1
|
Antonello J, Roy P. Damage-Associated Molecular Patterns (DAMPs) In Vascular Diseases. J Biol Chem 2025:110241. [PMID: 40381697 DOI: 10.1016/j.jbc.2025.110241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
Research into the role of chronic sterile inflammation (i.e. a prolonged inflammatory state not caused by an infectious agent), in vascular disease progression has continued to grow over the last few decades. DAMPs have a critical role in this research due to their ability to link stress-causing cardiovascular risk factors to inflammatory phenotypes seen in vascular disease. In this mini-review, we will briefly summarize the DAMPs and receptor signaling pathways that have been extensively studied in the context of vascular disease, including TLRs, RAGE, cGAS-STING, and the NLRP3 inflammasome. In particular, we will discuss how these pathways can promote the release of pro-inflammatory cytokines and chemokines as well as vascular remodeling. Next, we will summarize the results of studies which have linked the various pro-inflammatory effects of DAMPs with the phenotypes in the context of vascular diseases including atherosclerosis, fibrosis, aneurysm, ischemia, and hypertension. Finally, we will discuss some pre-clinical and clinical trials that have targeted DAMPs, their receptors, or the products of their signaling pathways, and discuss the outlook and future directions for the field at large.
Collapse
Affiliation(s)
| | - Partha Roy
- Bioengineering, University of Pittsburgh; Pathology, University of Pittsburgh.
| |
Collapse
|
2
|
Schmidt A, Coughlin M, Catalina MD, Przetak M, Kalatskaya I, Studham M, Shaw J, Bender AT, Strand F. Toll-like receptor 8 activation induces a neutrophil inflammatory phenotype: therapeutic implications for the utility of toll-like receptor 8 inhibition. J Leukoc Biol 2025; 117:qiaf036. [PMID: 40397760 DOI: 10.1093/jleuko/qiaf036] [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: 12/05/2024] [Revised: 02/11/2025] [Indexed: 05/23/2025] Open
Abstract
Excessive activation of toll-like receptor 7 and 8 (TLR7/8) plays a role in the pathogenesis of autoimmune diseases and is associated with negative outcomes from viral infections. Neutrophil activation is highly inflammatory and mediates tissue damage. We explored the effects of TLR7/8 activation in neutrophils to better understand neutrophil biology and evaluate the therapeutic utility of TLR7/8 inhibitors in indications where neutrophils contribute to disease pathogenesis. We found that TLR8, but not TLR7, is active in human neutrophils. TLR8 activation led to increased interleukin-8 (IL-8) secretion and resulted in significant changes in gene expression, as determined by RNA sequencing, with increased expression of genes encoding cytokines and other inflammatory mediators. Type I interferon (IFN) also induced gene expression changes distinct from those induced by TLR8. Additionally, neutrophil extracellular traps (NET) formation and DNA release, or NETosis, was induced by TLR8 activation in IFN-primed neutrophils. Treatment with a TLR7/8 inhibitor (CMPD2) effectively blocked IL-8 secretion and NETosis. In a Phase II clinical trial in COVID-19 pneumonia, TLR7/8 inhibition with enpatoran affected neutrophil counts. Expression of NFKBIZ was induced by TLR8 in neutrophils in vitro and found to also be reduced by enpatoran in patients with COVID-19, suggesting it may be useful as a marker for TLR8-activated neutrophils and for identifying candidate diseases and patients that may benefit from treatment with a TLR7/8 inhibitor. Overall, our findings provide new insights into TLR8 and neutrophil biology that have therapeutic implications in autoimmune diseases and immune-mediated inflammation.
Collapse
Affiliation(s)
- Angelika Schmidt
- Merck Healthcare KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Matthew Coughlin
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Michelle D Catalina
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Melinda Przetak
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Irina Kalatskaya
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Matthew Studham
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Jamie Shaw
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Andrew T Bender
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| | - Fatima Strand
- EMD Serono Research & Development Institute, Inc., 45 Middlesex Turnpike, Billerica, MA 01821, USA, an affiliate of Merck KGaA
| |
Collapse
|
3
|
Du Y, Faz-Lopez B, Ah Kioon MD, Cenac C, Pierides M, Lakin KS, Spiera RF, Chaumeil J, Truchetet ME, Gordon JK, Guéry JC, Barrat FJ. Altered X-chromosome inactivation of the TLR7/8 locus and heterogeneity of pDCs in systemic sclerosis. J Exp Med 2025; 222:e20231809. [PMID: 39670995 PMCID: PMC11639950 DOI: 10.1084/jem.20231809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that has a strong female predominance. Both the X-linked TLR7 and TLR8 can induce type I IFN (IFN-I) by plasmacytoid DCs (pDCs), which can promote fibrosis. We identified five subclusters of pDCs, including ISGhigh clusters that were over-represented in SSc patients. We observed that both TLR7 and TLR8 genes escape from X chromosome inactivation (XCI) at higher frequency in pDCs of SSc patients, which was associated with changes in TLR7 protein profile. Combined DNA/RNA FISH analysis revealed that the TLR7/8 locus is preferentially located outside of the inactive X (Xi) territory when TLR7 is expressed, suggesting that higher-order loop formation is linked to TLR7/8 expression from the Xi. Furthermore, the expression levels of XIST and the transcriptional repressor SPEN were reduced in SSc pDCs. Hence, our data revealed the heterogeneity of pDCs in SSc and suggested that altered XCI at the TLR7/8 locus may contribute to the chronic IFN-I activity of pDCs in female SSc patients.
Collapse
Affiliation(s)
- Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Bérénice Faz-Lopez
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Michael Pierides
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Kimberly S. Lakin
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Robert F. Spiera
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Julie Chaumeil
- Institut Cochin, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Marie-Elise Truchetet
- ImmunoConcEpt, CNRS, UMR 5164, University of Bordeaux, Talence, France
- Rheumatology Department, CHU de Bordeaux, Bordeaux, France
| | - Jessica K. Gordon
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
4
|
Finatto AN, Meurens F, de Oliveira Costa M. Piggybacking on nature: exploring the multifaceted world of porcine β-defensins. Vet Res 2025; 56:47. [PMID: 40033445 PMCID: PMC11877871 DOI: 10.1186/s13567-025-01465-4] [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: 12/31/2023] [Accepted: 12/16/2024] [Indexed: 03/05/2025] Open
Abstract
Porcine β-defensins (pBDs) are cationic peptides that are classically associated with the innate immune system. These molecules yield both antimicrobial and immunomodulatory properties, as evidenced by various in vitro and animal trials. Researchers have revealed that enhancing pBD expression can be achieved through dietary components and gene editing techniques in pigs and porcine cell models. This state-of-the-art review aims to encapsulate the pivotal findings and progress made in the field of pBD over recent decades, with a specific emphasis on the biological role of pBD in infection control and its usage in clinical trials, thereby offering a new landscape of opportunities for research aimed at identifying prophylactic and therapeutic alternatives for both swine medicine and translational purposes.
Collapse
Affiliation(s)
- Arthur Nery Finatto
- Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - François Meurens
- Swine and Poultry Infectious Diseases Research Center, Faculty of Veterinary Medicine, University of Montreal, St. Hyacinthe, QC, J2S 2M2, Canada
- Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada
| | - Matheus de Oliveira Costa
- Large Animal Clinical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, S7N 5E2, Canada.
- Department of Population Health, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
Zeng Y, Tao P, Wang J, Li T, Du Y, Wang X, Wang W, Peng S, Wang W, Ma M, Song H, Yu X, Zhou Q. Somatic gain-of-function mutation in TLR7 causes early-onset systemic lupus erythematosus. Ann Rheum Dis 2025; 84:442-450. [PMID: 39919974 DOI: 10.1016/j.ard.2025.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025]
Abstract
OBJECTIVES We identified a case of early-onset systemic lupus erythematosus (SLE) characterised by acute immune thrombocytopenia, recurrent fever, pneumonia, myocardial damage, thyroid dysfunction, lymphadenopathy, hepatosplenomegaly, and intracranial calcification. Our objective was to investigate the genetic and molecular mechanisms underlying the disease. METHODS Whole exome sequencing and targeted sequencing were performed and a somatic mutation in TLR7 was identified. RNA sequencing, quantitative polymerase chain reaction (qPCR), intracellular cytokine staining, and phospho-flow cytometry were performed to characterise inflammatory signatures. In addition, nuclear factor κB dual-luciferase reporter assays, qPCR, and RNA pull-down assays were performed to assess the functional impact of the TLR7 mutation on immune signalling. RESULTS We identified a novel somatic TLR7 mutation (p.Phe506Ser) that is likely to arise during early embryonic development. This mutation led to transcriptional upregulation of proinflammatory cytokines and interferon-stimulated genes, such as TNF and IFI27, with significant increases in intracellular cytokine expression, including TNF, following stimulation with the ligand single-stranded RNA (ssRNA) and the agonist R848 in the patient's peripheral blood mononuclear cells (PBMCs). In addition, functional analysis in HEK293T cells demonstrated that the mutant TLR7 exhibited increased binding affinity for ssRNA and enhanced responsiveness to agonists, resulting in hyperactivation of TLR7-mediated signalling. CONCLUSIONS We report the first case of early-onset SLE caused by a somatic TLR7 gain-of-function mutation. Our findings demonstrate that the TLR7 F506S mutation drives excessive proinflammatory signalling in the patient's PBMCs, contributing to disease pathogenesis.
