqi Mei
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | - Liangrong Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
- Department of Pharmacy, Yiwu Central Hospital, 322099, Yiwu, China
| | - Jianmin Li
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, 325000, Wenzhou, China
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, 55131, Mainz, Germany
| | - Jing Ruan
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, 325000, Wenzhou, China.
| | - Xu Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health); School of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
8
|
Devi MB, Bhattacharya A, Kumar A, Singh CT, Das S, Sarma HK, Mukherjee AK, Khan MR. Potential probiotic Lactiplantibacillus plantarum strains alleviate TNF-α by regulating ADAM17 protein and ameliorate gut integrity through tight junction protein expression in in vitro model. Cell Commun Signal 2024; 22:520. [PMID: 39468700 PMCID: PMC11514838 DOI: 10.1186/s12964-024-01900-7] [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] [Received: 08/01/2024] [Accepted: 10/19/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Lactiplantibacillus species are extensively studied for their ability to regulate host immune responses and functional therapeutic potentials. Nevertheless, there is a lack of understanding on the mechanisms of interactions with the hosts during immunoregulatory activities. METHODS Two Lactiplantibacillus plantarum strains MKMB01 and MKMB02 were tested for probiotic potential following Indian Council of Medical Research (ICMR) guidelines. Human colorectal adenocarcinoma cells such as HT-29, caco-2, and human monocytic cell THP-1 were also used to study the potential of MKMB01 and MKMB02 in regulating the host immune response when challenged with enteric pathogen Salmonella enterica typhimurium. Cells were pre-treated with MKMB01 and MKMB02 for 4 h and then stimulated with Salmonella. qRT-PCR and ELISA were used to analyze the genes and protein expression. Confocal microscopy and field emission scanning electron microscopy (FESEM) were used to visualize the effects. An Agilent Seahorse XF analyzer was used to determine real-time mitochondrial functioning. RESULTS Both probiotic strains could defend against Salmonella by maintaining gut integrity via expressing tight junction proteins (TJPs), MUC-2, and toll-like receptors (TLRs) negative regulators such as single Ig IL-1-related receptor (SIGIRR), toll-interacting protein (Tollip), interleukin-1 receptor-associated kinase (IRAK)-M, A20, and anti-inflammatory transforming growth factor-β and interleukin-10. Both strains also downregulated the expression of pro-inflammatory cytokines/chemokines interleukin-1β, monocyte chemoattractant protein (MCP)-1, tumor necrosis factor-alpha (TNF-α), interleukin 6, and nitric oxide (NO). Moreover, TNF-α sheddase protein, a disintegrin and metalloproteinase domain 17 (ADAM17), and its regulator iRhom2 were downregulated by both strains. Moreover, the bacteria also ameliorated Salmonella-induced mitochondrial dysfunction by restoring bioenergetic profiles, such as non-mitochondrial respiration, spare respiratory capacity (SRC), basal respiration, adenosine triphosphate (ATP) production, and maximal respiration. CONCLUSIONS MKMB01 and MKMB02 can reduce pathogen-induced gut-associated disorders and therefore should be further explored for their probiotic potential.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Anupam Bhattacharya
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Arun Kumar
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Chingtham Thanil Singh
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Santanu Das
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
| | - Hridip Kumar Sarma
- Department of Biotechnology, Gauhati University, Guwahati, 781014, Assam, India
| | - Ashis K Mukherjee
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India
- Microbial Biotechnology and Protein Research laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Mojibur R Khan
- Division of Life Sciences, Institute of Advanced Study in Science and Technology (IASST), Guwahati, 781035, Assam, India.
| |
Collapse
|
9
|
Wallaeys C, Garcia-Gonzalez N, Timmermans S, Vandewalle J, Vanderhaeghen T, De Beul S, Dufoor H, Eggermont M, Moens E, Bosteels V, De Rycke R, Thery F, Impens F, Verbanck S, Lienenklaus S, Janssens S, Blumberg RS, Iwawaki T, Libert C. Paneth cell TNF signaling induces gut bacterial translocation and sepsis. Cell Host Microbe 2024; 32:1725-1743.e7. [PMID: 39243761 DOI: 10.1016/j.chom.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 06/20/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
The cytokine tumor necrosis factor (TNF) plays important roles in limiting infection but is also linked to sepsis. The mechanisms underlying these paradoxical roles are unclear. Here, we show that TNF limits the antimicrobial activity of Paneth cells (PCs), causing bacterial translocation from the gut to various organs. This TNF-induced lethality does not occur in mice with a PC-specific deletion in the TNF receptor, P55. In PCs, TNF stimulates the IFN pathway and ablates the steady-state unfolded protein response (UPR), effects not observed in mice lacking P55 or IFNAR1. TNF triggers the transcriptional downregulation of IRE1 key genes Ern1 and Ern2, which are key mediators of the UPR. This UPR deficiency causes a significant reduction in antimicrobial peptide production and PC antimicrobial activity, causing bacterial translocation to organs and subsequent polymicrobial sepsis, organ failure, and death. This study highlights the roles of PCs in bacterial control and therapeutic targets for sepsis.
Collapse
Affiliation(s)
- Charlotte Wallaeys
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Natalia Garcia-Gonzalez
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Steven Timmermans
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Jolien Vandewalle
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Tineke Vanderhaeghen
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Somara De Beul
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Hester Dufoor
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Melanie Eggermont
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Elise Moens
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium
| | - Victor Bosteels
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Laboratory for ER Stress and Inflammation, VIB-UniversityGent Center for Inflammation Research, Ghent 9052, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent 9000, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium; VIB Center for Inflammation Research and Bioimaging Core, VIB, Ghent 9052, Belgium
| | - Fabien Thery
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent 9052, Belgium
| | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, VIB, Ghent 9052, Belgium; Department of Biomolecular Medicine, Ghent University, Ghent 9052, Belgium; VIB Proteomics Core, VIB, Ghent 9052, Belgium
| | - Serge Verbanck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Stefan Lienenklaus
- Institute of Laboratory Animal Science, Hannover Medical School, Hannover 30625, Germany
| | - Sophie Janssens
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Laboratory for ER Stress and Inflammation, VIB-UniversityGent Center for Inflammation Research, Ghent 9052, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent 9000, Belgium
| | - Richard S Blumberg
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku, Ishikawa 920-0293, Japan
| | - Claude Libert
- VIB Center for Inflammation Research, VIB, Ghent 9052, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent 9052, Belgium.
| |
Collapse
|
10
|
Du X, Xu J, Mei F, Shen J, Zhou B, Zhu Z, Li Z, Su X, Li J, Schlüter D, Ruan J, Wang X. Deubiquitination of RIPK2 by OTUB2 augments NOD2 signalling and protective effects in intestinal inflammation. Clin Transl Med 2024; 14:e70038. [PMID: 39358938 PMCID: PMC11446981 DOI: 10.1002/ctm2.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 09/02/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract, but the molecular mechanisms underlying IBD are incompletely understood. In this study, we explored the role and regulating mechanism of otubain 2 (OTUB2), a deubiquitinating enzyme, in IBD. METHODS To study the function of OTUB2 in IBD, we generated Otub2-/- mice and treated them with dextran sulfate sodium (DSS) to induce experimental colitis. Bone marrow transplantation was performed to identify the cell populations that were affected by OTUB2 in colitis. The molecular mechanism of OTUB2 in signal transduction was studied by various biochemical methods. RESULTS OTUB2 was highly expressed in colon-infiltrating macrophages in both humans with IBD and mice with DSS-induced experimental colitis. Colitis was significantly aggravated in Otub2-/- mice and bone marrow chimeric mice receiving Otub2-/- bone marrow. OTUB2-deficiency impaired the production of cytokines and chemokines in macrophages in response to the NOD2 agonist muramyl dipeptide (MDP). Upon MDP stimulation, OTUB2 promoted NOD2 signaling by stabilizing RIPK2. Mechanistically, OTUB2 inhibited the proteasomal degradation of RIPK2 by removing K48-linked polyubiquitination on RIPK2, which was mediated by the active C51 residue in OTUB2. In mice, OTUB2 ablation abolished the protective effects of MDP administration in colitis. CONCLUSION This study identified OTUB2 as a novel regulator of intestinal inflammation.
Collapse
Affiliation(s)
- Xue Du
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jun Xu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Fuqi Mei
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jiangyun Shen
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Bincheng Zhou
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Zhenhu Zhu
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Zhongding Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Xian Su
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| | - Jianmin Li
- Department of PathologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Dirk Schlüter
- Hannover Medical SchoolInstitute of Medical Microbiology and Hospital EpidemiologyHannoverGermany
| | - Jing Ruan
- Department of PathologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xu Wang
- School of Pharmaceutical SciencesWenzhou Medical UniversityWenzhouChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)WenzhouChina
| |
Collapse
|
11
|
Petta I, Thorp M, Ciers M, Blancke G, Boon L, Meese T, Van Nieuwerburgh F, Wullaert A, Grencis R, Elewaut D, van Loo G, Vereecke L. Myeloid A20 is critical for alternative macrophage polarization and type-2 immune-mediated helminth resistance. Front Immunol 2024; 15:1373745. [PMID: 38680500 PMCID: PMC11045979 DOI: 10.3389/fimmu.2024.1373745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 03/21/2024] [Indexed: 05/01/2024] Open
Abstract
Background Protective immunity against intestinal helminths requires induction of robust type-2 immunity orchestrated by various cellular and soluble effectors which promote goblet cell hyperplasia, mucus production, epithelial proliferation, and smooth muscle contractions to expel worms and re-establish immune homeostasis. Conversely, defects in type-2 immunity result in ineffective helminth clearance, persistent infection, and inflammation. Macrophages are highly plastic cells that acquire an alternatively activated state during helminth infection, but they were previously shown to be dispensable for resistance to Trichuris muris infection. Methods We use the in vivo mouse model A20myel-KO, characterized by the deletion of the potent anti-inflammatory factor A20 (TNFAIP3) specifically in the myeloid cells, the excessive type-1 cytokine production, and the development of spontaneous arthritis. We infect A20myel-KO mice with the gastrointestinal helminth Trichuris muris and we analyzed the innate and adaptive responses. We performed RNA sequencing on sorted myeloid cells to investigate the role of A20 on macrophage polarization and type-2 immunity. Moreover, we assess in A20myel-KO mice the pharmacological inhibition of type-1 cytokine pathways on helminth clearance and the infection with Salmonella typhimurium. Results We show that proper macrophage polarization is essential for helminth clearance, and we identify A20 as an essential myeloid factor for the induction of type-2 immune responses against Trichuris muris. A20myel-KO mice are characterized by persistent Trichuris muris infection and intestinal inflammation. Myeloid A20 deficiency induces strong classical macrophage polarization which impedes anti-helminth type-2 immune activation; however, it promotes detrimental Th1/Th17 responses. Antibody-mediated neutralization of the type-1 cytokines IFN-γ, IL-18, and IL-12 prevents myeloid-orchestrated Th1 polarization and re-establishes type-2-mediated protective immunity against T. muris in A20myel-KO mice. In contrast, the strong Th1-biased immunity in A20myel-KO mice offers protection against Salmonella typhimurium infection. Conclusions We hereby identify A20 as a critical myeloid factor for correct macrophage polarization and appropriate adaptive mucosal immunity in response to helminth and enteric bacterial infection.