Collapse
Affiliation(s)
- Yi Zeng
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China
| | - Panfeng Tao
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China; College of Life Sciences, Zhejiang University, Hangzhou, China.
| | - Jun Wang
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China
| | - Ting Li
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yue Du
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiuli Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Wei Wang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siming Peng
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Wei Wang
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mingsheng Ma
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongmei Song
- Department of Pediatrics, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Xiaomin Yu
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China; Department of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Qing Zhou
- Department of Rheumatology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine and Liangzhu Laboratory of Zhejiang University, Hangzhou, China.
| |
Collapse
|
6
|
Son GY, Zou A, Wahl A, Huang KT, Zorgit S, Vinu M, Zhou F, Wagner L, Idaghdour Y, Yule DI, Feske S, Lacruz RS. Loss of STIM1 and STIM2 in Salivary Glands Disrupts ANO1 Function but Does Not Induce Sjogren's Disease. FUNCTION 2025; 6:zqae047. [PMID: 39479800 PMCID: PMC11815586 DOI: 10.1093/function/zqae047] [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: 06/23/2024] [Revised: 10/02/2024] [Accepted: 10/28/2024] [Indexed: 11/06/2024] Open
Abstract
Ca2+ signaling via the store-operated Ca2+ entry (SOCE) mediated by STIM1 and STIM2 proteins and the ORAI1 Ca2+ channel is important in saliva fluid secretion and has been associated with Sjogren's disease (SjD). However, there are no studies addressing STIM1/2 dysfunction in salivary glands or SjD in animal models. We report that mice lacking Stim1 and Stim2 [Stim1/2K14Cre(+)] in salivary glands exhibited reduced Ca2+ levels and hyposalivate. SOCE was functionally required for the activation of the Ca2+ activated Cl- channel ANO1. Ageing Stim1/2K14Cre(+) mice showed no evidence of lymphocytic infiltration or increased levels of autoantibodies characteristic of SjD, possibly associated with a downregulation of toll-like receptor 8 (Tlr8) expression. Salivary gland biopsies of SjD patients showed increased expression of STIM1 and TLR7/8. Our study shows that SOCE activates ANO1 function and fluid secretion in salivary glands and highlights a potential link between SOCE and TLR signaling in SjD.
Collapse
Affiliation(s)
- Ga-Yeon Son
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| | - Anna Zou
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| | - Amanda Wahl
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Kai Ting Huang
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Saruul Zorgit
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Manikandan Vinu
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - Fang Zhou
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10010, USA
| | - Larry Wagner
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Youssef Idaghdour
- Biology Program, Division of Science and Mathematics, New York University Abu Dhabi, Abu Dhabi 129188, United Arab Emirates
| | - David I Yule
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York 14642, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, New York 10010, USA
| | - Rodrigo S Lacruz
- Department of Molecular Pathobiology, New York University College of Dentistry, New York 10010, USA
| |
Collapse
|
7
|
Rai P, Fessler MB. Mechanisms and effects of activation of innate immunity by mitochondrial nucleic acids. Int Immunol 2025; 37:133-142. [PMID: 39213393 DOI: 10.1093/intimm/dxae052] [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: 07/10/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
In recent years, a growing number of roles have been identified for mitochondria in innate immunity. One principal mechanism is that the translocation of mitochondrial nucleic acid species from the mitochondrial matrix to the cytosol and endolysosomal lumen in response to an array of microbial and non-microbial environmental stressors has been found to serve as a second messenger event in the cell signaling of the innate immune response. Thus, mitochondrial DNA and RNA have been shown to access the cytosol through several regulated mechanisms involving remodeling of the mitochondrial inner and outer membranes and to access lysosomes via vesicular transport, thereby activating cytosolic [e.g. cyclic GMP-AMP synthase (cGAS), retinoic acid-inducible gene I (RIG-I)-like receptors], and endolysosomal (Toll-like receptor 7, 9) nucleic acid receptors that induce type I interferons and pro-inflammatory cytokines. In this mini-review, we discuss these molecular mechanisms of mitochondrial nucleic acid mislocalization and their roles in host defense, autoimmunity, and auto-inflammatory disorders. The emergent paradigm is one in which host-derived DNA interestingly serves as a signal amplifier in the innate immune response and also as an alarm signal for disturbances in organellar homeostasis. The apparent vast excess of mitochondria and mitochondrial DNA nucleoids per cell may thus serve to sensitize the cell response to stressors while ensuring an underlying reserve of intact mitochondria to sustain cellular metabolism. An improved understanding of these molecular mechanisms will hopefully afford future opportunities for therapeutic intervention in human disease.
Collapse
Affiliation(s)
- Prashant Rai
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| | - Michael B Fessler
- Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, USA
| |
Collapse
|
8
|
Ehlers C, Thiele T, Biermann H, Traidl S, Bruns L, Ziegler A, Schefzyk M, Bartsch LM, Kalinke U, Witte T, Graalmann T. Toll-Like Receptor 8 is Expressed in Monocytes in Contrast to Plasmacytoid Dendritic Cells and Mediates Aberrant Interleukin-10 Responses in Patients With Systemic Sclerosis. Arthritis Rheumatol 2025; 77:59-66. [PMID: 39112920 PMCID: PMC11685002 DOI: 10.1002/art.42964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024]
Abstract
OBJECTIVE Systemic sclerosis (SSc) is a severe rheumatic disease causing fibrotic tissue rearrangement. Aberrant toll-like receptor (TLR) 8 transcripts in plasmacytoid dendritic cells (pDCs) were recently linked to SSc pathogenesis, which is at least partially mediated by increased type I interferon (IFN-I) responses. Here, we addressed the functional role of TLR8 signaling in different immune cell subsets of patients with SSc. METHODS Monocytes, conventional dendritic cells (cDCs), and pDCs from the blood and skin of patients with SSc were analyzed for TLR8 protein expression. To assess TLR function, cytokine responses upon TLR7 and TLR8 stimulation were studied. To identify relevant alterations specific for patients with SSc (n = 16), patients with primary Sjögren disease (pSS; n = 10) and healthy controls (HCs; n = 13) were included into the study. RESULTS In all individuals, TLR8 was expressed in monocytes and cDCs but not in pDCs. The TLR8 expression levels were overall similar in patients with SSc and pSS and HCs. Additionally, in all study participants, TLR8 stimulation of pDCs did not induce IFN-I expression. In contrast, monocytes from patients with SSc revealed increased interleukin (IL)-10 responses upon TLR8 (patients with SSc vs HCs, P = 0.0126) and TLR7/8 stimulation (patients with SSc vs HCs, P = 0.0170). CONCLUSION TLR8 protein is not expressed in pDCs of patients with SSc. Accordingly, they do not respond to TLR8 stimulation. In contrast, monocytes of patients with SSc respond to TLR8 stimulation with increased IL-10 responses. Therefore, TLR8 signaling in monocytes participates in SSc pathogenesis by conferring aberrant IL-10 expression.
Collapse
Affiliation(s)
- Christine Ehlers
- Junior Research Group for Translational Immunology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany; Biomedical Research in End‐Stage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL)HannoverGermany
| | - Thea Thiele
- Department for Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Hannah Biermann
- Junior Research Group for Translational Immunology, TWINCORE Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical SchoolHannoverGermany
| | - Stephan Traidl
- Department for Dermatology and AllergyHannover Medical SchoolHannoverGermany
| | - Luzia Bruns
- Department for Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Annett Ziegler
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical SchoolHannoverGermany
| | - Matthias Schefzyk
- Department for Dermatology and AllergyHannover Medical SchoolHannoverGermany
| | - Lea M. Bartsch
- Department for Gastroenterology, Hepatology, Infectious Diseases and EndocrinologyHannover Medical SchoolHannoverGermany
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical SchoolHannoverGermany
| | - Torsten Witte
- Department for Rheumatology and ImmunologyHannover Medical SchoolHannoverGermany
| | - Theresa Graalmann
- Junior Research Group for Translational Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany, Biomedical Research in End‐Stage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover, Germany, and Department for Rheumatology and Immunology, Hannover Medical SchoolHannoverGermany
| |
Collapse
|
9
|
Cui Y, Zhang Y, Yang C. TBK1 and Caspase-8 suppress necroptosis but activate NLRP3 inflammasome activation during CXCL4 and TLR8 costimulation. Cell Death Discov 2024; 10:511. [PMID: 39715768 DOI: 10.1038/s41420-024-02280-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/10/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Affiliation(s)
- Ying Cui
- Department of Blood Transfusion, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaan Xi, China
| | - Yaguang Zhang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China
| | - Chao Yang
- Department of Blood Transfusion, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaan Xi, China.
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
10
|
Li J, Zhu J, Yang H, Hou F. Sterile activation of RNA-sensing pathways in autoimmunity. J Mol Cell Biol 2024; 16:mjae029. [PMID: 39143032 PMCID: PMC11659683 DOI: 10.1093/jmcb/mjae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 06/27/2024] [Accepted: 08/13/2024] [Indexed: 08/16/2024] Open
Abstract
RNA-sensing pathways play a pivotal role in host defense against pathogenic infections to maintain cellular homeostasis. However, in the absence of infection, certain endogenous RNAs can serve as the activators of RNA-sensing pathways as well. The inappropriate activation of RNA-sensing pathways by self-ligands leads to systemic inflammation and autoimmune diseases. In this review, we summarize current findings on the sterile activation of RNA sensors, as well as its implications in autoimmunity, inflammatory diseases, and therapeutics.
Collapse
Affiliation(s)
- Jiaxin Li
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Junyan Zhu
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Fajian Hou
- Key Laboratory of RNA Science and Engineering, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
11
|
Hamerman JA, Barton GM. The path ahead for understanding Toll-like receptor-driven systemic autoimmunity. Curr Opin Immunol 2024; 91:102482. [PMID: 39353255 PMCID: PMC11863706 DOI: 10.1016/j.coi.2024.102482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
Five mammalian Toll-like receptors (TLR 3, 7, 8, 9, and 13) recognize nucleic acids (NA) and induce signals that control the function of multiple immune cell types and initiate both innate and adaptive immune responses. While these receptors enable recognition of diverse microbial threats, in some instances, they respond inappropriately to self-NA released from host cells and drive the development of autoimmune diseases. Specifically, activation of TLR7 and TLR8 by self-RNA and TLR9 by self-DNA has been linked to development of a collection of systemic autoimmune or autoinflammatory disorders, including systemic lupus erythematosus, systemic juvenile idiopathic arthritis, and macrophage activation syndrome. Here, we discuss recent progress in understanding how these receptors contribute to such diverse clinical phenotypes. We highlight how comparative studies between mice and humans have not only been beneficial in identifying key pathways relevant for disease but also reveal gaps in our understanding of disease mechanisms. We identify several challenges that we hope the field will tackle in the years ahead.