Collapse
Affiliation(s)
- Ioanna Petta
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Marie Thorp
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Maarten Ciers
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Gillian Blancke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | | | - Tim Meese
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
- NXTGNT, Ghent University, Ghent, Belgium
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Ghent University, Ghent, Belgium
- NXTGNT, Ghent University, Ghent, Belgium
| | - Andy Wullaert
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
- Cell Death Signaling Lab, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Richard Grencis
- Lydia Becker Institute of Immunology and Inflammation, Wellcome Centre for Cell Matrix Research, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Dirk Elewaut
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Lars Vereecke
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
12
|
Srinivasan S, Kancheva D, De Ren S, Saito T, Jans M, Boone F, Vandendriessche C, Paesmans I, Maurin H, Vandenbroucke RE, Hoste E, Voet S, Scheyltjens I, Pavie B, Lippens S, Schwabenland M, Prinz M, Saido T, Bottelbergs A, Movahedi K, Lamkanfi M, van Loo G. Inflammasome signaling is dispensable for ß-amyloid-induced neuropathology in preclinical models of Alzheimer's disease. Front Immunol 2024; 15:1323409. [PMID: 38352874 PMCID: PMC10863058 DOI: 10.3389/fimmu.2024.1323409] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Background Alzheimer's disease (AD) is the most common neurodegenerative disorder affecting memory and cognition. The disease is accompanied by an abnormal deposition of ß-amyloid plaques in the brain that contributes to neurodegeneration and is known to induce glial inflammation. Studies in the APP/PS1 mouse model of ß-amyloid-induced neuropathology have suggested a role for inflammasome activation in ß-amyloid-induced neuroinflammation and neuropathology. Methods Here, we evaluated the in vivo role of microglia-selective and full body inflammasome signalling in several mouse models of ß-amyloid-induced AD neuropathology. Results Microglia-specific deletion of the inflammasome regulator A20 and inflammasome effector protease caspase-1 in the AppNL-G-F and APP/PS1 models failed to identify a prominent role for microglial inflammasome signalling in ß-amyloid-induced neuropathology. Moreover, global inflammasome inactivation through respectively full body deletion of caspases 1 and 11 in AppNL-G-F mice and Nlrp3 deletion in APP/PS1 mice also failed to modulate amyloid pathology and disease progression. In agreement, single-cell RNA sequencing did not reveal an important role for Nlrp3 signalling in driving microglial activation and the transition into disease-associated states, both during homeostasis and upon amyloid pathology. Conclusion Collectively, these results question a generalizable role for inflammasome activation in preclinical amyloid-only models of neuroinflammation.
Collapse
Affiliation(s)
- Sahana Srinivasan
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Daliya Kancheva
- Brain and Systems Immunology Lab, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie De Ren
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
- Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Maude Jans
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Fleur Boone
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Charysse Vandendriessche
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ine Paesmans
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Hervé Maurin
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sofie Voet
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Isabelle Scheyltjens
- Brain and Systems Immunology Lab, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Benjamin Pavie
- VIB Center for Inflammation Research, Ghent, Belgium
- VIB Bioimaging Core, Ghent, Belgium
| | - Saskia Lippens
- VIB Center for Inflammation Research, Ghent, Belgium
- VIB Bioimaging Core, Ghent, Belgium
| | - Marius Schwabenland
- Institute of Neuropathology Medical Center, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology Medical Center, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama, Japan
| | - Astrid Bottelbergs
- Neuroscience Therapeutic Area, Janssen Research and Development, Beerse, Belgium
| | - Kiavash Movahedi
- Brain and Systems Immunology Lab, Brussels Center for Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mohamed Lamkanfi
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
13
|
Johann L, Soldati S, Müller K, Lampe J, Marini F, Klein M, Schramm E, Ries N, Schelmbauer C, Palagi I, Karram K, Assmann JC, Khan MA, Wenzel J, Schmidt MH, Körbelin J, Schlüter D, van Loo G, Bopp T, Engelhardt B, Schwaninger M, Waisman A. A20 regulates lymphocyte adhesion in murine neuroinflammation by restricting endothelial ICOSL expression in the CNS. J Clin Invest 2023; 133:e168314. [PMID: 37856217 PMCID: PMC10721159 DOI: 10.1172/jci168314] [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: 12/22/2022] [Accepted: 10/18/2023] [Indexed: 10/21/2023] Open
Abstract
A20 is a ubiquitin-modifying protein that negatively regulates NF-κB signaling. Mutations in A20/TNFAIP3 are associated with a variety of autoimmune diseases, including multiple sclerosis (MS). We found that deletion of A20 in central nervous system (CNS) endothelial cells (ECs) enhances experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. A20ΔCNS-EC mice showed increased numbers of CNS-infiltrating immune cells during neuroinflammation and in the steady state. While the integrity of the blood-brain barrier (BBB) was not impaired, we observed a strong activation of CNS-ECs in these mice, with dramatically increased levels of the adhesion molecules ICAM-1 and VCAM-1. We discovered ICOSL to be expressed by A20-deficient CNS-ECs, which we found to function as adhesion molecules. Silencing of ICOSL in CNS microvascular ECs partly reversed the phenotype of A20ΔCNS-EC mice without reaching statistical significance and delayed the onset of EAE symptoms in WT mice. In addition, blocking of ICOSL on primary mouse brain microvascular ECs impaired the adhesion of T cells in vitro. Taken together, we propose that CNS EC-ICOSL contributes to the firm adhesion of T cells to the BBB, promoting their entry into the CNS and eventually driving neuroinflammation.
Collapse
Affiliation(s)
- Lisa Johann
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Kristin Müller
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Josephine Lampe
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany
| | - Federico Marini
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI)
- Research Center for Immunotherapy (FZI), and
| | - Matthias Klein
- Institute for Immunology, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Eva Schramm
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Nathalie Ries
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Carsten Schelmbauer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Ilaria Palagi
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | - Julian C. Assmann
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Mahtab A. Khan
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Jan Wenzel
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany
| | - Mirko H.H. Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Jakob Körbelin
- University Medical Center Hamburg-Eppendorf, Department of Oncology, Hematology and Bone Marrow Transplantation, Hamburg, Germany
| | - Dirk Schlüter
- Hannover Medical School, Institute of Medical Microbiology and Hospital Epidemiology, Hannover, Germany
| | - Geert van Loo
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Tobias Bopp
- Research Center for Immunotherapy (FZI), and
- Institute for Immunology, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
| | | | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
- DZHK (German Research Centre for Cardiovascular Research), Hamburg-Lübeck-Kiel, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg, University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), and
| |
Collapse
|
14
|
Van Damme KFA, Hertens P, Martens A, Gilis E, Priem D, Bruggeman I, Fossoul A, Declercq J, Aegerter H, Wullaert A, Hochepied T, Hoste E, Vande Walle L, Lamkanfi M, Savvides SN, Elewaut D, Lambrecht BN, van Loo G. Protein citrullination and NET formation do not contribute to the pathology of A20/TNFAIP3 mutant mice. Sci Rep 2023; 13:17992. [PMID: 37865713 PMCID: PMC10590390 DOI: 10.1038/s41598-023-45324-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023] Open
Abstract
A20 serves as a critical brake on NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been linked to various inflammatory disorders, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Experimental gene knockout studies in mice have confirmed A20 as a susceptibility gene for SLE and RA. Here, we examine the significance of protein citrullination and NET formation in the autoimmune pathology of A20 mutant mice because autoimmunity directed against citrullinated antigens released by neutrophil extracellular traps (NETs) is central to the pathogenesis of RA and SLE. Furthermore, genetic variants impairing the deubiquitinase (DUB) function of A20 have been shown to contribute to autoimmune susceptibility. Our findings demonstrate that genetic disruption of A20 DUB function in A20 C103R knockin mice does not result in autoimmune pathology. Moreover, we show that PAD4 deficiency, which abolishes protein citrullination and NET formation, does not prevent the development of autoimmunity in A20 deficient mice. Collectively, these findings provide experimental confirmation that PAD4-dependent protein citrullination and NET formation do not serve as pathogenic mechanisms in the development of RA and SLE pathology in mice with A20 mutations.
Collapse
Affiliation(s)
- Karel F A Van Damme
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, 9052, Ghent, Belgium
| | - Pieter Hertens
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Arne Martens
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Elisabeth Gilis
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Dario Priem
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Inge Bruggeman
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Amelie Fossoul
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Jozefien Declercq
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, 9052, Ghent, Belgium
| | - Helena Aegerter
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, 9052, Ghent, Belgium
| | - Andy Wullaert
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Laboratory of Proteinscience, Proteomics and Epigenetic Signalling (PPES), Department of Biomedical Sciences, University of Antwerp, 2610, Antwerp, Belgium
| | - Tino Hochepied
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Esther Hoste
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | | | - Mohamed Lamkanfi
- Department of Internal Medicine and Pediatrics, Ghent University, 9052, Ghent, Belgium
| | - Savvas N Savvides
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, 9052, Ghent, Belgium
| | - Dirk Elewaut
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Rheumatology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Bart N Lambrecht
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, 9052, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Geert van Loo
- VIB, Center for Inflammation Research, Technologiepark 71, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
15
|
Thiran A, Petta I, Blancke G, Thorp M, Planckaert G, Jans M, Andries V, Barbry K, Gilis E, Coudenys J, Hochepied T, Vanhove C, Gracey E, Dumas E, Manuelo T, Josipovic I, van Loo G, Elewaut D, Vereecke L. Sterile triggers drive joint inflammation in TNF- and IL-1β-dependent mouse arthritis models. EMBO Mol Med 2023; 15:e17691. [PMID: 37694693 PMCID: PMC10565626 DOI: 10.15252/emmm.202317691] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023] Open
Abstract
Arthritis is the most common extra-intestinal complication in inflammatory bowel disease (IBD). Conversely, arthritis patients are at risk for developing IBD and often display subclinical gut inflammation. These observations suggest a shared disease etiology, commonly termed "the gut-joint-axis." The clinical association between gut and joint inflammation is further supported by the success of common therapeutic strategies and microbiota dysbiosis in both conditions. Most data, however, support a correlative relationship between gut and joint inflammation, while causative evidence is lacking. Using two independent transgenic mouse arthritis models, either TNF- or IL-1β dependent, we demonstrate that arthritis develops independently of the microbiota and intestinal inflammation, since both lines develop full-blown articular inflammation under germ-free conditions. In contrast, TNF-driven gut inflammation is fully rescued in germ-free conditions, indicating that the microbiota is driving TNF-induced gut inflammation. Together, our study demonstrates that although common inflammatory pathways may drive both gut and joint inflammation, the molecular triggers initiating such pathways are distinct in these tissues.
Collapse
|
16
|
Verboom L, Anderson CJ, Jans M, Petta I, Blancke G, Martens A, Sze M, Hochepied T, Ravichandran KS, Vereecke L, van Loo G. OTULIN protects the intestinal epithelium from apoptosis during inflammation and infection. Cell Death Dis 2023; 14:534. [PMID: 37598207 PMCID: PMC10439912 DOI: 10.1038/s41419-023-06058-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023]
Abstract
The intestinal epithelium is a single cell layer that is constantly renewed and acts as a physical barrier that separates intestinal microbiota from underlying tissues. In inflammatory bowel disease (IBD) in humans, as well as in experimental mouse models of IBD, this barrier is impaired, causing microbial infiltration and inflammation. Deficiency in OTU deubiquitinase with linear linkage specificity (OTULIN) causes OTULIN-related autoinflammatory syndrome (ORAS), a severe inflammatory pathology affecting multiple organs including the intestine. We show that mice with intestinal epithelial cell (IEC)-specific OTULIN deficiency exhibit increased susceptibility to experimental colitis and are highly sensitive to TNF toxicity, due to excessive apoptosis of OTULIN deficient IECs. OTULIN deficiency also increases intestinal pathology in mice genetically engineered to secrete excess TNF, confirming that chronic exposure to TNF promotes epithelial cell death and inflammation in OTULIN deficient mice. Mechanistically we demonstrate that upon TNF stimulation, OTULIN deficiency impairs TNF receptor complex I formation and LUBAC recruitment, and promotes the formation of the cytosolic complex II inducing epithelial cell death. Finally, we show that OTULIN deficiency in IECs increases susceptibility to Salmonella infection, further confirming the importance of OTULIN for intestinal barrier integrity. Together, these results identify OTULIN as a major anti-apoptotic protein in the intestinal epithelium and provide mechanistic insights into how OTULIN deficiency drives gastrointestinal inflammation in ORAS patients.
Collapse
Affiliation(s)
- Lien Verboom
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Christopher J Anderson
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Maude Jans
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Ioanna Petta
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Gillian Blancke
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Arne Martens
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Mozes Sze
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Tino Hochepied
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Kodi S Ravichandran
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
17
|
Carman LE, Samulevich ML, Aneskievich BJ. Repressive Control of Keratinocyte Cytoplasmic Inflammatory Signaling. Int J Mol Sci 2023; 24:11943. [PMID: 37569318 PMCID: PMC10419196 DOI: 10.3390/ijms241511943] [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: 06/23/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
The overactivity of keratinocyte cytoplasmic signaling contributes to several cutaneous inflammatory and immune pathologies. An important emerging complement to proteins responsible for this overactivity is signal repression brought about by several proteins and protein complexes with the native role of limiting inflammation. The signaling repression by these proteins distinguishes them from transmembrane receptors, kinases, and inflammasomes, which drive inflammation. For these proteins, defects or deficiencies, whether naturally arising or in experimentally engineered skin inflammation models, have clearly linked them to maintaining keratinocytes in a non-activated state or returning cells to a post-inflamed state after a signaling event. Thus, together, these proteins help to resolve acute inflammatory responses or limit the development of chronic cutaneous inflammatory disease. We present here an integrated set of demonstrated or potentially inflammation-repressive proteins or protein complexes (linear ubiquitin chain assembly complex [LUBAC], cylindromatosis lysine 63 deubiquitinase [CYLD], tumor necrosis factor alpha-induced protein 3-interacting protein 1 [TNIP1], A20, and OTULIN) for a comprehensive view of cytoplasmic signaling highlighting protein players repressing inflammation as the needed counterpoints to signal activators and amplifiers. Ebb and flow of players on both sides of this inflammation equation would be of physiological advantage to allow acute response to damage or pathogens and yet guard against chronic inflammatory disease. Further investigation of the players responsible for repressing cytoplasmic signaling would be foundational to developing new chemical-entity pharmacologics to stabilize or enhance their function when clinical intervention is needed to restore balance.
Collapse
Affiliation(s)
- Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (L.E.C.); (M.L.S.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
18
|
Chen C, Lan B, Xie G, Liu Z. Analysis and identification of ferroptosis-related genes in ulcerative colitis. Scand J Gastroenterol 2023; 58:1422-1433. [PMID: 37530128 DOI: 10.1080/00365521.2023.2240927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/21/2023] [Indexed: 08/03/2023]
Abstract
BACKGROUND Previous studies have shown that ferroptosis is associated with the pathogenesis of ulcerative colitis (UC). Therefore, this study aimed to identify key ferroptosis-related genes (FRGs) associated with the diagnosis of UC. METHODS UC-related expression datasets were downloaded from the Gene Expression Omnibus (GEO) database. First, Weighted Gene Co-expression Network Analysis (WGCNA) was used to identify UC-related genes (UCRGs). Differentially expressed genes (DEGs) between normal and UC groups were screened in GSE87466, and DEGs were subjected to an intersection analysis with FRGs and UCRGs to obtain ferroptosis-related DEGs (FR DEGs). Then a protein-protein interaction (PPI) network was constructed for FR DEGs. The hub genes were extracted based on the degree, Maximum Neighborhood Component (MNC), closeness, and Maximal Clique Centrality (MCC). Biomarkers with diagnostic values were screened by support vector machine (SVM) and the least absolute shrinkage and selection operator (LASSO) algorithms. Next, the infiltration of immune cells was compared between UC and normal groups, and the correlation between different immune cells and diagnostic genes was analyzed. The biological functions, classical pathways, and intermolecular interaction networks of diagnostic genes were characterized utilizing ingenuity pathway analysis (IPA). Finally, a TF-mRNA network was constructed and potential small-molecule compounds were screened. RESULTS Thirty-six FR DEGs were obtained, and these were enriched in biological processes such as positive regulation of cytokine production, cytokine-mediated signalling pathway, long-chain fatty acid-CoA ligase activity, etc. Among 18 hub genes, five genes (ALOX5, TIMP1, TNFAIP3, SOCS1, DUOX2) were captured with diagnostic values for UC, and they displayed significant differences between UC and normal groups. Sixteen immune cell infiltrates were significantly different between UC and normal groups, such as activated dendritic cells and resting dendritic cells. TNFAIP3 and ALOX5 were positively correlated with neutrophils, and TIMP1, SOCS1, ALOX5, and DUOX2 were negatively correlated with M2 macrophages. IPA showed that diagnostic genes were related to 43 function modules and activated 17 pathways. The constructed TF-mRNA regulatory network comprised three diagnostic genes and 17 differentially expressed TFs. Potential small-molecule compounds including helveticoside and cymarin were identified. CONCLUSION Our findings yielded several promising FRGs for UC, providing a scientific reference for further studies on the pathogenesis of UC.
Collapse
Affiliation(s)
- Chen Chen
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, P.R. China
| | - Bo Lan
- Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, P.R. China
| | - Guanghong Xie
- Department of Emergency Internal Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, P.R. China
| | - Zhaoyang Liu
- Department of Emergency Internal Medicine, Guizhou Provincial People's Hospital, Guiyang, Guizhou Province, P.R. China
| |
Collapse
|
19
|
Uwada J, Nakazawa H, Muramatsu I, Masuoka T, Yazawa T. Role of Muscarinic Acetylcholine Receptors in Intestinal Epithelial Homeostasis: Insights for the Treatment of Inflammatory Bowel Disease. Int J Mol Sci 2023; 24:ijms24076508. [PMID: 37047478 PMCID: PMC10095461 DOI: 10.3390/ijms24076508] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
Inflammatory bowel disease (IBD), which includes Crohn’s disease and ulcerative colitis, is an intestinal disorder that causes prolonged inflammation of the gastrointestinal tract. Currently, the etiology of IBD is not fully understood and treatments are insufficient to completely cure the disease. In addition to absorbing essential nutrients, intestinal epithelial cells prevent the entry of foreign antigens (micro-organisms and undigested food) through mucus secretion and epithelial barrier formation. Disruption of the intestinal epithelial homeostasis exacerbates inflammation. Thus, the maintenance and reinforcement of epithelial function may have therapeutic benefits in the treatment of IBD. Muscarinic acetylcholine receptors (mAChRs) are G protein-coupled receptors for acetylcholine that are expressed in intestinal epithelial cells. Recent studies have revealed the role of mAChRs in the maintenance of intestinal epithelial homeostasis. The importance of non-neuronal acetylcholine in mAChR activation in epithelial cells has also been recognized. This review aimed to summarize recent advances in research on mAChRs for intestinal epithelial homeostasis and the involvement of non-neuronal acetylcholine systems, and highlight their potential as targets for IBD therapy.
Collapse
|
20
|
Baillo A, Villena J, Albarracín L, Tomokiyo M, Elean M, Fukuyama K, Quilodrán-Vega S, Fadda S, Kitazawa H. Lactiplantibacillus plantarum Strains Modulate Intestinal Innate Immune Response and Increase Resistance to Enterotoxigenic Escherichia coli Infection. Microorganisms 2022; 11:microorganisms11010063. [PMID: 36677354 PMCID: PMC9863675 DOI: 10.3390/microorganisms11010063] [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: 11/30/2022] [Revised: 12/19/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Currently, probiotic bacteria with not transferable antibiotic resistance represent a sustainable strategy for the treatment and prevention of enterotoxigenic Escherichia coli (ETEC) in farm animals. Lactiplantibacillus plantarum is among the most versatile species used in the food industry, either as starter cultures or probiotics. In the present work, the immunobiotic potential of L. plantarum CRL681 and CRL1506 was studied to evaluate their capability to improve the resistance to ETEC infection. In vitro studies using porcine intestinal epithelial (PIE) cells and in vivo experiments in mice were undertaken. Expression analysis indicated that both strains were able to trigger IL-6 and IL-8 expression in PIE cells in steady-state conditions. Furthermore, mice orally treated with these strains had significantly improved levels of IFN-γ and TNF-α in the intestine as well as enhanced activity of peritoneal macrophages. The ability of CRL681 and CRL1506 to beneficially modulate intestinal immunity was further evidenced in ETEC-challenge experiments. In vitro, the CRL1506 and CRL681 strains modulated the expression of inflammatory cytokines (IL-6) and chemokines (IL-8, CCL2, CXCL5 and CXCL9) in ETEC-stimulated PIE cells. In vivo experiments demonstrated the ability of both strains to beneficially regulate the immune response against this pathogen. Moreover, the oral treatment of mice with lactic acid bacteria (LAB) strains significantly reduced ETEC counts in jejunum and ileum and prevented the spread of the pathogen to the spleen and liver. Additionally, LAB treated-mice had improved levels of intestinal IL-10 both at steady state and after the challenge with ETEC. The protective effect against ETEC infection was not observed for the non-immunomodulatory TL2677 strain. Furthermore, the study showed that L. plantarum CRL1506 was more efficient than the CRL681 strain to modulate mucosal immunity highlighting the strain specific character of this probiotic activity. Our results suggest that the improved intestinal epithelial defenses and innate immunity induced by L. plantarum CRL1506 and CRL681 would increase the clearance of ETEC and at the same time, protect the host against detrimental inflammation. These constitute valuable features for future probiotic products able to improve the resistance to ETEC infection.
Collapse
Affiliation(s)
- Ayelen Baillo
- Laboratory of Technology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina
| | - Julio Villena
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Correspondence: (J.V.); (S.F.); (H.K.)
| | - Leonardo Albarracín
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina
| | - Mikado Tomokiyo
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Mariano Elean
- Laboratory of Immunobiotechnology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina
| | - Kohtaro Fukuyama
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
| | - Sandra Quilodrán-Vega
- Laboratory of Food Microbiology, Faculty of Veterinary Sciences, University of Concepción, Chillán 3820572, Chile
| | - Silvina Fadda
- Laboratory of Technology, Reference Centre for Lactobacilli (CERELA-CONICET), Tucuman CP4000, Argentina
- Correspondence: (J.V.); (S.F.); (H.K.)
| | - Haruki Kitazawa
- Food and Feed Immunology Group, Laboratory of Animal Food Function, Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Livestock Immunology Unit, International Education and Research Center for Food and Agricultural Immunology (CFAI), Graduate School of Agricultural Science, Tohoku University, Sendai 981-8555, Japan
- Correspondence: (J.V.); (S.F.); (H.K.)
| |
Collapse
|
21
|
Martens A, Hertens P, Priem D, Rinotas V, Meletakos T, Gennadi M, Van Hove L, Louagie E, Coudenys J, De Muynck A, Gaublomme D, Sze M, van Hengel J, Catrysse L, Hoste E, Zajac JD, Davey RA, Van Hoorebeke L, Hochepied T, Bertrand MJM, Armaka M, Elewaut D, van Loo G. A20 controls RANK-dependent osteoclast formation and bone physiology. EMBO Rep 2022; 23:e55233. [PMID: 36194667 PMCID: PMC9724664 DOI: 10.15252/embr.202255233] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/05/2022] Open
Abstract
The anti-inflammatory protein A20 serves as a critical brake on NF-κB signaling and NF-κB-dependent inflammation. In humans, polymorphisms in or near the TNFAIP3/A20 gene have been associated with several inflammatory disorders, including rheumatoid arthritis (RA), and experimental studies in mice have demonstrated that myeloid-specific A20 deficiency causes the development of a severe polyarthritis resembling human RA. Myeloid A20 deficiency also promotes osteoclastogenesis in mice, suggesting a role for A20 in the regulation of osteoclast differentiation and bone formation. We show here that osteoclast-specific A20 knockout mice develop severe osteoporosis, but not inflammatory arthritis. In vitro, osteoclast precursor cells from A20 deficient mice are hyper-responsive to RANKL-induced osteoclastogenesis. Mechanistically, we show that A20 is recruited to the RANK receptor complex within minutes of ligand binding, where it restrains NF-κB activation independently of its deubiquitinating activity but through its zinc finger (ZnF) 4 and 7 ubiquitin-binding functions. Together, these data demonstrate that A20 acts as a regulator of RANK-induced NF-κB signaling to control osteoclast differentiation, assuring proper bone development and turnover.