Collapse
Affiliation(s)
- Jessica A Hamerman
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA 98101, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| | - Gregory M Barton
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
12
|
Rael VE, Yano JA, Huizar JP, Slayden LC, Weiss MA, Turcotte EA, Terry JM, Zuo W, Thiffault I, Pastinen T, Farrow EG, Jenkins JL, Becker ML, Wong SC, Stevens AM, Otten C, Allenspach EJ, Bonner DE, Bernstein JA, Wheeler MT, Saxton RA, Liu B, Majer O, Barton GM. Large-scale mutational analysis identifies UNC93B1 variants that drive TLR-mediated autoimmunity in mice and humans. J Exp Med 2024; 221:e20232005. [PMID: 38780621 PMCID: PMC11116816 DOI: 10.1084/jem.20232005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/21/2024] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Nucleic acid-sensing Toll-like receptors (TLR) 3, 7/8, and 9 are key innate immune sensors whose activities must be tightly regulated to prevent systemic autoimmune or autoinflammatory disease or virus-associated immunopathology. Here, we report a systematic scanning-alanine mutagenesis screen of all cytosolic and luminal residues of the TLR chaperone protein UNC93B1, which identified both negative and positive regulatory regions affecting TLR3, TLR7, and TLR9 responses. We subsequently identified two families harboring heterozygous coding mutations in UNC93B1, UNC93B1+/T93I and UNC93B1+/R336C, both in key negative regulatory regions identified in our screen. These patients presented with cutaneous tumid lupus and juvenile idiopathic arthritis plus neuroinflammatory disease, respectively. Disruption of UNC93B1-mediated regulation by these mutations led to enhanced TLR7/8 responses, and both variants resulted in systemic autoimmune or inflammatory disease when introduced into mice via genome editing. Altogether, our results implicate the UNC93B1-TLR7/8 axis in human monogenic autoimmune diseases and provide a functional resource to assess the impact of yet-to-be-reported UNC93B1 mutations.
Collapse
Affiliation(s)
- Victoria E. Rael
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Julian A. Yano
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - John P. Huizar
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Leianna C. Slayden
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Madeleine A. Weiss
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Elizabeth A. Turcotte
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Jacob M. Terry
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Wenqi Zuo
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Isabelle Thiffault
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital, Kansas City, MO, USA
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Tomi Pastinen
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Emily G. Farrow
- Department of Pathology and Laboratory Medicine, Children’s Mercy Hospital, Kansas City, MO, USA
- Genomic Medicine Center, Children’s Mercy Hospital, Kansas City, MO, USA
- University of Missouri Kansas City School of Medicine, Kansas City, MO, USA
| | - Janda L. Jenkins
- Department of Genetics, Children’s Mercy Hospital, Kansas City, MO, USA
| | - Mara L. Becker
- Division of Rheumatology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Stephen C. Wong
- Division of Rheumatology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA, USA
| | - Anne M. Stevens
- Division of Rheumatology, Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA, USA
- Johnson & Johnson Innovative Medicine, Spring House, PA, USA
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | - Catherine Otten
- Division of Pediatric Neurology, Department of Neurology, Seattle Children’s Hospital, University of Washington, Seattle, WA, USA
| | - Eric J. Allenspach
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Devon E. Bonner
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Medical Genetics, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan A. Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Medical Genetics, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew T. Wheeler
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert A. Saxton
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Bo Liu
- Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Olivia Majer
- Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gregory M. Barton
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
13
|
Ah Kioon MD, Laurent P, Chaudhary V, Du Y, Crow MK, Barrat FJ. Modulation of plasmacytoid dendritic cells response in inflammation and autoimmunity. Immunol Rev 2024; 323:241-256. [PMID: 38553621 DOI: 10.1111/imr.13331] [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] [Indexed: 05/18/2024]
Abstract
The discovery of toll-like receptors (TLRs) and the subsequent recognition that endogenous nucleic acids (NAs) could serve as TLR ligands have led to essential insights into mechanisms of healthy immune responses as well as pathogenic mechanisms relevant to systemic autoimmune and inflammatory diseases. In systemic lupus erythematosus, systemic sclerosis, and rheumatoid arthritis, NA-containing immune complexes serve as TLR ligands, with distinct implications depending on the additional immune stimuli available. Plasmacytoid dendritic cells (pDCs), the robust producers of type I interferon (IFN-I), are providing critical insights relevant to TLR-mediated healthy immune responses and tissue repair, as well as generation of inflammation, autoimmunity and fibrosis, processes central to the pathogenesis of many autoimmune diseases. In this review, we describe recent data characterizing the role of platelets and NA-binding chemokines in modulation of TLR signaling in pDCs, as well as implications for how the IFN-I products of pDCs contribute to the generation of inflammation and wound healing responses by monocyte/macrophages. Chemokine modulators of TLR-mediated B cell tolerance mechanisms and interactions between TLR signaling and metabolic pathways are also considered. The modulators of TLR signaling and their contribution to the pathogenesis of systemic autoimmune diseases suggest new opportunities for identification of novel therapeutic targets.
Collapse
Affiliation(s)
| | - Paôline Laurent
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Vidyanath Chaudhary
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Yong Du
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
| | - Mary K Crow
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, New York, New York, USA
- Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Franck J Barrat
- HSS Research Institute, Hospital for Special Surgery, New York, New York, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, New York, USA
- David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, New York, USA
| |
Collapse
|
14
|
Wang T, Song D, Li X, Luo Y, Yang D, Liu X, Kong X, Xing Y, Bi S, Zhang Y, Hu T, Zhang Y, Dai S, Shao Z, Chen D, Hou J, Ballestar E, Cai J, Zheng F, Yang JY. MiR-574-5p activates human TLR8 to promote autoimmune signaling and lupus. Cell Commun Signal 2024; 22:220. [PMID: 38589923 PMCID: PMC11000404 DOI: 10.1186/s12964-024-01601-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/28/2024] [Indexed: 04/10/2024] Open
Abstract
Endosomal single-stranded RNA-sensing Toll-like receptor-7/8 (TLR7/8) plays a pivotal role in inflammation and immune responses and autoimmune diseases. However, the mechanisms underlying the initiation of the TLR7/8-mediated autoimmune signaling remain to be fully elucidated. Here, we demonstrate that miR-574-5p is aberrantly upregulated in tissues of lupus prone mice and in the plasma of lupus patients, with its expression levels correlating with the disease activity. miR-574-5p binds to and activates human hTLR8 or its murine ortholog mTlr7 to elicit a series of MyD88-dependent immune and inflammatory responses. These responses include the overproduction of cytokines and interferons, the activation of STAT1 signaling and B lymphocytes, and the production of autoantigens. In a transgenic mouse model, the induction of miR-574-5p overexpression is associated with increased secretion of antinuclear and anti-dsDNA antibodies, increased IgG and C3 deposit in the kidney, elevated expression of inflammatory genes in the spleen. In lupus-prone mice, lentivirus-mediated silencing of miR-574-5p significantly ameliorates major symptoms associated with lupus and lupus nephritis. Collectively, these results suggest that the miR-574-5p-hTLR8/mTlr7 signaling is an important axis of immune and inflammatory responses, contributing significantly to the development of lupus and lupus nephritis.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
- The Key Laboratory of Urinary Tract Tumors and Calculi, Department of Urology, School of Medicine, The First Affiliated Hospital of Xiamen University, Xiamen University, Xiamen, 361003, China
| | - Dan Song
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Xuejuan Li
- Wuhu Hospital of East China Normal University, Wuhu, Anhui, 241000, China
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China
- Department of Nephrology, The Second Hospital, Dalian Medical University, Dalian, 116144, China
| | - Yu Luo
- School of Nursing, The Third Military Medical University, Chongqing, 400038, China
| | - Dianqiang Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Xiaoyan Liu
- Department of Nephrology, The Second Hospital, Dalian Medical University, Dalian, 116144, China
| | - Xiaodan Kong
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yida Xing
- Department of Rheumatology, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | - Shulin Bi
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Yan Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Tao Hu
- College of Medicine, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Yunyun Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Shuang Dai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Zhiqiang Shao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Dahan Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Jinpao Hou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China
| | - Esteban Ballestar
- Wuhu Hospital of East China Normal University, Wuhu, Anhui, 241000, China
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, 08916, Spain
| | - Jianchun Cai
- Department of Gastrointestinal Surgery, Institute of Gastrointestinal Oncology, Zhongshan Hospital of Xiamen University, Medical College of Xiamen University, Xiamen, Fujian, 361005, China.
| | - Feng Zheng
- Wuhu Hospital of East China Normal University, Wuhu, Anhui, 241000, China.
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China.
- Department of Nephrology, The Second Hospital, Dalian Medical University, Dalian, 116144, China.
- The Advanced Institute for Molecular Medicine, Dalian Medical University, Dalian, 116144, China.
| | - James Y Yang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China.
- Wuhu Hospital of East China Normal University, Wuhu, Anhui, 241000, China.
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China.
| |
Collapse
|
15
|
Gillard J, Suffiotti M, Brazda P, Venkatasubramanian PB, Versteegen P, de Jonge MI, Kelly D, Bibi S, Pinto MV, Simonetti E, Babiceanu M, Kettring A, Teodosio C, de Groot R, Berbers G, Stunnenberg HG, Schanen B, Fenwick C, Huynen MA, Diavatopoulos DA. Antiviral responses induced by Tdap-IPV vaccination are associated with persistent humoral immunity to Bordetella pertussis. Nat Commun 2024; 15:2133. [PMID: 38459022 PMCID: PMC10923912 DOI: 10.1038/s41467-024-46560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/01/2024] [Indexed: 03/10/2024] Open
Abstract
Many countries continue to experience pertussis epidemics despite widespread vaccination. Waning protection after booster vaccination has highlighted the need for a better understanding of the immunological factors that promote durable protection. Here we apply systems vaccinology to investigate antibody responses in adolescents in the Netherlands (N = 14; NL) and the United Kingdom (N = 12; UK) receiving a tetanus-diphtheria-acellular pertussis-inactivated poliovirus (Tdap-IPV) vaccine. We report that early antiviral and interferon gene expression signatures in blood correlate to persistence of pertussis-specific antibody responses. Single-cell analyses of the innate response identified monocytes and myeloid dendritic cells (MoDC) as principal responders that upregulate antiviral gene expression and type-I interferon cytokine production. With public data, we show that Tdap vaccination stimulates significantly lower antiviral/type-I interferon responses than Tdap-IPV, suggesting that IPV may promote antiviral gene expression. Subsequent in vitro stimulation experiments demonstrate TLR-dependent, IPV-specific activation of the pro-inflammatory p38 MAP kinase pathway in MoDCs. Together, our data provide insights into the molecular host response to pertussis booster vaccination and demonstrate that IPV enhances innate immune activity associated with persistent, pertussis-specific antibody responses.