Collapse
Affiliation(s)
- Arne Martens
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Pieter Hertens
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Dario Priem
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Vagelis Rinotas
- Biomedical Sciences Research Center 'Alexander Fleming'VariGreece
| | | | - Meropi Gennadi
- Biomedical Sciences Research Center 'Alexander Fleming'VariGreece
| | - Lisette Van Hove
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Els Louagie
- Center for Inflammation Research VIBGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
| | - Julie Coudenys
- Center for Inflammation Research VIBGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
| | | | - Djoere Gaublomme
- Center for Inflammation Research VIBGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
| | - Mozes Sze
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | | | - Leen Catrysse
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Esther Hoste
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Jeffrey D Zajac
- Department of Medicine, Austin HealthUniversity of MelbourneHeidelbergVictoriaAustralia
| | - Rachel A Davey
- Department of Medicine, Austin HealthUniversity of MelbourneHeidelbergVictoriaAustralia
| | | | - Tino Hochepied
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Mathieu J M Bertrand
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| | - Marietta Armaka
- Biomedical Sciences Research Center 'Alexander Fleming'VariGreece
| | - Dirk Elewaut
- Center for Inflammation Research VIBGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
| | - Geert van Loo
- Center for Inflammation Research VIBGhentBelgium
- Department of Biomedical Molecular BiologyGhent UniversityGhentBelgium
| |
Collapse
|
22
|
Abstract
Tumour necrosis factor (TNF) is a central cytokine in inflammatory reactions, and biologics that neutralize TNF are among the most successful drugs for the treatment of chronic inflammatory and autoimmune pathologies. In recent years, it became clear that TNF drives inflammatory responses not only directly by inducing inflammatory gene expression but also indirectly by inducing cell death, instigating inflammatory immune reactions and disease development. Hence, inhibitors of cell death are being considered as a new therapy for TNF-dependent inflammatory diseases.
Collapse
|
23
|
Deubiquitinases in cell death and inflammation. Biochem J 2022; 479:1103-1119. [PMID: 35608338 PMCID: PMC9162465 DOI: 10.1042/bcj20210735] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/07/2022] [Accepted: 05/10/2022] [Indexed: 11/20/2022]
Abstract
Apoptosis, pyroptosis, and necroptosis are distinct forms of programmed cell death that eliminate infected, damaged, or obsolete cells. Many proteins that regulate or are a part of the cell death machinery undergo ubiquitination, a post-translational modification made by ubiquitin ligases that modulates protein abundance, localization, and/or activity. For example, some ubiquitin chains target proteins for degradation, while others function as scaffolds for the assembly of signaling complexes. Deubiquitinases (DUBs) are the proteases that counteract ubiquitin ligases by cleaving ubiquitin from their protein substrates. Here, we review the DUBs that have been found to suppress or promote apoptosis, pyroptosis, or necroptosis.
Collapse
|
24
|
The Multi-Omics Analysis Revealed a Metabolic Regulatory System of Cecum in Rabbit with Diarrhea. Animals (Basel) 2022; 12:ani12091194. [PMID: 35565618 PMCID: PMC9099945 DOI: 10.3390/ani12091194] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/05/2023] Open
Abstract
With the comprehensive prohibition of antibiotics in the feed industry in China, the incidence of diarrhea in rabbits increased, such as loss of appetite, vomiting, and excretion of atheromatous feces. In order to explore the pathological and the molecular mechanisms of the diarrhea in the rabbitry fed with antibiotic-free diet, we used microbial metagenomics, transcriptome, and non-targeted metabolomics sequencing. The results showed that the Firmicutes level was significantly decreased (p < 0.001) and the Proteobacteria level was significantly increased (p < 0.05). The functional enrichment of cecum revealed that most differentially expressed genes (DEGs) were expressed in immune, inflammatory, and metabolic processes. The enrichment of the cecal fecal metabolites focused on the bile secretion, antifolate resistance, and tryptophan metabolism pathways, which are mainly associated with inflammation. The results of correlation analysis showed that Fournierella was positively correlated with myricetin, ursolic acid, and furtherly might cause bile secretion and tryptophan metabolism disorder, aggravate intestinal inflammation, change intestinal permeability, and reduce host immunity, leading to diarrhea in rabbits. This study provides a theoretical basis for illustrating the reason for diarrhea and developing new feeds for the health of rabbits.
Collapse
|
25
|
Perez-Sanchez C, Barbera Betancourt A, Lyons PA, Zhang Z, Suo C, Lee JC, McKinney EF, Modis LK, Ellson C, Smith KG. miR-374a-5p regulates inflammatory genes and monocyte function in patients with inflammatory bowel disease. J Exp Med 2022; 219:e20211366. [PMID: 35363256 PMCID: PMC8980842 DOI: 10.1084/jem.20211366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/23/2021] [Accepted: 02/17/2022] [Indexed: 02/02/2023] Open
Abstract
MicroRNAs are critical regulators of gene expression controlling cellular processes including inflammation. We explored their role in the pathogenesis of inflammatory bowel disease (IBD) and identified reduced expression of miR-374a-5p in IBD monocytes that correlated with a module of up-regulated genes related to the inflammatory response. Key proinflammatory module genes, including for example TNFα, IL1A, IL6, and OSM, were inversely correlated with miR-374a-5p and were validated in vitro. In colonic biopsies, miR-374a-5p was again reduced in expression and inversely correlated with the same inflammatory module, and its levels predicted subsequent response to anti-TNF therapy. Increased miR-374a-5p expression was shown to control macrophage-driven inflammation by suppressing proinflammatory mediators and to reduce the capacity of monocytes to migrate and activate T cells. Our findings suggest that miR-374a-5p reduction is a central driver of inflammation in IBD, and its therapeutic supplementation could reduce monocyte-driven inflammation in IBD or other immune-mediated diseases.
Collapse
Affiliation(s)
- Carlos Perez-Sanchez
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Rheumatology Service, Reina Sofia University Hospital, Maimonides Biomedical Research Institute of Córdoba, University of Cordoba, Cordoba, Spain
| | - Ariana Barbera Betancourt
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Zinan Zhang
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology and Clinical Genomics Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Chenqu Suo
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Paediatrics, Cambridge University Hospitals, Cambridge, UK
| | - James C. Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | - Eoin F. McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| | | | | | - Kenneth G.C. Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
26
|
Ruan J, Schlüter D, Naumann M, Waisman A, Wang X. Ubiquitin-modifying enzymes as regulators of colitis. Trends Mol Med 2022; 28:304-318. [PMID: 35177326 DOI: 10.1016/j.molmed.2022.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/24/2022] [Accepted: 01/24/2022] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), is a chronic inflammatory disorder of the gastrointestinal tract. Although the pathophysiology of IBD is multifaceted, ubiquitination, a post-translational modification, has been shown to have essential roles in its pathogenesis and development. Ubiquitin-modifying enzymes (UMEs) work in synergy to orchestrate the optimal ubiquitination of target proteins, thereby maintaining intestinal homeostasis. Genome-wide association studies (GWAS) have identified multiple UME genes as IBD susceptibility loci, implying the importance of UMEs in IBD. Furthermore, accumulative evidence demonstrates that UMEs affect intestinal inflammation by regulating various aspects, such as intestinal barrier functions and immune responses. Considering the significant functions of UMEs in IBD, targeting UMEs could become a favorable therapeutic approach for IBD.
Collapse
Affiliation(s)
- Jing Ruan
- Department of Pathology, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dirk Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Michael Naumann
- Institute of Experimental Internal Medicine, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Xu Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China; Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, China; Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
27
|
Rusu I, Mennillo E, Bain JL, Li Z, Sun X, Ly KM, Rosli YY, Naser M, Wang Z, Advincula R, Achacoso P, Shao L, Razani B, Klein OD, Marson A, Turnbaugh JA, Turnbaugh PJ, Malynn BA, Ma A, Kattah MG. Microbial signals, MyD88, and lymphotoxin drive TNF-independent intestinal epithelial tissue damage. J Clin Invest 2022; 132:154993. [PMID: 35077396 PMCID: PMC8884902 DOI: 10.1172/jci154993] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/19/2022] [Indexed: 11/18/2022] Open
Abstract
Anti-TNF antibodies are effective for treating patients with inflammatory bowel disease (IBD), but many patients fail to respond to anti-TNF therapy, highlighting the importance of TNF-independent disease. We previously demonstrated that acute deletion of 2 IBD susceptibility genes, A20 (Tnfaip3) and Abin-1 (Tnip1), in intestinal epithelial cells (IECs) sensitized mice to both TNF-dependent and TNF-independent death. Here we show that TNF-independent IEC death after A20 and Abin-1 deletion was rescued by germ-free derivation or deletion of MyD88, while deletion of Trif provided only partial protection. Combined deletion of Ripk3 and Casp8, which inhibits both apoptotic and necroptotic death, completely protected against death after acute deletion of A20 and Abin-1 in IECs. A20- and Abin-1–deficient IECs were sensitized to TNF-independent, TNFR1-mediated death in response to lymphotoxin α (LTα) homotrimers. Blockade of LTα in vivo reduced weight loss and improved survival when combined with partial deletion of MyD88. Biopsies of inflamed colon mucosa from patients with IBD exhibited increased LTA and IL1B expression, including a subset of patients with active colitis on anti-TNF therapy. These data show that microbial signals, MyD88, and LTα all contribute to TNF-independent intestinal injury.
Collapse
Affiliation(s)
- Iulia Rusu
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Elvira Mennillo
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Jared L. Bain
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Zhongmei Li
- Department of Medicine, UCSF, San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
| | - Xiaofei Sun
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Yenny Y. Rosli
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Mohammad Naser
- Biological Imaging Development CoLab, UCSF, San Francisco, California, USA
| | - Zunqiu Wang
- Department of Medicine, UCSF, San Francisco, California, USA
| | | | - Philip Achacoso
- Department of Medicine, UCSF, San Francisco, California, USA
| | - Ling Shao
- Department of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Ophir D. Klein
- Departments of Orofacial Sciences and Pediatrics, Program in Craniofacial Biology, and
| | - Alexander Marson
- Department of Medicine, UCSF, San Francisco, California, USA
- Gladstone Institutes, San Francisco, California, USA
- Department of Microbiology and Immunology and
- Institute for Human Genetics, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | | | | | | - Averil Ma
- Department of Medicine, UCSF, San Francisco, California, USA
| | | |
Collapse
|
28
|
Zhang C, Han X, Sun L, Yang S, Peng J, Chen Y, Jin Y, Xu F, Liu Z, Zhou Q. OUP accepted manuscript. Clin Kidney J 2022; 15:2027-2038. [PMID: 36325013 PMCID: PMC9613433 DOI: 10.1093/ckj/sfac130] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background Heterozygous loss-of-function mutations in the tumour necrosis factor alpha induced protein 3 (TNFAIP3) gene cause an early-onset auto-inflammatory disease named haploinsufficiency of A20 (HA20). Here we describe three unrelated patients with autoimmune lupus nephritis (LN) phenotypes carrying three novel mutations in the TNFAIP3 gene. Methods Whole-exome sequencing (WES) was used to identify the causative mutations in three biopsy-proven LN patients. Sanger sequencing and quantitative polymerase chain reaction (qPCR) were used to validate the mutations identified by WES. RNA sequencing, qPCR and cytometric bead array was used to detect inflammatory signatures in the patients. Results The patients predominantly presented with an autoimmune phenotype, including autoimmune haemolytic anaemia, multipositive autoantibodies and LN. Additionally, novel phenotypes of allergy and pericardial effusion were first reported. WES identified three novel heterozygous mutations in the TNFAIP3 gene, including a novel splicing mutation located in the canonical splicing site (c.634+2T>C) resulting in an intron 4 insertion containing a premature stop codon, a de novo novel copy number variation (exon 7–8 deletion) and a novel nonsense mutation c.1300_1301delinsTA causing a premature stop codon. We further identified hyperactivation signatures of nuclear factor- kappa B and type I IFN signalling and overproduction of pro-inflammatory cytokines in the blood. This report expanded the phenotype to a later age, as two girls were diagnosed at age 3 years and one man at age 29 years. Conclusions Kidney involvement may be the main feature of the clinical spectrum of HA20, even in adults. Genetic screening should be considered for early-onset LN patients.