Collapse
Affiliation(s)
- Joshua Gillard
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Peter Brazda
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Pauline Versteegen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dominic Kelly
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sagida Bibi
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Marta Valente Pinto
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Caparica, Almada, Portugal
| | - Elles Simonetti
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Cristina Teodosio
- Leiden University Medical Center, Immunohematology & Blood Transfusion, Leiden, The Netherlands
| | - Ronald de Groot
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Guy Berbers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | | | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Martijn A Huynen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
16
|
Tong AJ, Leylek R, Herzner AM, Rigas D, Wichner S, Blanchette C, Tahtinen S, Kemball CC, Mellman I, Haley B, Freund EC, Delamarre L. Nucleotide modifications enable rational design of TLR7-selective ligands by blocking RNase cleavage. J Exp Med 2024; 221:e20230341. [PMID: 38095631 PMCID: PMC10720541 DOI: 10.1084/jem.20230341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 10/10/2023] [Accepted: 11/21/2023] [Indexed: 12/17/2023] Open
Abstract
Toll-like receptors 7 (TLR7) and 8 (TLR8) each sense single-stranded RNA (ssRNA), but their activation results in different immune activation profiles. Attempts to selectively target either TLR7 or TLR8 have been hindered by their high degree of homology. However, recent studies revealed that TLR7 and TLR8 bind different ligands resulting from the processing of ssRNA by endolysosomal RNases. We demonstrate that by introducing precise 2' sugar-modified bases into oligoribonucleotides (ORNs) containing known TLR7 and TLR8 binding motifs, we could prevent RNase-mediated degradation into the monomeric uridine required for TLR8 activation while preserving TLR7 activation. Furthermore, a novel, optimized protocol for CRISPR-Cas9 knockout in primary human plasmacytoid dendritic cells showed that TLR7 activation is dependent on RNase processing of ORNs and revealed a previously undescribed role for RNase 6 in degrading ORNs into TLR ligands. Finally, 2' sugar-modified ORNs demonstrated robust innate immune activation in mice. Altogether, we identified a strategy for creating tunable TLR7-selective agonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ira Mellman
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | |
Collapse
|
17
|
Son GY, Zou A, Wahl A, Huang KT, Vinu M, Zorgit S, Zhou F, Wagner L, Idaghdour Y, Yule DI, Feske S, Lacruz RS. Loss of STIM1 and STIM2 in salivary glands disrupts ANO1 function but does not induce Sjogren's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574702. [PMID: 38260625 PMCID: PMC10802497 DOI: 10.1101/2024.01.08.574702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Sjogren's disease (SjD) is an autoimmune disease characterized by xerostomia (dry mouth), lymphocytic infiltration into salivary glands and the presence of SSA and SSB autoantibodies. Xerostomia is caused by hypofunction of the salivary glands and has been involved in the development of SjD. Saliva production is regulated by parasympathetic input into the glands initiating intracellular Ca 2+ signals that activate the store operated Ca 2+ entry (SOCE) pathway eliciting sustained Ca 2+ influx. SOCE is mediated by the STIM1 and STIM2 proteins and the ORAI1 Ca 2+ channel. However, there are no studies on the effects of lack of STIM1/2 function in salivary acini in animal models and its impact on SjD. Here we report that male and female mice lacking Stim1 and Stim2 ( Stim1/2 K14Cre ) in salivary glands showed reduced intracellular Ca 2+ levels via SOCE in parotid acini and hyposalivate upon pilocarpine stimulation. Bulk RNASeq of the parotid glands of Stim1/2 K14Cre mice showed a decrease in the expression of Stim1/2 but no other Ca 2+ associated genes mediating saliva fluid secretion. SOCE was however functionally required for the activation of the Ca 2+ activated chloride channel ANO1. Despite hyposalivation, ageing Stim1/2 K14Cre mice showed no evidence of lymphocytic infiltration in the glands or elevated levels of SSA or SSB autoantibodies in the serum, which may be linked to the downregulation of the toll-like receptor 8 ( Tlr8 ). By contrast, salivary gland biopsies of SjD patients showed increased STIM1 and TLR8 expression, and induction of SOCE in a salivary gland cell line increased the expression of TLR8 . Our data demonstrate that SOCE is an important activator of ANO1 function and saliva fluid secretion in salivary glands. They also provide a novel link between SOCE and TLR8 signaling which may explain why loss of SOCE does not result in SjD.
Collapse
|
18
|
Abstract
Systemic autoinflammatory diseases (SAIDs) are a heterogeneous group of disorders caused by excess activation of the innate immune system in an antigen-independent manner. Starting with the discovery of the causal gene for familial Mediterranean fever, more than 50 monogenic SAIDs have been described. These discoveries, paired with advances in immunology and genomics, have allowed our understanding of these diseases to improve drastically in the last decade. The genetic causes of SAIDs are complex and include both germline and somatic pathogenic variants that affect various inflammatory signaling pathways. We provide an overview of the acquired SAIDs from a genetic perspective and summarize the clinical phenotypes and mechanism(s) of inflammation, aiming to provide a comprehensive understanding of the pathogenesis of autoinflammatory diseases.
Collapse
Affiliation(s)
- Jiahui Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA;
| | - Qing Zhou
- Life Sciences Institute, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China;
| |
Collapse
|
19
|
Ishizaka ST, Hawkins L, Chen Q, Tago F, Yagi T, Sakaniwa K, Zhang Z, Shimizu T, Shirato M. A novel Toll-like receptor 7/8-specific antagonist E6742 ameliorates clinically relevant disease parameters in murine models of lupus. Eur J Pharmacol 2023; 957:175962. [PMID: 37544422 DOI: 10.1016/j.ejphar.2023.175962] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
The sensing of self RNA by the endosomal Toll-like receptors (TLRs) 7 and 8 initiates pathogenic mechanisms underlying the autoimmune disease lupus. A blockade of the TLR7/8 signals may, therefore, be a novel therapeutic intervention for lupus. To test the hypothesis, a novel compound E6742 that blocks TLR7/8 activation was identified. The mode of action of E6742 was investigated by analysis of the tertiary structure of TLR7 and 8 in complex with E6742. The in vitro activities of the compound were examined in cellular systems and its therapeutic potential was evaluated in murine lupus models. Tertiary structures of the extracellular domain of TLR7 and 8 in complex with E6742 showed that E6742 binds specifically and non-covalently to the hydrophobic pocket located at the interface of TLR7 or TLR8 homodimers. E6742 potently and selectively inhibited several TLR7/8-mediated cytokine responses in human PBMC. In two mouse models of lupus, oral dosing of E6742 after the onset of disease suppressed increase in autoantibodies and blocked the advance of organ damage. Collectively, the data show that TLR7/8 activation contributes to disease progression and its blocking by E6742 has potential as a therapeutic intervention for lupus.
Collapse
Affiliation(s)
- Sally T Ishizaka
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | - Lynn Hawkins
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | - Qian Chen
- Eisai Inc., Eisai Center for Genetics Guided Dementia Discovery, MA, USA
| | | | | | - Kentaro Sakaniwa
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Zhikuan Zhang
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Manabu Shirato
- Eisai Co., Ltd., Tsukuba Research Laboratories, Ibaraki, Japan.
| |
Collapse
|
20
|
Maria NI, Papoin J, Raparia C, Sun Z, Josselsohn R, Lu A, Katerji H, Syeda MM, Polsky D, Paulson R, Kalfa T, Barnes BJ, Zhang W, Blanc L, Davidson A. Human TLR8 induces inflammatory bone marrow erythromyeloblastic islands and anemia in SLE-prone mice. Life Sci Alliance 2023; 6:e202302241. [PMID: 37495396 PMCID: PMC10372407 DOI: 10.26508/lsa.202302241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023] Open
Abstract
Anemia commonly occurs in systemic lupus erythematosus, a disease characterized by innate immune activation by nucleic acids. Overactivation of cytoplasmic sensors by self-DNA or RNA can cause erythroid cell death, while sparing other hematopoietic cell lineages. Whereas chronic inflammation is involved in this mechanism, less is known about the impact of systemic lupus erythematosus on the BM erythropoietic niche. We discovered that expression of the endosomal ssRNA sensor human TLR8 induces fatal anemia in Sle1.Yaa lupus mice. We observed that anemia was associated with a decrease in erythromyeloblastic islands and a block in differentiation at the CFU-E to proerythroblast transition in the BM. Single-cell RNAseq analyses of isolated BM erythromyeloblastic islands from human TLR8-expressing mice revealed that genes associated with essential central macrophage functions including adhesion and provision of nutrients were down-regulated. Although compensatory stress erythropoiesis occurred in the spleen, red blood cell half-life decreased because of hemophagocytosis. These data implicate the endosomal RNA sensor TLR8 as an additional innate receptor whose overactivation causes acquired failure of erythropoiesis via myeloid cell dysregulation.