Collapse
Affiliation(s)
| | | | - Li Sun
- Department of Rheumatology, Children's Hospital of Fudan University, Shanghai, China
| | - Sirui Yang
- Department of Pediatric Rheumatology and Allergy, First Hospital, Jilin University, Changchun, China
| | - Jiahui Peng
- Kidney Diseases Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Yinghua Chen
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing, China
| | - Ying Jin
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing, China
| | - Feng Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing, China
| | | | - Qing Zhou
- Correspondence to: Qing Zhou; E-mail:
| |
Collapse
|
29
|
Zou M, Zeng QS, Nie J, Yang JH, Luo ZY, Gan HT. The Role of E3 Ubiquitin Ligases and Deubiquitinases in Inflammatory Bowel Disease: Friend or Foe? Front Immunol 2021; 12:769167. [PMID: 34956195 PMCID: PMC8692584 DOI: 10.3389/fimmu.2021.769167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/17/2021] [Indexed: 02/05/2023] Open
Abstract
Inflammatory bowel disease (IBD), which include Crohn’s disease (CD) and ulcerative colitis (UC), exhibits a complex multifactorial pathogenesis involving genetic susceptibility, imbalance of gut microbiota, mucosal immune disorder and environmental factors. Recent studies reported associations between ubiquitination and deubiquitination and the occurrence and development of inflammatory bowel disease. Ubiquitination modification, one of the most important types of post-translational modifications, is a multi-step enzymatic process involved in the regulation of various physiological processes of cells, including cell cycle progression, cell differentiation, apoptosis, and innate and adaptive immune responses. Alterations in ubiquitination and deubiquitination can lead to various diseases, including IBD. Here, we review the role of E3 ubiquitin ligases and deubiquitinases (DUBs) and their mediated ubiquitination and deubiquitination modifications in the pathogenesis of IBD. We highlight the importance of this type of posttranslational modification in the development of inflammation, and provide guidance for the future development of targeted therapeutics in IBD.
Collapse
Affiliation(s)
- Min Zou
- Department of Gastroenterology and the Center of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China.,Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Qi-Shan Zeng
- Department of Gastroenterology and the Center of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China.,Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Nie
- Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Department of Geriatrics and National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Jia-Hui Yang
- Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Department of Geriatrics and National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen-Yi Luo
- Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Department of Geriatrics and National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| | - Hua-Tian Gan
- Department of Gastroenterology and the Center of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China.,Lab of Inflammatory Bowel Disease, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China.,Department of Geriatrics and National Clinical Research Center for Geriatric, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Hausmann A, Felmy B, Kunz L, Kroon S, Berthold DL, Ganz G, Sandu I, Nakamura T, Zangger NS, Zhang Y, Dolowschiak T, Fattinger SA, Furter M, Müller-Hauser AA, Barthel M, Vlantis K, Wachsmuth L, Kisielow J, Tortola L, Heide D, Heikenwälder M, Oxenius A, Kopf M, Schroeder T, Pasparakis M, Sellin ME, Hardt WD. Intercrypt sentinel macrophages tune antibacterial NF-κB responses in gut epithelial cells via TNF. J Exp Med 2021; 218:e20210862. [PMID: 34529751 PMCID: PMC8480669 DOI: 10.1084/jem.20210862] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/21/2021] [Accepted: 08/25/2021] [Indexed: 12/14/2022] Open
Abstract
Intestinal epithelial cell (IEC) NF-κB signaling regulates the balance between mucosal homeostasis and inflammation. It is not fully understood which signals tune this balance and how bacterial exposure elicits the process. Pure LPS induces epithelial NF-κB activation in vivo. However, we found that in mice, IECs do not respond directly to LPS. Instead, tissue-resident lamina propria intercrypt macrophages sense LPS via TLR4 and rapidly secrete TNF to elicit epithelial NF-κB signaling in their immediate neighborhood. This response pattern is relevant also during oral enteropathogen infection. The macrophage-TNF-IEC axis avoids responses to luminal microbiota LPS but enables crypt- or tissue-scale epithelial NF-κB responses in proportion to the microbial threat. Thereby, intercrypt macrophages fulfill important sentinel functions as first responders to Gram-negative microbes breaching the epithelial barrier. The tunability of this crypt response allows the induction of defense mechanisms at an appropriate scale according to the localization and intensity of microbial triggers.
Collapse
Affiliation(s)
- Annika Hausmann
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Boas Felmy
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Leo Kunz
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Sanne Kroon
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Dorothée Lisa Berthold
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Giverny Ganz
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Ioana Sandu
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Toshihiro Nakamura
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Nathan Sébastien Zangger
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Yang Zhang
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Tamas Dolowschiak
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Stefan Alexander Fattinger
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Markus Furter
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Anna Angelika Müller-Hauser
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Manja Barthel
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Katerina Vlantis
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Laurens Wachsmuth
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Jan Kisielow
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Luigi Tortola
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center, Heidelberg, Germany
| | - Annette Oxenius
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zurich, Basel, Switzerland
| | - Manolis Pasparakis
- Institute for Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Mikael Erik Sellin
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Heida A, Gruben N, Catrysse L, Koehorst M, Koster M, Kloosterhuis NJ, Gerding A, Havinga R, Bloks VW, Bongiovanni L, Wolters JC, van Dijk T, van Loo G, de Bruin A, Kuipers F, Koonen DPY, van de Sluis B. The hepatocyte IKK:NF-κB axis promotes liver steatosis by stimulating de novo lipogenesis and cholesterol synthesis. Mol Metab 2021; 54:101349. [PMID: 34626855 PMCID: PMC8581577 DOI: 10.1016/j.molmet.2021.101349] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/20/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Obesity-related chronic inflammation plays an important role in the development of Metabolic Associated Fatty Liver Disease (MAFLD). Although the contribution of the pro-inflammatory NF-κB signaling pathway to the progression from simple steatosis to non-alcoholic steatohepatitis (NASH) is well-established, its role as an initiator of hepatic steatosis and the underlying mechanism remains unclear. Here, we investigated the hypothesis that the hepatocytic NF-κB signaling pathway acts as a metabolic regulator, thereby promoting hepatic steatosis development. METHODS A murine model expressing a constitutively active form of IKKβ in hepatocytes (Hep-IKKβca) was used to activate hepatocyte NF-κB. In addition, IKKβca was also expressed in hepatocyte A20-deficient mice (IKKβca;A20LKO). A20 is an NF-κB-target gene that inhibits the activation of the NF-κB signaling pathway upstream of IKKβ. These mouse models were fed a sucrose-rich diet for 8 weeks. Hepatic lipid levels were measured and using [1-13C]-acetate de novo lipogenesis and cholesterol synthesis rate were determined. Gene expression analyses and immunoblotting were used to study the lipogenesis and cholesterol synthesis pathways. RESULTS Hepatocytic NF-κB activation by expressing IKKβca in hepatocytes resulted in hepatic steatosis without inflammation. Ablation of hepatocyte A20 in Hep-IKKβca mice (IKKβca;A20LKO mice) exacerbated hepatic steatosis, characterized by macrovesicular accumulation of triglycerides and cholesterol, and increased plasma cholesterol levels. Both De novo lipogenesis (DNL) and cholesterol synthesis were found elevated in IKKβca;A20LKO mice. Phosphorylation of AMP-activated kinase (AMPK) - a suppressor in lipogenesis and cholesterol synthesis - was decreased in IKKβca;A20LKO mice. This was paralleled by elevated protein levels of hydroxymethylglutaryl-CoA synthase 1 (HMGCS1) and reduced phosphorylation of HMG-CoA reductase (HMGCR) both key enzymes in the cholesterol synthesis pathway. Whereas inflammation was not observed in young IKKβca;A20LKO mice sustained hepatic NF-κB activation resulted in liver inflammation, together with elevated hepatic and plasma cholesterol levels in middle-aged mice. CONCLUSIONS The hepatocytic IKK:NF-κB axis is a metabolic regulator by controlling DNL and cholesterol synthesis, independent of its central role in inflammation. The IKK:NF-κB axis controls the phosphorylation levels of AMPK and HMGCR and the protein levels of HMGCS1. Chronic IKK-mediated NF-κB activation may contribute to the initiation of hepatic steatosis and cardiovascular disease risk in MAFLD patients.
Collapse
Affiliation(s)
- Andries Heida
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nanda Gruben
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leen Catrysse
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Martijn Koehorst
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Mirjam Koster
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Albert Gerding
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Rick Havinga
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Vincent W Bloks
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Laura Bongiovanni
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy
| | - Justina C Wolters
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Theo van Dijk
- Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Geert van Loo
- VIB Inflammation Research Center, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Alain de Bruin
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, the Netherlands
| | - Folkert Kuipers
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Departments of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Debby P Y Koonen
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Bart van de Sluis
- Departments of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
32
|
Woznicki JA, Saini N, Flood P, Rajaram S, Lee CM, Stamou P, Skowyra A, Bustamante-Garrido M, Regazzoni K, Crawford N, McDade SS, Longley DB, Aza-Blanc P, Shanahan F, Zulquernain SA, McCarthy J, Melgar S, McRae BL, Nally K. TNF-α synergises with IFN-γ to induce caspase-8-JAK1/2-STAT1-dependent death of intestinal epithelial cells. Cell Death Dis 2021; 12:864. [PMID: 34556638 PMCID: PMC8459343 DOI: 10.1038/s41419-021-04151-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 08/16/2021] [Accepted: 09/08/2021] [Indexed: 12/25/2022]
Abstract
Rewiring of host cytokine networks is a key feature of inflammatory bowel diseases (IBD) such as Crohn's disease (CD). Th1-type cytokines-IFN-γ and TNF-α-occupy critical nodes within these networks and both are associated with disruption of gut epithelial barrier function. This may be due to their ability to synergistically trigger the death of intestinal epithelial cells (IECs) via largely unknown mechanisms. In this study, through unbiased kinome RNAi and drug repurposing screens we identified JAK1/2 kinases as the principal and nonredundant drivers of the synergistic killing of human IECs by IFN-γ/TNF-α. Sensitivity to IFN-γ/TNF-α-mediated synergistic IEC death was retained in primary patient-derived intestinal organoids. Dependence on JAK1/2 was confirmed using genetic loss-of-function studies and JAK inhibitors (JAKinibs). Despite the presence of biochemical features consistent with canonical TNFR1-mediated apoptosis and necroptosis, IFN-γ/TNF-α-induced IEC death was independent of RIPK1/3, ZBP1, MLKL or caspase activity. Instead, it involved sustained activation of JAK1/2-STAT1 signalling, which required a nonenzymatic scaffold function of caspase-8 (CASP8). Further modelling in gut mucosal biopsies revealed an intercorrelated induction of the lethal CASP8-JAK1/2-STAT1 module during ex vivo stimulation of T cells. Functional studies in CD-derived organoids using inhibitors of apoptosis, necroptosis and JAKinibs confirmed the causative role of JAK1/2-STAT1 in cytokine-induced death of primary IECs. Collectively, we demonstrate that TNF-α synergises with IFN-γ to kill IECs via the CASP8-JAK1/2-STAT1 module independently of canonical TNFR1 and cell death signalling. This non-canonical cell death pathway may underpin immunopathology driven by IFN-γ/TNF-α in diverse autoinflammatory diseases such as IBD, and its inhibition may contribute to the therapeutic efficacy of anti-TNFs and JAKinibs.