Collapse
Affiliation(s)
- Naomi I Maria
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Julien Papoin
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Chirag Raparia
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Zeguo Sun
- Department of Medicine, Mount Sinai Medical Center, New York, NY, USA
| | - Rachel Josselsohn
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Ailing Lu
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hani Katerji
- Department of Pathology, University of Rochester, Rochester, NY, USA
| | - Mahrukh M Syeda
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - David Polsky
- The Ronald O. Perelman Department of Dermatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Robert Paulson
- Department of Veterinary and Biomedical Sciences, Penn State College of Agricultural Sciences, University Park, PA, USA
| | - Theodosia Kalfa
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Betsy J Barnes
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Weijia Zhang
- Department of Medicine, Mount Sinai Medical Center, New York, NY, USA
| | - Lionel Blanc
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| | - Anne Davidson
- Institute of Molecular Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Northwell Health, Hempstead, NY, USA
| |
Collapse
|
21
|
Youness A, Cenac C, Faz-López B, Grunenwald S, Barrat FJ, Chaumeil J, Mejía JE, Guéry JC. TLR8 escapes X chromosome inactivation in human monocytes and CD4 + T cells. Biol Sex Differ 2023; 14:60. [PMID: 37723501 PMCID: PMC10506212 DOI: 10.1186/s13293-023-00544-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 09/04/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND Human endosomal Toll-like receptors TLR7 and TLR8 recognize self and non-self RNA ligands, and are important mediators of innate immunity and autoimmune pathogenesis. TLR7 and TLR8 are, respectively, encoded by adjacent X-linked genes. We previously established that TLR7 evades X chromosome inactivation (XCI) in female immune cells. Whether TLR8 also evades XCI, however, has not yet been explored. METHOD In the current study, we used RNA fluorescence in situ hybridization (RNA FISH) to directly visualize, on a single-cell basis, primary transcripts of TLR7 and TLR8 relative to X chromosome territories in CD14+ monocytes and CD4+ T lymphocytes from women, Klinefelter syndrome (KS) men, and euploid men. To assign X chromosome territories in cells lacking robust expression of a XIST compartment, we designed probes specific for X-linked genes that do not escape XCI and therefore robustly label the active X chromosome. We also assessed whether XCI escape of TLR8 was associated with sexual dimorphism in TLR8 protein expression by western blot and flow cytometry. RESULTS Using RNA FISH, we show that TLR8, like TLR7, evades XCI in immune cells, and that cells harboring simultaneously TLR7 and TLR8 transcript foci are more frequent in women and KS men than in euploid men, resulting in a sevenfold difference in frequency. This transcriptional bias was again observable when comparing the single X of XY males with the active X of cells from females or KS males. Interestingly, TLR8 protein expression was significantly higher in female mononuclear blood cells, including all monocyte subsets, than in male cells. CONCLUSIONS TLR8, mirroring TLR7, escapes XCI in human monocytes and CD4+ T cells. Co-dependent transcription from the active X chromosome and escape from XCI could both contribute to higher TLR8 protein abundance in female cells, which may have implications for the response to viruses and bacteria, and the risk of developing inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Ali Youness
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), UMR 1291 INSERM, CNRS, Hôpital Purpan, Université de Toulouse, 31024, Toulouse, France
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), UMR 1291 INSERM, CNRS, Hôpital Purpan, Université de Toulouse, 31024, Toulouse, France
| | - Berenice Faz-López
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), UMR 1291 INSERM, CNRS, Hôpital Purpan, Université de Toulouse, 31024, Toulouse, France
| | - Solange Grunenwald
- Service d'Endocrinologie, Maladies Métaboliques et Nutrition, Hôpital Larrey, Centre Hospitalier Universitaire (CHU) de Toulouse, 31059, Toulouse, France
| | - Franck J Barrat
- Hospital for Special Surgery, HSS Research Institute and David Z. Rosensweig Genomics Research Center, New York, NY, 10021, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, 10021, USA
| | - Julie Chaumeil
- INSERM, CNRS, Université Paris Cité, Institut Cochin, 75014, Paris, France
| | - José Enrique Mejía
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), UMR 1291 INSERM, CNRS, Hôpital Purpan, Université de Toulouse, 31024, Toulouse, France.
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires (INFINITY), UMR 1291 INSERM, CNRS, Hôpital Purpan, Université de Toulouse, 31024, Toulouse, France.
| |
Collapse
|
22
|
Shibata T, Sato R, Taoka M, Saitoh SI, Komine M, Yamaguchi K, Goyama S, Motoi Y, Kitaura J, Izawa K, Yamauchi Y, Tsukamoto Y, Ichinohe T, Fujita E, Hiranuma R, Fukui R, Furukawa Y, Kitamura T, Takai T, Tojo A, Ohtsuki M, Ohto U, Shimizu T, Ozawa M, Yoshida N, Isobe T, Latz E, Mukai K, Taguchi T, Hemmi H, Akira S, Miyake K. TLR7/8 stress response drives histiocytosis in SLC29A3 disorders. J Exp Med 2023; 220:e20230054. [PMID: 37462944 PMCID: PMC10354536 DOI: 10.1084/jem.20230054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 05/02/2023] [Accepted: 06/09/2023] [Indexed: 07/21/2023] Open
Abstract
Loss-of-function mutations in the lysosomal nucleoside transporter SLC29A3 cause lysosomal nucleoside storage and histiocytosis: phagocyte accumulation in multiple organs. However, little is known about the mechanism by which lysosomal nucleoside storage drives histiocytosis. Herein, histiocytosis in Slc29a3-/- mice was shown to depend on Toll-like receptor 7 (TLR7), which senses a combination of nucleosides and oligoribonucleotides (ORNs). TLR7 increased phagocyte numbers by driving the proliferation of Ly6Chi immature monocytes and their maturation into Ly6Clow phagocytes in Slc29a3-/- mice. Downstream of TLR7, FcRγ and DAP10 were required for monocyte proliferation. Histiocytosis is accompanied by inflammation in SLC29A3 disorders. However, TLR7 in nucleoside-laden splenic monocytes failed to activate inflammatory responses. Enhanced production of proinflammatory cytokines was observed only after stimulation with ssRNAs, which would increase lysosomal ORNs. Patient-derived monocytes harboring the G208R SLC29A3 mutation showed enhanced survival and proliferation in a TLR8-antagonist-sensitive manner. These results demonstrated that TLR7/8 responses to lysosomal nucleoside stress drive SLC29A3 disorders.
Collapse
Affiliation(s)
- Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masato Taoka
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mayumi Komine
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Susumu Goyama
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuji Motoi
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Jiro Kitaura
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Kumi Izawa
- Atopy Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yoshio Yamauchi
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Yumiko Tsukamoto
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsuko Fujita
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| | - Ryosuke Hiranuma
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Arinobu Tojo
- Department of Hematology and Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Mamitaro Ohtsuki
- Department of Dermatology, Jichi Medical University, Tochigi, Japan
| | - Umeharu Ohto
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiyuki Shimizu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Manabu Ozawa
- Laboratory of Developmental Genetics, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Nobuaki Yoshida
- Laboratory of Developmental Genetics, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Isobe
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Kojiro Mukai
- Department of Integrative Life Sciences, Laboratory of Organelle Pathophysiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Tomohiko Taguchi
- Department of Integrative Life Sciences, Laboratory of Organelle Pathophysiology, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Hiroaki Hemmi
- Laboratory of Immunology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier Institute—Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka, Japan
- Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Xu X, Qu S, Zhang C, Zhang M, Qin W, Ren G, Bao H, Li L, Zen K, Liu Z. CD8 T Cell-Derived Exosomal miR-186-5p Elicits Renal Inflammation via Activating Tubular TLR7/8 Signal Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301492. [PMID: 37395441 PMCID: PMC10477851 DOI: 10.1002/advs.202301492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/27/2023] [Indexed: 07/04/2023]
Abstract
T cells play an important role in the development of focal segmental glomerulosclerosis (FSGS). The mechanism underlying such T cell-based kidney disease, however, remains elusive. Here the authors report that activated CD8 T cells elicit renal inflammation and tissue injury via releasing miR-186-5p-enriched exosomes. Continuing the cohort study identifying the correlation of plasma level of miR-186-5p with proteinuria in FSGS patients, it is demonstrated that circulating miR-186-5p is mainly derived from activated CD8 T cell exosomes. Renal miR-186-5p, which is markedly increased in FSGS patients and mice with adriamycin-induced renal injury, is mainly delivered by CD8 T cell exosomes. Depleting miR-186-5p strongly attenuates adriamycin-induced mouse renal injury. Supporting the function of exosomal miR-186-5p as a key circulating pathogenic factor, intravenous injection of miR-186-5p or miR-186-5p-containing T cell exosomes results in mouse renal inflammation and tissue injury. Tracing the injected T cell exosomes shows their preferential distribution in mouse renal tubules, not glomerulus. Mechanistically, miR-186-5p directly activates renal tubular TLR7/8 signal and initiates tubular cell apoptosis. Mutating the TLR7-binding sequence on miR-186-5p or deleting mouse TLR7 largely abolishes renal tubular injuries induced by miR-186-5p or adriamycin. These findings reveal a causative role of exosomal miR-186-5p in T cell-mediated renal dysfunction.
Collapse
Affiliation(s)
- Xiaodong Xu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Shuang Qu
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Changming Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Weisong Qin
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Guisheng Ren
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Hao Bao
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| | - Limin Li
- School of Life Science and TechnologyChina Pharmaceutical University639 Longmian AvenueNanjingJiangsu211198China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical BiotechnologyNanjing University School of Life SciencesNanjingJiangsu210093China
| | - Zhihong Liu
- National Clinical Research Center of Kidney DiseasesJinling HospitalNanjing University School of MedicineNanjingJiangsu210002China
| |
Collapse
|
24
|
Shisha T, Posch MG, Lehmann J, Feifel R, Junt T, Hawtin S, Schuemann J, Avrameas A, Danekula R, Misiolek P, Siegel R, Gergely P. First-in-Human Study of the Safety, Pharmacokinetics, and Pharmacodynamics of MHV370, a Dual Inhibitor of Toll-Like Receptors 7 and 8, in Healthy Adults. Eur J Drug Metab Pharmacokinet 2023; 48:553-566. [PMID: 37532923 PMCID: PMC10480294 DOI: 10.1007/s13318-023-00847-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND AND OBJECTIVE MHV370, a dual antagonist of human Toll-like receptors (TLR) 7 and 8, suppresses cytokines and interferon-stimulated genes in vitro and in vivo, and has demonstrated efficacy in murine models of lupus. This first-in-human study aimed to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of single and multiple doses of MHV370 in healthy adults, as well as the effects of food consumption on a single dose of MHV370. METHODS This was a phase 1, randomised, placebo-controlled study conducted in three parts. In part A, participants received (3:1) a single ascending dose (SAD) of 1, 3, 10, 20, 40, 80, 160, 320, 640 and 1000 mg MHV370 or placebo. In part B, participants received (3:1) multiple ascending doses (MAD) of 25, 50, 100, 200 and 400 mg MHV370 twice daily (b.i.d) or placebo for 14 days. In part C, participants received an open-label single dose of 200 mg MHV370 under fasted or fed conditions. Safety, pharmacokinetic and pharmacodynamic parameters were evaluated. RESULTS MHV370 was well tolerated, and no safety signal was observed in the study. No dose-limiting adverse events occurred across the dose range evaluated. Plasma concentrations of MHV370 increased with dose (mean [SD] maximum plasma concentrations ranged from 0.97 [0.48] to 1670 [861.0] ng/mL for SAD of 3-1000 mg, 29.5 [7.98] to 759 [325.0] ng/mL for MAD of 25-400 mg b.i.d. on day 1). The intake of food did not have a relevant impact on the pharmacokinetics of MHV370. Pharmacodynamic data indicated time- and dose-dependent inhibition of TLR7-mediated CD69 expression on B cells (100% inhibition at 24 h post-dose starting from SAD 160 mg and MAD 50 mg b.i.d.) and TLR8-mediated TNF release after ex vivo stimulation (>90% inhibition at 24 h post-dose starting from SAD 320 mg and MAD 100 mg b.i.d.). CONCLUSION The safety, pharmacokinetic and pharmacodynamic data support the further development of MHV370 in systemic autoimmune diseases driven by the overactivation of TLR7 and TLR8.
Collapse
Affiliation(s)
- Tamas Shisha
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland.
| | | | | | - Roland Feifel
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Tobias Junt
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Stuart Hawtin
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Jens Schuemann
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Alexandre Avrameas
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Rambabu Danekula
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Patrycja Misiolek
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Richard Siegel
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| | - Peter Gergely
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, 4002, Basel, Switzerland
| |
Collapse
|
25
|
Alper P, Betschart C, André C, Boulay T, Cheng D, Deane J, Faller M, Feifel R, Glatthar R, Han D, Hemmig R, Jiang T, Knoepfel T, Maginnis J, Mutnick D, Pei W, Ruzzante G, Syka P, Zhang G, Zhang Y, Zink F, Zipfel G, Hawtin S, Junt T, Michellys PY. Discovery of the TLR7/8 Antagonist MHV370 for Treatment of Systemic Autoimmune Diseases. ACS Med Chem Lett 2023; 14:1054-1062. [PMID: 37583811 PMCID: PMC10424326 DOI: 10.1021/acsmedchemlett.3c00136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/30/2023] [Indexed: 08/17/2023] Open
Abstract
Toll-like receptor (TLR) 7 and TLR8 are endosomal sensors of the innate immune system that are activated by GU-rich single stranded RNA (ssRNA). Multiple genetic and functional lines of evidence link chronic activation of TLR7/8 to the pathogenesis of systemic autoimmune diseases (sAID) such as Sjögren's syndrome (SjS) and systemic lupus erythematosus (SLE). This makes targeting TLR7/8-induced inflammation with small-molecule inhibitors an attractive approach for the treatment of patients suffering from systemic autoimmune diseases. Here, we describe how structure-based optimization of compound 2 resulted in the discovery of 34 (MHV370, (S)-N-(4-((5-(1,6-dimethyl-1H-pyrazolo[3,4-b]pyridin-4-yl)-3-methyl-4,5,6,7-tetrahydro-1H-pyrazolo[4,3-c]pyridin-1-yl)methyl)bicyclo[2.2.2]octan-1-yl)morpholine-3-carboxamide). Its in vivo activity allows for further profiling toward clinical trials in patients with autoimmune disorders, and a Phase 2 proof of concept study of MHV370 has been initiated, testing its safety and efficacy in patients with Sjögren's syndrome and mixed connective tissue disease.
Collapse
Affiliation(s)
- Phil Alper
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Claudia Betschart
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Cédric André
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Thomas Boulay
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Dai Cheng
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Jonathan Deane
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Michael Faller
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Roland Feifel
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Ralf Glatthar
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Dong Han
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Rene Hemmig
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Tao Jiang
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Thomas Knoepfel
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Jillian Maginnis
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Daniel Mutnick
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Wei Pei
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Giulia Ruzzante
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Peter Syka
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Guobao Zhang
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Yi Zhang
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| | - Florence Zink
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Géraldine Zipfel
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Stuart Hawtin
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Tobias Junt
- Novartis
Institutes for Biomedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Pierre-Yves Michellys
- Novartis
Institutes for Biomedical Research, 10675 John Jay Hopkins Drive, San Diego, California 92121, United States
| |
Collapse
|
26
|
Kim JY, Rosenberger MG, Rutledge NS, Esser-Kahn AP. Next-Generation Adjuvants: Applying Engineering Methods to Create and Evaluate Novel Immunological Responses. Pharmaceutics 2023; 15:1687. [PMID: 37376133 PMCID: PMC10300703 DOI: 10.3390/pharmaceutics15061687] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Adjuvants are a critical component of vaccines. Adjuvants typically target receptors that activate innate immune signaling pathways. Historically, adjuvant development has been laborious and slow, but has begun to accelerate over the past decade. Current adjuvant development consists of screening for an activating molecule, formulating lead molecules with an antigen, and testing this combination in an animal model. There are very few adjuvants approved for use in vaccines, however, as new candidates often fail due to poor clinical efficacy, intolerable side effects, or formulation limitations. Here, we consider new approaches using tools from engineering to improve next-generation adjuvant discovery and development. These approaches will create new immunological outcomes that will be evaluated with novel diagnostic tools. Potential improved immunological outcomes include reduced vaccine reactogenicity, tunable adaptive responses, and enhanced adjuvant delivery. Evaluations of these outcomes can leverage computational approaches to interpret "big data" obtained from experimentation. Applying engineering concepts and solutions will provide alternative perspectives, further accelerating the field of adjuvant discovery.
Collapse
Affiliation(s)
| | | | | | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago, 5640 South Ellis Avenue, Chicago, IL 60637, USA; (J.Y.K.); (M.G.R.); (N.S.R.)
| |
Collapse
|
27
|
García-Martínez K, Chen J, Jones J, Woo A, Aucapina A, Brito I, Leifer CA. Stimulator of interferon genes is required for Toll-Like Receptor-8 induced interferon response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540812. [PMID: 37292640 PMCID: PMC10245589 DOI: 10.1101/2023.05.15.540812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The innate immune system is equipped with multiple receptors to detect microbial nucleic acids and induce type I interferon (IFN) to restrict viral replication. When dysregulated these receptor pathways induce inflammation in response to host nucleic acids and promote development and persistence of autoimmune diseases like Systemic Lupus Erythematosus (SLE). IFN production is regulated by the Interferon Regulatory Factor (IRF) transcription factor family of proteins that function downstream of several innate immune receptors such as Toll-like receptors (TLRs) and Stimulator of Interferon Genes (STING). Although both TLRs and STING activate the same downstream molecules, the pathway by which TLRs and STING activate IFN response are thought to be independent. Here we show that STING plays a previously undescribed role in human TLR8 signaling. Stimulation with the TLR8 ligands induced IFN secretion in primary human monocytes, and inhibition of STING reduced IFN secretion from primary monocytes from 8 healthy donors. We demonstrate that TLR8-induced IRF activity was reduced by STING inhibitors. Moreover, TLR8-induced IRF activity was blocked by inhibition or loss of IKKε, but not TBK1. Bulk RNA transcriptomic analysis supported a model where TLR8 induces transcriptional responses associated with SLE that can be downregulated by inhibition of STING. These data demonstrate that STING is required for full TLR8-to-IRF signaling and provide evidence for a new framework of crosstalk between cytosolic and endosomal innate immune receptors, which could be leveraged to treat IFN driven autoimmune diseases. Background High levels of type I interferon (IFN) is characteristic of multiple autoimmune diseases, and while TLR8 is associated with autoimmune disease and IFN production, the mechanisms of TLR8-induced IFN production are not fully understood. Results STING is phosphorylated following TLR8 signaling, which is selectively required for the IRF arm of TLR8 signaling and for TLR8-induced IFN production in primary human monocytes. Conclusion STING plays a previously unappreciated role in TLR8-induced IFN production. Significance Nucleic acid-sensing TLRs contribute to development and progression of autoimmune disease including interferonopathies, and we show a novel role for STING in TLR-induced IFN production that could be a therapeutic target.
Collapse
|
28
|
Hawtin S, André C, Collignon-Zipfel G, Appenzeller S, Bannert B, Baumgartner L, Beck D, Betschart C, Boulay T, Brunner HI, Ceci M, Deane J, Feifel R, Ferrero E, Kyburz D, Lafossas F, Loetscher P, Merz-Stoeckle C, Michellys P, Nuesslein-Hildesheim B, Raulf F, Rush JS, Ruzzante G, Stein T, Zaharevitz S, Wieczorek G, Siegel R, Gergely P, Shisha T, Junt T. Preclinical characterization of the Toll-like receptor 7/8 antagonist MHV370 for lupus therapy. Cell Rep Med 2023; 4:101036. [PMID: 37196635 DOI: 10.1016/j.xcrm.2023.101036] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/17/2022] [Accepted: 04/12/2023] [Indexed: 05/19/2023]
Abstract
Genetic and in vivo evidence suggests that aberrant recognition of RNA-containing autoantigens by Toll-like receptors (TLRs) 7 and 8 drives autoimmune diseases. Here we report on the preclinical characterization of MHV370, a selective oral TLR7/8 inhibitor. In vitro, MHV370 inhibits TLR7/8-dependent production of cytokines in human and mouse cells, notably interferon-α, a clinically validated driver of autoimmune diseases. Moreover, MHV370 abrogates B cell, plasmacytoid dendritic cell, monocyte, and neutrophil responses downstream of TLR7/8. In vivo, prophylactic or therapeutic administration of MHV370 blocks secretion of TLR7 responses, including cytokine secretion, B cell activation, and gene expression of, e.g., interferon-stimulated genes. In the NZB/W F1 mouse model of lupus, MHV370 halts disease. Unlike hydroxychloroquine, MHV370 potently blocks interferon responses triggered by specific immune complexes from systemic lupus erythematosus patient sera, suggesting differentiation from clinical standard of care. These data support advancement of MHV370 to an ongoing phase 2 clinical trial.
Collapse
Affiliation(s)
- Stuart Hawtin
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Cédric André
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | | | - Simone Appenzeller
- Department of Orthopedics, Rheumatology, and Traumatology, School of Medical Science, University of Campinas (UNICAMP), Campinas, 13083-887 São Paulo, Brazil
| | - Bettina Bannert
- Department of Rheumatology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Lea Baumgartner
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Damian Beck
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Claudia Betschart
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Thomas Boulay
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Hermine I Brunner
- Division of Rheumatology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Melanie Ceci
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Jonathan Deane
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | - Roland Feifel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Enrico Ferrero
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Diego Kyburz
- Department of Rheumatology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Frederique Lafossas
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Pius Loetscher
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | | | - Pierre Michellys
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | | | - Friedrich Raulf
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - James S Rush
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Giulia Ruzzante
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Thomas Stein
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Samantha Zaharevitz
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, La Jolla, CA 92121, USA
| | - Grazyna Wieczorek
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Richard Siegel
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Peter Gergely
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Tamas Shisha
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Tobias Junt
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland.
| |
Collapse
|
29
|
The link between rheumatic disorders and inborn errors of immunity. EBioMedicine 2023; 90:104501. [PMID: 36870198 PMCID: PMC9996386 DOI: 10.1016/j.ebiom.2023.104501] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/11/2022] [Accepted: 02/10/2023] [Indexed: 03/06/2023] Open
Abstract
Inborn errors of immunity (IEIs) are immunological disorders characterized by variable susceptibility to infections, immune dysregulation and/or malignancies, as a consequence of damaging germline variants in single genes. Though initially identified among patients with unusual, severe or recurrent infections, non-infectious manifestations and especially immune dysregulation in the form of autoimmunity or autoinflammation can be the first or dominant phenotypic aspect of IEIs. An increasing number of IEIs causing autoimmunity or autoinflammation, including rheumatic disease have been reported over the last decade. Despite their rarity, identification of those disorders provided insight into the pathomechanisms of immune dysregulation, which may be relevant for understanding the pathogenesis of systemic rheumatic disorders. In this review, we present novel IEIs primarily causing autoimmunity or autoinflammation along with their pathogenic mechanisms. In addition, we explore the likely pathophysiological and clinical relevance of IEIs in systemic rheumatic disorders.