Collapse
Affiliation(s)
| | - Nisha Saini
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Peter Flood
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Ciaran M Lee
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | | | | | | | - Nyree Crawford
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Simon S McDade
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Daniel B Longley
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Pedro Aza-Blanc
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland
| | - Syed A Zulquernain
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland
| | - Jane McCarthy
- Department of Gastroenterology, Mercy University Hospital, Cork, Ireland
| | - Silvia Melgar
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Bradford L McRae
- Immunology Discovery, Abbvie Bioresearch Center, Worcester, MA, 01605, USA
| | - Ken Nally
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
33
|
Catrysse L, Maes B, Mehrotra P, Martens A, Hoste E, Martens L, Maueröder C, Remmerie A, Bujko A, Slowicka K, Sze M, Vikkula H, Ghesquière B, Scott CL, Saeys Y, van de Sluis B, Ravichandran K, Janssens S, van Loo G. A20 deficiency in myeloid cells protects mice from diet-induced obesity and insulin resistance due to increased fatty acid metabolism. Cell Rep 2021; 36:109748. [PMID: 34551300 DOI: 10.1016/j.celrep.2021.109748] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 04/04/2021] [Accepted: 09/02/2021] [Indexed: 10/20/2022] Open
Abstract
Obesity-induced inflammation is a major driving force in the development of insulin resistance, type 2 diabetes (T2D), and related metabolic disorders. During obesity, macrophages accumulate in the visceral adipose tissue, creating a low-grade inflammatory environment. Nuclear factor κB (NF-κB) signaling is a central coordinator of inflammatory responses and is tightly regulated by the anti-inflammatory protein A20. Here, we find that myeloid-specific A20-deficient mice are protected from diet-induced obesity and insulin resistance despite an inflammatory environment in their metabolic tissues. Macrophages lacking A20 show impaired mitochondrial respiratory function and metabolize more palmitate both in vitro and in vivo. We hypothesize that A20-deficient macrophages rely more on palmitate oxidation and metabolize the fat present in the diet, resulting in a lean phenotype and protection from metabolic disease. These findings reveal a role for A20 in regulating macrophage immunometabolism.
Collapse
Affiliation(s)
- Leen Catrysse
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bastiaan Maes
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, B-9052 Ghent, Belgium
| | - Parul Mehrotra
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Arne Martens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Liesbet Martens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Christian Maueröder
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Anneleen Remmerie
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Anna Bujko
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Karolina Slowicka
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Mozes Sze
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Hanna Vikkula
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bart Ghesquière
- Metabolomics Core Facility, VIB Center for Cancer Biology, VIB, B-3000 Leuven, Belgium
| | - Charlotte L Scott
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Yvan Saeys
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, B-9052 Ghent, Belgium
| | - Bart van de Sluis
- Department of Pediatrics, Molecular Genetics Section, University of Groningen, University Medical Center Groningen, NL- 9713 Groningen, the Netherlands
| | - Kodi Ravichandran
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; Center for Cell Clearance and Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Sophie Janssens
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, B-9052 Ghent, Belgium
| | - Geert van Loo
- VIB Center for Inflammation Research, B-9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
34
|
Barnabei L, Laplantine E, Mbongo W, Rieux-Laucat F, Weil R. NF-κB: At the Borders of Autoimmunity and Inflammation. Front Immunol 2021; 12:716469. [PMID: 34434197 PMCID: PMC8381650 DOI: 10.3389/fimmu.2021.716469] [Citation(s) in RCA: 340] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/22/2021] [Indexed: 12/18/2022] Open
Abstract
The transcription factor NF-κB regulates multiple aspects of innate and adaptive immune functions and serves as a pivotal mediator of inflammatory response. In the first part of this review, we discuss the NF-κB inducers, signaling pathways, and regulators involved in immune homeostasis as well as detail the importance of post-translational regulation by ubiquitination in NF-κB function. We also indicate the stages of central and peripheral tolerance where NF-κB plays a fundamental role. With respect to central tolerance, we detail how NF-κB regulates medullary thymic epithelial cell (mTEC) development, homeostasis, and function. Moreover, we elaborate on its role in the migration of double-positive (DP) thymocytes from the thymic cortex to the medulla. With respect to peripheral tolerance, we outline how NF-κB contributes to the inactivation and destruction of autoreactive T and B lymphocytes as well as the differentiation of CD4+-T cell subsets that are implicated in immune tolerance. In the latter half of the review, we describe the contribution of NF-κB to the pathogenesis of autoimmunity and autoinflammation. The recent discovery of mutations involving components of the pathway has both deepened our understanding of autoimmune disease and informed new therapeutic approaches to treat these illnesses.
Collapse
Affiliation(s)
- Laura Barnabei
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Emmanuel Laplantine
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - William Mbongo
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| | - Frédéric Rieux-Laucat
- INSERM UMR 1163, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, Imagine Institute Paris Descartes Sorbonne Paris Cité University, Paris, France
| | - Robert Weil
- Sorbonne Universités, Institut National de la Santé et de la Recherche Médicale (INSERM, UMR1135), Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CMI, Paris, France
| |
Collapse
|
35
|
Microbes exploit death-induced nutrient release by gut epithelial cells. Nature 2021; 596:262-267. [PMID: 34349263 DOI: 10.1038/s41586-021-03785-9] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 06/30/2021] [Indexed: 01/13/2023]
Abstract
Regulated cell death is an integral part of life, and has broad effects on organism development and homeostasis1. Malfunctions within the regulated cell death process, including the clearance of dying cells, can manifest in diverse pathologies throughout various tissues including the gastrointestinal tract2. A long appreciated, yet elusively defined relationship exists between cell death and gastrointestinal pathologies with an underlying microbial component3-6, but the direct effect of dying mammalian cells on bacterial growth is unclear. Here we advance a concept that several Enterobacteriaceae, including patient-derived clinical isolates, have an efficient growth strategy to exploit soluble factors that are released from dying gut epithelial cells. Mammalian nutrients released after caspase-3/7-dependent apoptosis boosts the growth of multiple Enterobacteriaceae and is observed using primary mouse colonic tissue, mouse and human cell lines, several apoptotic triggers, and in conventional as well as germ-free mice in vivo. The mammalian cell death nutrients induce a core transcriptional response in pathogenic Salmonella, and we identify the pyruvate formate-lyase-encoding pflB gene as a key driver of bacterial colonization in three contexts: a foodborne infection model, a TNF- and A20-dependent cell death model, and a chemotherapy-induced mucositis model. These findings introduce a new layer to the complex host-pathogen interaction, in which death-induced nutrient release acts as a source of fuel for intestinal bacteria, with implications for gut inflammation and cytotoxic chemotherapy treatment.
Collapse
|
36
|
Inhibitory feedback control of NF-κB signalling in health and disease. Biochem J 2021; 478:2619-2664. [PMID: 34269817 PMCID: PMC8286839 DOI: 10.1042/bcj20210139] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/14/2022]
Abstract
Cells must adapt to changes in their environment to maintain cell, tissue and organismal integrity in the face of mechanical, chemical or microbiological stress. Nuclear factor-κB (NF-κB) is one of the most important transcription factors that controls inducible gene expression as cells attempt to restore homeostasis. It plays critical roles in the immune system, from acute inflammation to the development of secondary lymphoid organs, and also has roles in cell survival, proliferation and differentiation. Given its role in such critical processes, NF-κB signalling must be subject to strict spatiotemporal control to ensure measured and context-specific cellular responses. Indeed, deregulation of NF-κB signalling can result in debilitating and even lethal inflammation and also underpins some forms of cancer. In this review, we describe the homeostatic feedback mechanisms that limit and ‘re-set’ inducible activation of NF-κB. We first describe the key components of the signalling pathways leading to activation of NF-κB, including the prominent role of protein phosphorylation and protein ubiquitylation, before briefly introducing the key features of feedback control mechanisms. We then describe the array of negative feedback loops targeting different components of the NF-κB signalling cascade including controls at the receptor level, post-receptor signalosome complexes, direct regulation of the critical ‘inhibitor of κB kinases’ (IKKs) and inhibitory feedforward regulation of NF-κB-dependent transcriptional responses. We also review post-transcriptional feedback controls affecting RNA stability and translation. Finally, we describe the deregulation of these feedback controls in human disease and consider how feedback may be a challenge to the efficacy of inhibitors.
Collapse
|
37
|
Microenvironmental innate immune signaling and cell mechanical responses promote tumor growth. Dev Cell 2021; 56:1884-1899.e5. [PMID: 34197724 DOI: 10.1016/j.devcel.2021.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/01/2021] [Accepted: 06/09/2021] [Indexed: 01/08/2023]
Abstract
Tissue homeostasis is achieved by balancing stem cell maintenance, cell proliferation and differentiation, as well as the purging of damaged cells. Elimination of unfit cells maintains tissue health; however, the underlying mechanisms driving competitive growth when homeostasis fails, for example, during tumorigenesis, remain largely unresolved. Here, using a Drosophila intestinal model, we find that tumor cells outcompete nearby enterocytes (ECs) by influencing cell adhesion and contractility. This process relies on activating the immune-responsive Relish/NF-κB pathway to induce EC delamination and requires a JNK-dependent transcriptional upregulation of the peptidoglycan recognition protein PGRP-LA. Consequently, in organisms with impaired PGRP-LA function, tumor growth is delayed and lifespan extended. Our study identifies a non-cell-autonomous role for a JNK/PGRP-LA/Relish signaling axis in mediating death of neighboring normal cells to facilitate tumor growth. We propose that intestinal tumors "hijack" innate immune signaling to eliminate enterocytes in order to support their own growth.
Collapse
|
38
|
An Overview on Diffuse Large B-Cell Lymphoma Models: Towards a Functional Genomics Approach. Cancers (Basel) 2021; 13:cancers13122893. [PMID: 34207773 PMCID: PMC8226720 DOI: 10.3390/cancers13122893] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Lymphoma research is a paradigm of integrating basic and applied research within the fields of molecular marker-based diagnosis and therapy. In recent years, major advances in next-generation sequencing have substantially improved the understanding of the genomics underlying diffuse large B-cell lymphoma (DLBCL), the most frequent type of B-cell lymphoma. This review addresses the various approaches that have helped unveil the biology and intricate alterations in this pathology, from cell lines to more sophisticated last-generation experimental models, such as organoids. We also provide an overview of the most recent findings in the field, their potential relevance for designing targeted therapies and the corresponding applicability to personalized medicine. Abstract Lymphoma research is a paradigm of the integration of basic and clinical research within the fields of diagnosis and therapy. Clinical, phenotypic, and genetic data are currently used to predict which patients could benefit from standard treatment. However, alternative therapies for patients at higher risk from refractoriness or relapse are usually empirically proposed, based on trial and error, without considering the genetic complexity of aggressive B-cell lymphomas. This is primarily due to the intricate mosaic of genetic and epigenetic alterations in lymphomas, which are an obstacle to the prediction of which drug will work for any given patient. Matching a patient’s genes to drug sensitivity by directly testing live tissues comprises the “precision medicine” concept. However, in the case of lymphomas, this concept should be expanded beyond genomics, eventually providing better treatment options for patients in need of alternative therapeutic approaches. We provide an overview of the most recent findings in diffuse large B-cell lymphomas genomics, from the classic functional models used to study tumor biology and the response to experimental treatments using cell lines and mouse models, to the most recent approaches with spheroid/organoid models. We also discuss their potential relevance and applicability to daily clinical practice.
Collapse
|
39
|
Henneke P, Kierdorf K, Hall LJ, Sperandio M, Hornef M. Perinatal development of innate immune topology. eLife 2021; 10:67793. [PMID: 34032570 PMCID: PMC8149122 DOI: 10.7554/elife.67793] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 05/06/2021] [Indexed: 12/17/2022] Open
Abstract
At the transition from intrauterine to postnatal life, drastic alterations are mirrored by changes in cellular immunity. These changes are in part immune cell intrinsic, originate in the replacement of fetal cells, or result from global regulatory mechanisms and adaptation to changes in the tissue microenvironment. Overall, longer developmental trajectories are intersected by events related to mother-infant separation, birth cues, acquisition of microbiota and metabolic factors. Perinatal alterations particularly affect immune niches, where structures with discrete functions meet, the intestinal mucosa, epidermis and lung. Accordingly, the following questions will be addressed in this review. How does the preprogrammed development supported by endogenous cues, steer innate immune cell differentiation, adaptation to tissue structures, and immunity to infection? How does the transition at birth impact on tissue immune make-up including its topology? How do postnatal cues guide innate immune cell differentiation and function at immunological niches?