Collapse
|
30
|
O'Reilly S. Toll-like receptor triggering in systemic sclerosis: time to target. Rheumatology (Oxford) 2023; 62:SI12-SI19. [PMID: 35863054 DOI: 10.1093/rheumatology/keac421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/04/2022] [Accepted: 07/08/2022] [Indexed: 02/07/2023] Open
Abstract
SSc is an autoimmune disease that has features of vascular abnormalities, inflammation and skin and lung fibrosis. Toll-like receptors (TLRs) are sentinel receptors that serve to recognize pathogens or internal danger signals leading to downstream signalling pathways that ultimately lead to inflammation and modification of adaptive immunity. Inflammation and fibrosis appear intricately connected in this disease and TLR ligation on fibroblasts can directly activate these cells to produce copious amounts of collagen, a hallmark of disease. The presence of damage-associated molecular patterns in association with fibrosis has been highlighted. Given their prominent role in disease, this review discusses the evidence of their expression and role in disease pathogenesis and possible therapeutic intervention to mitigate fibrosis.
Collapse
|
31
|
Leśniak M, Lipniarska J, Majka P, Kopyt W, Lejman M, Zawitkowska J. The Role of TRL7/8 Agonists in Cancer Therapy, with Special Emphasis on Hematologic Malignancies. Vaccines (Basel) 2023; 11:vaccines11020277. [PMID: 36851155 PMCID: PMC9967151 DOI: 10.3390/vaccines11020277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Toll-like receptors (TLR) belong to the pattern recognition receptors (PRR). TLR7 and the closely correlated TLR8 affiliate with toll-like receptors family, are located in endosomes. They recognize single-stranded ribonucleic acid (RNA) molecules and synthetic deoxyribonucleic acid (DNA)/RNA analogs-oligoribonucleotides. TLRs are primarily expressed in hematopoietic cells. There is compiling evidence implying that TLRs also direct the formation of blood cellular components and make a contribution to the pathogenesis of certain hematopoietic malignancies. The latest research shows a positive effect of therapy with TRL agonists on the course of hemato-oncological diseases. Ligands impact activation of antigen-presenting cells which results in production of cytokines, transfer of mentioned cells to the lymphoid tissue and co-stimulatory surface molecules expression required for T-cell activation. Toll-like receptor agonists have already been used in oncology especially in the treatment of dermatological neoplastic lesions. The usage of these substances in the treatment of solid tumors is being investigated. The present review discusses the direct and indirect influence that TLR7/8 agonists, such as imiquimod, imidazoquinolines and resiquimod have on neoplastic cells and their promising role as adjuvants in anticancer vaccines.
Collapse
Affiliation(s)
- Maria Leśniak
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Justyna Lipniarska
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Patrycja Majka
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Weronika Kopyt
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence:
| |
Collapse
|
32
|
Zsurka G, Appel MLT, Nastaly M, Hallmann K, Hansen N, Nass D, Baumgartner T, Surges R, Hartmann G, Bartok E, Kunz WS. Loss of the Immunomodulatory Transcription Factor BATF2 in Humans Is Associated with a Neurological Phenotype. Cells 2023; 12:227. [PMID: 36672163 PMCID: PMC9856319 DOI: 10.3390/cells12020227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/21/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Epilepsy and mental retardation are known to be associated with pathogenic mutations in a broad range of genes that are expressed in the brain and have a role in neurodevelopment. Here, we report on a family with three affected individuals whose clinical symptoms closely resemble a neurodevelopmental disorder. Whole-exome sequencing identified a homozygous stop-gain mutation, p.Gln19*, in the BATF2 gene in the patients. The BATF2 transcription factor is predominantly expressed in macrophages and monocytes and has been reported to modulate AP-1 transcription factor-mediated pro-inflammatory responses. Transcriptome analysis showed altered base-level expression of interferon-stimulated genes in the patients' blood, typical for type I interferonopathies. Peripheral blood mononuclear cells from all three patients demonstrated elevated responses to innate immune stimuli, which could be reproduced in CRISPR-Cas9-generated BATF2-/- human monocytic cell lines. BATF2 is, therefore, a novel disease-associated gene candidate for severe epilepsy and mental retardation related to dysregulation of immune responses, which underscores the relevance of neuroinflammation for epilepsy.
Collapse
Affiliation(s)
- Gábor Zsurka
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Maximilian L. T. Appel
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Institute of Experimental Haematology and Transfusion Medicine, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Maximilian Nastaly
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Kerstin Hallmann
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Niels Hansen
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Daniel Nass
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Tobias Baumgartner
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Rainer Surges
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University of Bonn, 53127 Bonn, Germany
| | - Eva Bartok
- Institute of Experimental Haematology and Transfusion Medicine, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, 2000 Antwerp, Belgium
| | - Wolfram S. Kunz
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, 53127 Bonn, Germany
- Department of Epileptology, University Hospital Bonn, 53127 Bonn, Germany
| |
Collapse
|
33
|
Liu S, Yang X. Intestinal flora plays a role in the progression of hepatitis-cirrhosis-liver cancer. Front Cell Infect Microbiol 2023; 13:1140126. [PMID: 36968098 PMCID: PMC10034054 DOI: 10.3389/fcimb.2023.1140126] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
The liver is a vital metabolism and detoxification organ of human body, which is involved in the biotransformation and metabolism of the organism. Hepatitis - cirrhosis - liver cancer are significant and common part of liver diseases. The pathogenesis of liver diseases is generally as followed: inflammation and other pathogenic factors cause persistent damage to the liver, leading to the activation of hepatic stellate cells (HSCs) and excessive deposition of extracellular matrix. Patients with chronic hepatitis have a high risk of developing into liver fibrosis, cirrhosis, and even life-threatening liver cancer, which poses a great threat to public health.As the first organ to come into contact with blood from the gut, the liver is profoundly affected by the intestinal flora and its metabolites, with leaky gut and flora imbalance being the triggers of the liver's pathological response. So far, no one has reviewed the role of intestinal flora in this process from the perspective of the progression of hepatitis-cirrhosis-liver cancer and this article reviews the evidence supporting the effect of intestinal flora in the progression of liver disease.
Collapse
|
34
|
Bolouri H, Ries RE, Wiedeman AE, Hylkema T, Scheiding S, Gersuk VH, O'Brien K, Nguyen QA, Smith JL, Alice Long S, Meshinchi S. Inflammatory bone marrow signaling in pediatric acute myeloid leukemia distinguishes patients with poor outcomes. Nat Commun 2022; 13:7186. [PMID: 36418348 PMCID: PMC9684530 DOI: 10.1038/s41467-022-34965-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
High levels of the inflammatory cytokine IL-6 in the bone marrow are associated with poor outcomes in pediatric acute myeloid leukemia (pAML), but its etiology remains unknown. Using RNA-seq data from pre-treatment bone marrows of 1489 children with pAML, we show that > 20% of patients have concurrent IL-6, IL-1, IFNα/β, and TNFα signaling activity and poorer outcomes. Targeted sequencing of pre-treatment bone marrow samples from affected patients (n = 181) revealed 5 highly recurrent patterns of somatic mutation. Using differential expression analyses of the most common genomic subtypes (~60% of total), we identify high expression of multiple potential drivers of inflammation-related treatment resistance. Regardless of genomic subtype, we show that JAK1/2 inhibition reduces receptor-mediated inflammatory signaling by leukemic cells in-vitro. The large number of high-risk pAML genomic subtypes presents an obstacle to the development of mutation-specific therapies. Our findings suggest that therapies targeting inflammatory signaling may be effective across multiple genomic subtypes of pAML.
Collapse
Affiliation(s)
- Hamid Bolouri
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA.
| | - Rhonda E Ries
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Alice E Wiedeman
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Tiffany Hylkema
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
| | - Sheila Scheiding
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Vivian H Gersuk
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Kimberly O'Brien
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Quynh-Anh Nguyen
- Center for Systems Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Jenny L Smith
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA
- Research Scientific Computing, Seattle Children's Research Institute, 818 Stewart Street, Seattle, WA, USA
| | - S Alice Long
- Center for Translational Immunology, Benaroya Research Institute, 1201 9th Ave, Seattle, WA, USA
| | - Soheil Meshinchi
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA, USA.
| |
Collapse
|
35
|
Alexopoulou L. Nucleic acid-sensing toll-like receptors: Important players in Sjögren’s syndrome. Front Immunol 2022; 13:980400. [PMID: 36389822 PMCID: PMC9659959 DOI: 10.3389/fimmu.2022.980400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/12/2022] [Indexed: 11/30/2022] Open
Abstract
Sjögren’s syndrome (SS) is a chronic systemic autoimmune disease that affects the salivary and lacrimal glands, as well as other organ systems like the lungs, kidneys and nervous system. SS can occur alone or in combination with another autoimmune disease, such as systemic lupus erythematosus (SLE) or rheumatoid arthritis. The etiology of SS is unknown but recent studies have revealed the implication of the activation of innate immune receptors, including Toll-like receptors (TLRs), mainly through the detection of endogenous nucleic acids, in the pathogenesis of systemic autoimmune diseases. Studies on SS mouse models suggest that TLRs and especially TLR7 that detects single-stranded RNA of microbial or endogenous origin can drive the development of SS and findings in SS patients corroborate those in mouse models. In this review, we will give an overview of the function and signaling of nucleic acid-sensing TLRs, the interplay of TLR7 with TLR8 and TLR9 in the context of autoimmunity, summarize the evidence for the critical role of TLR7 in the pathogenesis of SS and present a possible connection between SARS-CoV-2 and SS.