Collapse
Affiliation(s)
- Philipp Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Pediatrics and Adolescent Medicine, University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, United Kingdom.,Norwich Medical School, University of East Anglia, Norwich, United Kingdom.,Intestinal Microbiome, School of Life Sciences, and ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Markus Sperandio
- Institute of Cardiovascular Physiology and Pathophysiology, Walter Brendel Center of Experimental Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, RWTH University Hospital Aachen, Aachen, Germany
| |
Collapse
|
40
|
Nissim-Eliraz E, Nir E, Marsiano N, Yagel S, Shpigel NY. NF-kappa-B activation unveils the presence of inflammatory hotspots in human gut xenografts. PLoS One 2021; 16:e0243010. [PMID: 33939711 PMCID: PMC8092666 DOI: 10.1371/journal.pone.0243010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/17/2021] [Indexed: 12/13/2022] Open
Abstract
The single-epithelial cell layer of the gut mucosa serves as an essential barrier between the host and luminal microflora and plays a major role in innate immunity against invading pathogens. Nuclear factor kB (NF-κB), a central component of the cellular signaling machinery, regulates immune response and inflammation. NF-κB proteins are activated by signaling pathways downstream to microbial recognition receptors and cytokines receptors. Highly regulated NF-κB activity in intestinal epithelial cells (IEC) is essential for normal gut homeostasis; dysregulated activity has been linked to a number of disease states, including inflammatory bowel diseases (IBD) such as Crohn's Disease (CD). Our aim was to visualize and quantify spatial and temporal dynamics of NF-κB activity in steady state and inflamed human gut. Lentivirus technology was used to transduce the IEC of human gut xenografts in SCID mice with a NF-κB luminescence reporter system. NF-κB signaling was visualized and quantified using low resolution, intravital imaging of the whole body and high resolution, immunofluorescence microscopic imaging of the tissues. We show that NF-κB is activated in select subset of IEC with low "leaky" NF-κB activity. These unique inflammatory epithelial cells are clustered in the gut into discrete hotspots of NF-κB activity that are visible in steady state and selectively activated by systemic LPS and human TNFα or luminal bacteria. The presence of inflammatory hotspots in the normal and inflamed gut might explain the patchy mucosal lesions characterizing CD and thus could have important implications for diagnosis and therapy.
Collapse
Affiliation(s)
- Einat Nissim-Eliraz
- Department of Basic Sciences, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Eilam Nir
- Department of Basic Sciences, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Noga Marsiano
- Department of Basic Sciences, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| | - Simcha Yagel
- Department of Obstetrics and Gynecology, Hadassah University Hospital, Jerusalem, Israel
| | - Nahum Y. Shpigel
- Department of Basic Sciences, Koret School of Veterinary Medicine, Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
41
|
Speir M, Djajawi TM, Conos SA, Tye H, Lawlor KE. Targeting RIP Kinases in Chronic Inflammatory Disease. Biomolecules 2021; 11:biom11050646. [PMID: 33924766 PMCID: PMC8146010 DOI: 10.3390/biom11050646] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 02/08/2023] Open
Abstract
Chronic inflammatory disorders are characterised by aberrant and exaggerated inflammatory immune cell responses. Modes of extrinsic cell death, apoptosis and necroptosis, have now been shown to be potent drivers of deleterious inflammation, and mutations in core repressors of these pathways underlie many autoinflammatory disorders. The receptor-interacting protein (RIP) kinases, RIPK1 and RIPK3, are integral players in extrinsic cell death signalling by regulating the production of pro-inflammatory cytokines, such as tumour necrosis factor (TNF), and coordinating the activation of the NOD-like receptor protein 3 (NLRP3) inflammasome, which underpin pathological inflammation in numerous chronic inflammatory disorders. In this review, we firstly give an overview of the inflammatory cell death pathways regulated by RIPK1 and RIPK3. We then discuss how dysregulated signalling along these pathways can contribute to chronic inflammatory disorders of the joints, skin, and gastrointestinal tract, and discuss the emerging evidence for targeting these RIP kinases in the clinic.
Collapse
Affiliation(s)
- Mary Speir
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Tirta M. Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Stephanie A. Conos
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Hazel Tye
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
| | - Kate E. Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia; (M.S.); (T.M.D.); (S.A.C.); (H.T.)
- Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
- Correspondence: ; Tel.: +61-85722700
| |
Collapse
|
42
|
Hu J, Yi S, Wang C, Zhang Y, Tang J, Huang X, Yang L, Yang J, Li H. A20 Inhibits Intraocular Inflammation in Mice by Regulating the Function of CD4+T Cells and RPE Cells. Front Immunol 2021; 11:603939. [PMID: 33613524 PMCID: PMC7890008 DOI: 10.3389/fimmu.2020.603939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 12/21/2020] [Indexed: 11/20/2022] Open
Abstract
A20 is a negative regulator of inflammation and immunity and plays a role in several autoimmune and inflammatory diseases. Here, we demonstrate that A20 overexpression significantly ameliorates severity of EAU by inhibiting the infiltration of Th1 and Th17 cells, and by protecting integrity of the blood retinal barrier. In vitro studies showed that A20 silencing could promote CD4+T cells toward a Th1 and Th17 phenotype. A decreased expression of A20 in CD4+T cells was noticed in active BD patients but not in VKH patients. Furthermore, silencing of A20 in hRPE cells induced the production of IL-6, IL-8, and MCP-1 and downregulated ZO-1 and occludin expression which is mediated by inhibition of MAPK and NF-κB pathways. This study reveals a mechanism by which A20 prevents autoimmune uveitis.
Collapse
Affiliation(s)
- Jianping Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Chaokui Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Yiting Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Jihong Tang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Xinyue Huang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Lu Yang
- Department of Ophthalmology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Jinglu Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| | - Hong Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology, Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
43
|
Xie Y, Zhao Y, Shi L, Li W, Chen K, Li M, Chen X, Zhang H, Li T, Matsuzawa-Ishimoto Y, Yao X, Shao D, Ke Z, Li J, Chen Y, Zhang X, Cui J, Cui S, Leng Q, Cadwell K, Li X, Wei H, Zhang H, Li H, Xiao H. Gut epithelial TSC1/mTOR controls RIPK3-dependent necroptosis in intestinal inflammation and cancer. J Clin Invest 2020; 130:2111-2128. [PMID: 31961824 DOI: 10.1172/jci133264] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/14/2020] [Indexed: 02/06/2023] Open
Abstract
Although Western diet and dysbiosis are the most prominent environmental factors associated with inflammatory bowel diseases (IBDs), the corresponding host factors and cellular mechanisms remain poorly defined. Here we report that the TSC1/mTOR pathway in the gut epithelium represents a metabolic and innate immune checkpoint for intestinal dysfunction and inflammation. mTOR hyperactivation triggered by Western diet or Tsc1 ablation led to epithelium necroptosis, barrier disruption, and predisposition to dextran sulfate sodium-induced colitis and inflammation-associated colon cancer. Mechanistically, our results uncovered a critical role for TSC1/mTOR in restraining the expression and activation of RIPK3 in the gut epithelium through TRIM11-mediated ubiquitination and autophagy-dependent degradation. Notably, microbiota depletion by antibiotics or gnotobiotics attenuated RIPK3 expression and activation, thereby alleviating epithelial necroptosis and colitis driven by mTOR hyperactivation. mTOR primarily impinged on RIPK3 to potentiate necroptosis induced by TNF and by microbial pathogen-associated molecular patterns (PAMPs), and hyperactive mTOR and aberrant necroptosis were intertwined in human IBDs. Together, our data reveal a previously unsuspected link between the Western diet, microbiota, and necroptosis and identify the mTOR/RIPK3/necroptosis axis as a driving force for intestinal inflammation and cancer.
Collapse
Affiliation(s)
- Yadong Xie
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China.,Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Yifan Zhao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Lei Shi
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Wei Li
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Kun Chen
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Min Li
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Xia Chen
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Tiantian Li
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Yu Matsuzawa-Ishimoto
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Xiaomin Yao
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Dianhui Shao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Zunfu Ke
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jian Li
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Xiaoming Zhang
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Jun Cui
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuzhong Cui
- State Key Laboratory of Respiratory Diseases, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qibin Leng
- State Key Laboratory of Respiratory Diseases, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, New York, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Hong Wei
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, CAS, Shanghai, China
| | - Huabin Li
- Center for Allergic and Inflammatory Diseases & Department of Otolaryngology, Head and Neck Surgery, Affiliated Eye, Ear, Nose and Throat Hospital, Fudan University, Shanghai, China.,Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Hui Xiao
- The Center for Microbes, Development and Health, Chinese Academy of Sciences (CAS) Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, CAS, Shanghai, China
| |
Collapse
|
44
|
Koudstaal T, van Hulst JAC, Das T, Neys SFH, Merkus D, Bergen IM, de Raaf MA, Bogaard HJ, Boon L, van Loo G, Aerts JGJV, Boomars KA, Kool M, Hendriks RW. DNGR1-Cre-mediated Deletion of Tnfaip3/A20 in Conventional Dendritic Cells Induces Pulmonary Hypertension in Mice. Am J Respir Cell Mol Biol 2020; 63:665-680. [PMID: 32755457 DOI: 10.1165/rcmb.2019-0443oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic perivascular inflammation is a prominent feature in the lungs of idiopathic pulmonary arterial hypertension. Although the proportions of conventional dendritic cells (cDCs) and plasmacytoid DCs are increased in idiopathic pulmonary arterial hypertension lungs, it remains unknown whether activated cDCs play a pathogenic role. The Tnfaip3 gene encodes the ubiquitin-binding protein A20, which is a negative regulator of NF-κB, critically involved in DC activation. Targeting of Tnfaip3/A20 in cDCs was achieved by Clec9a (DNGR1)-Cre-mediated excision of the Tnfaip3 gene in Tnfaip3DNGR1-KO mice. Mice were evaluated for signs of pulmonary hypertension (PH) using right heart catheterization, echocardiography, and measurement of the Fulton index. Inflammation was assessed by immunohistochemistry and flow cytometry. Pulmonary cDCs and monocyte-derived DCs from 31-week-old Tnfaip3DNGR1-KO mice showed modulated expression of cell surface activation markers compared with Tnfaip3DNGR1-WT mice. Tnfaip3DNGR1-KO mice developed elevated right ventricular systolic pressure and right ventricular hypertrophy. The lungs of these mice displayed increased vascular remodeling and perivascular and peribronchial immune cell infiltration resembling tertiary lymphoid organs. Proportions of activated T cells and expression of IL-1β, IL-6, and IL-10 were enhanced in the lungs of Tnfaip3DNGR1-KO mice. Autoreactive IgA and IgG1 was detected in BAL and autoreactive IgA recognizing pulmonary endothelial antigens was present in the serum of Tnfaip3DNGR1-KO mice. All signs of PH were ameliorated in Tnfaip3DNGR1-KO mice by anti-IL-6 antibody treatment. These results indicate that activation of the NF-κB pathway in DCs, through deletion of A20/Tnfaip3, leads to experimental PH with accompanied pulmonary inflammation in an IL-6-dependent fashion.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michiel A de Raaf
- Department of Neonatology, Erasmus Medical Centre, Rotterdam, the Netherlands.,VU Medical Centre, Amsterdam, the Netherlands
| | | | - Louis Boon
- Polpharma Biologics, Utrecht, the Netherlands
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium; and.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | | | | | | |
Collapse
|
45
|
Abstract
A20/TNFAIP3 is a TNF induced gene that plays a profound role in preserving cellular and organismal homeostasis (Lee, et al., 2000; Opipari etal., 1990). This protein has been linked to multiple human diseases via genetic, epigenetic, and an emerging series of patients with mono-allelic coding mutations. Diverse cellular functions of this pleiotropically expressed protein include immune-suppressive, anti-inflammatory, and cell protective functions. The A20 protein regulates ubiquitin dependent cell signals; however, the biochemical mechanisms by which it performs these functions is surprisingly complex. Deciphering these cellular and biochemical facets of A20 dependent biology should greatly improve our understanding of murine and human disease pathophysiology as well as unveil new mechanisms of cell and tissue biology.
Collapse
Affiliation(s)
- Bahram Razani
- Department of Dermatology, University of California, San Francisco, CA, United States
| | - Barbara A Malynn
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, San Francisco, CA, United States.
| |
Collapse
|
46
|
Xiao Y, Huang Q, Wu Z, Chen W. Roles of protein ubiquitination in inflammatory bowel disease. Immunobiology 2020; 225:152026. [PMID: 33190004 DOI: 10.1016/j.imbio.2020.152026] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 08/19/2020] [Accepted: 10/18/2020] [Indexed: 02/08/2023]
Abstract
Inflammatory bowel disease (IBD) including Crohn's disease (CD) and ulcerative colitis (UC) seriously affects the quality of life for patients. The pathogenesis of IBD contains the environmental, host genetic and epigenetic factors. In recent years, the studies of protein ubiquitination, an important protein post-translational modification as an epigenetic factor, have emerged in the pathogenesis and development of IBD. In the past few years, accumulative evidence illustrated that six E3 ubiquitin ligases, namely, ring finger protein (RNF) 183, RNF 20, A20, Pellino 3, TRIM62 and Itch, exhibited clear mechanisms in the development of IBD. They regulate the intestinal inflammation by facilitating the ubiquitination of targeted proteins which participate in different inflammatory signaling pathways. Besides, it was reported that some deubiquitinating enzymes such as Cylindromatosis and USP7 were involved in the development of IBD, but the molecular mechanism was still unclear. This review summarized the role and regulatory mechanism of protein ubiquitination in the pathogenesis and development of IBD, providing insights to develop a new therapeutic strategy in IBD treatments.
Collapse
Affiliation(s)
- Yue Xiao
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Department of Immunology, School of Medicine, Shenzhen University, Shenzhen 516080, China
| | - Qi Huang
- Department of Pediatric Orthopaedics, Shenzhen Children's Hospital, Shenzhen 518035, China
| | - Zherui Wu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Department of Immunology, School of Medicine, Shenzhen University, Shenzhen 516080, China
| | - Weilin Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Marshall Laboratory of Biomedical Engineering, Department of Immunology, School of Medicine, Shenzhen University, Shenzhen 516080, China.
| |
Collapse
|
47
|
Comprehensive Targeted Sequencing Identifies Monogenic Disorders in Patients With Early-onset Refractory Diarrhea. J Pediatr Gastroenterol Nutr 2020; 71:333-339. [PMID: 32487952 DOI: 10.1097/mpg.0000000000002796] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Causes of early-onset refractory diarrhea include exudative diarrhea associated with very early-onset inflammatory bowel diseases, osmotic or secretory diarrhea, and protein-losing enteropathy. Monogenic disorders are included in these diseases, yet a comprehensive genetic analysis has not been fully established. METHODS We established targeted gene panels covering all responsible genes for early-onset diarrhea. In total, 108 patients from 15 institutions were enrolled in this study. We collected clinical data from all patients. Seventy-three patients with exudative diarrhea, 4 with osmotic or secretory diarrhea and 8 with protein-losing enteropathy were subjected to genetic analysis. RESULTS A total of 15 out of the 108 enrolled patients (13.9%) were identified as monogenic. We identified 1 patient with RELA, 2 with TNFAIP3, 1 with CTLA4, 1 with SLCO2A1, 4 with XIAP, 3 with IL10RA, 1 with HPS1, 1 with FOXP3, and 1 with CYBB gene mutations. We also identified 1 patient with NFKB2 and 1 with TERT mutations from the gene panel for primary immunodeficiency syndromes. The patient with refractory diarrhea caused by heterozygous truncated RelA protein expression is the first case identified worldwide, and functional analysis revealed that the mutation affected nuclear factor kappa B signaling. Genotypes were significantly associated with the clinical and pathological findings in each patient. CONCLUSIONS We identified variable monogenic diseases in the patients and found that genes responsible for primary immunodeficiency diseases were frequently involved in molecular pathogenesis. Comprehensive genetic analysis was useful for accurate molecular diagnosis, understanding of underlying pathogenesis, and selecting the optimal treatment for patients with early-onset refractory diarrhea.An infographic for this article is available at: http://links.lww.com/MPG/B853.
Collapse
|
48
|
Mifflin L, Ofengeim D, Yuan J. Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target. Nat Rev Drug Discov 2020; 19:553-571. [PMID: 32669658 PMCID: PMC7362612 DOI: 10.1038/s41573-020-0071-y] [Citation(s) in RCA: 302] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2020] [Indexed: 02/08/2023]
Abstract
Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) is a key mediator of cell death and inflammation. The unique hydrophobic pocket in the allosteric regulatory domain of RIPK1 has enabled the development of highly selective small-molecule inhibitors of its kinase activity, which have demonstrated safety in preclinical models and clinical trials. Potential applications of these RIPK1 inhibitors for the treatment of monogenic and polygenic autoimmune, inflammatory, neurodegenerative, ischaemic and acute conditions, such as sepsis, are emerging. This article reviews RIPK1 biology and disease-associated mutations in RIPK1 signalling pathways, highlighting clinical trials of RIPK1 inhibitors and potential strategies to mitigate development challenges. Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) — a key mediator of cell death and inflammation — is activated in human diseases. Here, Yuan and colleagues discuss current understanding of RIPK1 biology and its association with diseases including inflammatory and autoimmune disorders, neurodegenerative diseases and sepsis. The clinical development of small-molecule RIPK1 inhibitors and associated challenges are discussed.
Collapse
Affiliation(s)
- Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Dimitry Ofengeim
- Rare and Neurologic Disease Research, Sanofi, Framingham, MA, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Mikuda N, Schmidt-Ullrich R, Kärgel E, Golusda L, Wolf J, Höpken UE, Scheidereit C, Kühl AA, Kolesnichenko M. Deficiency in IκBα in the intestinal epithelium leads to spontaneous inflammation and mediates apoptosis in the gut. J Pathol 2020; 251:160-174. [PMID: 32222043 DOI: 10.1002/path.5437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/25/2020] [Accepted: 03/19/2020] [Indexed: 12/15/2022]
Abstract
The IκB kinase (IKK)-NF-κB signaling pathway plays a multifaceted role in inflammatory bowel disease (IBD): on the one hand, it protects from apoptosis; on the other, it activates transcription of numerous inflammatory cytokines and chemokines. Although several murine models of IBD rely on disruption of IKK-NF-κB signaling, these involve either knockouts of a single family member of NF-κB or of upstream kinases that are known to have additional, NF-κB-independent, functions. This has made the distinct contribution of NF-κB to homeostasis in intestinal epithelium cells difficult to assess. To examine the role of constitutive NF-κB activation in intestinal epithelial cells, we generated a mouse model with a tissue-specific knockout of the direct inhibitor of NF-κB, Nfkbia/IκBα. We demonstrate that constitutive activation of NF-κB in intestinal epithelial cells induces several hallmarks of IBD including increased apoptosis, mucosal inflammation in both the small intestine and the colon, crypt hyperplasia, and depletion of Paneth cells, concomitant with aberrant Wnt signaling. To determine which NF-κB-driven phenotypes are cell-intrinsic, and which are extrinsic and thus require the immune compartment, we established a long-term organoid culture. Constitutive NF-κB promoted stem-cell proliferation, mis-localization of Paneth cells, and sensitization of intestinal epithelial cells to apoptosis in a cell-intrinsic manner. Increased number of stem cells was accompanied by a net increase in Wnt activity in organoids. Because aberrant Wnt signaling is associated with increased risk of cancer in IBD patients and because NFKBIA has recently emerged as a risk locus for IBD, our findings have critical implications for the clinic. In a context of constitutive NF-κB, our findings imply that general anti-inflammatory or immunosuppressive therapies should be supplemented with direct targeting of NF-κB within the epithelial compartment in order to attenuate apoptosis, inflammation, and hyperproliferation. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Nadine Mikuda
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Ruth Schmidt-Ullrich
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Eva Kärgel
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Laura Golusda
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, iPATH.Berlin - Core Unit for Immunopathology, Berlin, Germany
| | - Jana Wolf
- Mathematical Modelling of Cellular Processes, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Uta E Höpken
- Microenvironmental Regulation in Autoimmunity and Cancer, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Claus Scheidereit
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Anja A Kühl
- Charité-Universitätsmedizin Berlin, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health, iPATH.Berlin - Core Unit for Immunopathology, Berlin, Germany
| | - Marina Kolesnichenko
- Signal Transduction in Tumour Cells, Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| |
Collapse
|
50
|
Evaluation of the Immunomodulatory Ability of Lactic Acid Bacteria Isolated from Feedlot Cattle Against Mastitis Using a Bovine Mammary Epithelial Cells In Vitro Assay. Pathogens 2020; 9:pathogens9050410. [PMID: 32466097 PMCID: PMC7281661 DOI: 10.3390/pathogens9050410] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 12/27/2022] Open
Abstract
Bovine mastitis, the inflammation of the mammary gland, affects the quality and quantity of milk yield. Mastitis control relies on single or multiple combinations of antibiotic therapy. Due to increasing antibiotic resistance in pathogens, the intramammary infusion of lactic acid bacteria (LAB) has been considered as a potential alternative to antibiotics for treating and preventing bovine mastitis through the improvement of the host immunity. Probiotic effects are a strain-dependent characteristic; therefore, candidate LAB strains have to be evaluated efficiently to find out the ones with the best potential. Here, we investigated LAB strains originally isolated from feedlot cattle’s environment regarding their ability in inducing the Toll-like receptor (TLR)-triggered inflammatory responses in bovine mammary epithelial (BME) cells in vitro. The BME cells were pre-stimulated with the LAB strains individually for 12, 24, and 48 h and then challenged with Escherichia coli-derived lipopolysaccharide (LPS) for 12 h. The mRNA expression of selected immune genes—interleukin 1 alpha (IL-1α), IL-1β, monocyte chemotactic protein 1 (MCP-1), IL-8, chemokine (C-X-C motif) ligand 2 (CXCL2), and CXCL3 were quantified by real-time quantitative PCR (RT-qPCR). Results indicated that pretreatment with some Lactobacillus strains were able to differentially regulate the LPS inflammatory response in BME cells; however, strain-dependent differences were found. The most remarkable effects were found for Lactobacillus acidophilus CRL2074, which reduced the expression of IL-1α, IL-1β, MCP-1, IL-8, and CXCL3, whereas Lactobacillus rhamnosus CRL2084 diminished IL-1β, MCP-1, and IL-8 expression. The pre-stimulation of BME cells with the CRL2074 strain resulted in the upregulated expression of three negative regulators of the TLRs, including the ubiquitin-editing enzyme A20 (also called tumor necrosis factor alpha-induced protein 3, TNFAIP3), single immunoglobin IL-1 single receptor (SIGIRR), and Toll interacting protein (Tollip) after the LPS challenge. The CRL2084 pre-stimulation upregulated only Tollip expression. Our results demonstrated that the L. acidophilus CRL2074 strain possess remarkable immunomodulatory abilities against LPS-induced inflammation in BME cells. This Lactobacillus strain could be used as candidate for in vivo testing due to its beneficial effects in bovine mastitis through intramammary infusion. Our findings also suggest that the BME cells immunoassay system could be of value for the in vitro evaluation of the immunomodulatory abilities of LAB against the inflammation resulting from the intramammary infection with mastitis-related pathogens.
Collapse
|