Collapse
|
36
|
Liu ZM, Yang MH, Yu K, Lian ZX, Deng SL. Toll-like receptor (TLRs) agonists and antagonists for COVID-19 treatments. Front Pharmacol 2022; 13:989664. [PMID: 36188605 PMCID: PMC9518217 DOI: 10.3389/fphar.2022.989664] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) rapidly infects humans and animals which make coronavirus disease 2019 (COVID-19) a grievous epidemic worldwide which broke out in 2020. According to data analysis of the other coronavirus family, for instance severe acute respiratory syndrome SARS coronavirus (SARS-CoV), can provide experience for the mutation of SARS-CoV-2 and the prevention and treatment of COVID-19. Toll-like receptors (TLRs) as a pattern recognition receptor (PRRs), have an indispensable function in identifying the invader even activate the innate immune system. It is possible for organism to activate different TLR pathways which leads to secretion of proinflammatory cytokines such as Interleukin 1 (IL-1), Interleukin 6 (IL-6), Tumor necrosis factor α (TNFα) and type Ⅰ interferon. As a component of non-specific immunity, TLRs pathway may participate in the SARS-CoV-2 pathogenic processes, due to previous works have proved that TLRs are involved in the invasion and infection of SARS-CoV and MERS to varying degrees. Different TLR, such as TLR2, TLR4, TLR7, TLR8 and TLR9 probably have a double-sided in COVID-19 infection. Therefore, it is of great significance for a correctly acknowledging how TLR take part in the SARS-CoV-2 pathogenic processes, which will be the development of treatment and prevention strategies.
Collapse
Affiliation(s)
- Zhi-Mei Liu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ming-Hui Yang
- Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing, China
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| | - Shou-Long Deng
- NHC Key Laboratory of Human Disease Comparative Medicine, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
- *Correspondence: Zheng-Xing Lian, ; Shou-Long Deng,
| |
Collapse
|
37
|
Abt ER, Rashid K, Le TM, Li S, Lee HR, Lok V, Li L, Creech AL, Labora AN, Mandl HK, Lam AK, Cho A, Rezek V, Wu N, Abril-Rodriguez G, Rosser EW, Mittelman SD, Hugo W, Mehrling T, Bantia S, Ribas A, Donahue TR, Crooks GM, Wu TT, Radu CG. Purine nucleoside phosphorylase enables dual metabolic checkpoints that prevent T cell immunodeficiency and TLR7-associated autoimmunity. J Clin Invest 2022; 132:e160852. [PMID: 35653193 PMCID: PMC9374381 DOI: 10.1172/jci160852] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/31/2022] [Indexed: 01/27/2023] Open
Abstract
Purine nucleoside phosphorylase (PNP) enables the breakdown and recycling of guanine nucleosides. PNP insufficiency in humans is paradoxically associated with both immunodeficiency and autoimmunity, but the mechanistic basis for these outcomes is incompletely understood. Here, we identify two immune lineage-dependent consequences of PNP inactivation dictated by distinct gene interactions. During T cell development, PNP inactivation is synthetically lethal with downregulation of the dNTP triphosphohydrolase SAMHD1. This interaction requires deoxycytidine kinase activity and is antagonized by microenvironmental deoxycytidine. In B lymphocytes and macrophages, PNP regulates Toll-like receptor 7 signaling by controlling the levels of its (deoxy)guanosine nucleoside ligands. Overriding this regulatory mechanism promotes germinal center formation in the absence of exogenous antigen and accelerates disease in a mouse model of autoimmunity. This work reveals that one purine metabolism gene protects against immunodeficiency and autoimmunity via independent mechanisms operating in distinct immune lineages and identifies PNP as a potentially novel metabolic immune checkpoint.
Collapse
Affiliation(s)
- Evan R. Abt
- Department of Molecular and Medical Pharmacology and
| | - Khalid Rashid
- Department of Molecular and Medical Pharmacology and
| | - Thuc M. Le
- Department of Molecular and Medical Pharmacology and
| | - Suwen Li
- Department of Molecular and Medical Pharmacology and
| | - Hailey R. Lee
- Department of Molecular and Medical Pharmacology and
| | - Vincent Lok
- Department of Molecular and Medical Pharmacology and
| | - Luyi Li
- Department of Surgery, UCLA, Los Angeles, California, USA
| | | | | | - Hanna K. Mandl
- Department of Surgery, UCLA, Los Angeles, California, USA
| | - Alex K. Lam
- Department of Molecular and Medical Pharmacology and
| | - Arthur Cho
- Department of Nuclear Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | | | - Nanping Wu
- Department of Surgery, UCLA, Los Angeles, California, USA
| | | | | | - Steven D. Mittelman
- Division of Pediatric Endocrinology, UCLA Children’s Discovery and Innovation Institute, and
| | - Willy Hugo
- Division of Dermatology, Department of Medicine, UCLA, Los Angeles, California, USA
| | | | | | - Antoni Ribas
- Department of Molecular and Medical Pharmacology and
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Division of Hematology/Oncology, Department of Medicine
- Division of Surgical Oncology, Department of Surgery
- Jonsson Comprehensive Cancer Center
| | - Timothy R. Donahue
- Department of Molecular and Medical Pharmacology and
- Department of Surgery, UCLA, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center
| | - Gay M. Crooks
- Division of Pediatric Hematology-Oncology, Department of Pediatrics
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, and
- Department of Pathology and Laboratory Medicine, UCLA, Los Angeles, California, USA
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology and
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology and
- Jonsson Comprehensive Cancer Center
| |
Collapse
|
38
|
Miyake K, Shibata T, Fukui R, Sato R, Saitoh SI, Murakami Y. Nucleic Acid Sensing by Toll-Like Receptors in the Endosomal Compartment. Front Immunol 2022; 13:941931. [PMID: 35812450 PMCID: PMC9259784 DOI: 10.3389/fimmu.2022.941931] [Citation(s) in RCA: 12] [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: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptors (TLRs) respond to pathogen constituents, such as microbial lipids and nucleic acids (NAs). TLRs recognize NAs in endosomal compartments. Structural and functional studies have shown that recognition of NAs by TLRs depends on NA processing by RNases and DNases. DNase II-dependent DNA degradation is required for TLR9 responses to single-stranded DNAs, whereas RNase T2-dependent RNA degradation enables TLR7 and TLR8 to respond to nucleosides and oligoribonucleotides. In contrast, RNases and DNases negatively regulate TLR responses by degrading their ligands. RNase T2 negatively regulates TLR3 responses to degrading the TLR3 ligand double-stranded RNAs. Therefore, NA metabolism in the endosomal compartments affects the endosomal TLR responses. Dysregulation of NA metabolism in the endosomal compartment drives the TLR-dependent pathologies in human diseases.
Collapse
Affiliation(s)
- Kensuke Miyake
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
- *Correspondence: Kensuke Miyake,
| | - Takuma Shibata
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryota Sato
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Shin-Ichiroh Saitoh
- Division of Innate Immunity, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Yusuke Murakami
- Faculty of Pharmacy, Department of Pharmaceutical Sciences and Research Institute of Pharmaceutical Sciences, Musashino University, Tokyo, Japan
| |
Collapse
|
39
|
Nilsen KE, Skjesol A, Frengen Kojen J, Espevik T, Stenvik J, Yurchenko M. TIRAP/Mal Positively Regulates TLR8-Mediated Signaling via IRF5 in Human Cells. Biomedicines 2022; 10:biomedicines10071476. [PMID: 35884781 PMCID: PMC9312982 DOI: 10.3390/biomedicines10071476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/09/2022] [Accepted: 06/17/2022] [Indexed: 11/21/2022] Open
Abstract
Toll-like receptor 8 (TLR8) recognizes single-stranded RNA of viral and bacterial origin as well as mediates the secretion of pro-inflammatory cytokines and type I interferons by human monocytes and macrophages. TLR8, as other endosomal TLRs, utilizes the MyD88 adaptor protein for initiation of signaling from endosomes. Here, we addressed the potential role of the Toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) in the regulation of TLR8 signaling in human primary monocyte-derived macrophages (MDMs). To accomplish this, we performed TIRAP gene silencing, followed by the stimulation of cells with synthetic ligands or live bacteria. Cytokine-gene expression and secretion were analyzed by quantitative PCR or Bioplex assays, respectively, while nuclear translocation of transcription factors was addressed by immunofluorescence and imaging, as well as by cell fractionation and immunoblotting. Immunoprecipitation and Akt inhibitors were also used to dissect the signaling mechanisms. Overall, we show that TIRAP is recruited to the TLR8 Myddosome signaling complex, where TIRAP contributes to Akt-kinase activation and the nuclear translocation of interferon regulatory factor 5 (IRF5). Recruitment of TIRAP to the TLR8 signaling complex promotes the expression and secretion of the IRF5-dependent cytokines IFNβ and IL-12p70 as well as, to a lesser degree, TNF. These findings reveal a new and unconventional role of TIRAP in innate immune defense.
Collapse
Affiliation(s)
- Kaja Elisabeth Nilsen
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Astrid Skjesol
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - June Frengen Kojen
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Terje Espevik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Jørgen Stenvik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Infectious Diseases, Clinic of Medicine, St. Olavs Hospital HF, Trondheim University Hospital, NO-7006 Trondheim, Norway
| | - Maria Yurchenko
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (K.E.N.); (A.S.); (J.F.K.); (T.E.); (J.S.)
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
- Department of Infectious Diseases, Clinic of Medicine, St. Olavs Hospital HF, Trondheim University Hospital, NO-7006 Trondheim, Norway
- Correspondence:
| |
Collapse
|
40
|
Yang C, Bachu M, Du Y, Brauner C, Yuan R, Ah Kioon MD, Chesi G, Barrat FJ, Ivashkiv LB. CXCL4 synergizes with TLR8 for TBK1-IRF5 activation, epigenomic remodeling and inflammatory response in human monocytes. Nat Commun 2022; 13:3426. [PMID: 35701499 PMCID: PMC9195402 DOI: 10.1038/s41467-022-31132-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 06/06/2022] [Indexed: 01/11/2023] Open
Abstract
Regulation of endosomal Toll-like receptor (TLR) responses by the chemokine CXCL4 is implicated in inflammatory and fibrotic diseases, with CXCL4 proposed to potentiate TLR responses by binding to nucleic acid TLR ligands and facilitating their endosomal delivery. Here we report that in human monocytes/macrophages, CXCL4 initiates signaling cascades and downstream epigenomic reprogramming that change the profile of the TLR8 response by selectively amplifying inflammatory gene transcription and interleukin (IL)-1β production, while partially attenuating the interferon response. Mechanistically, costimulation by CXCL4 and TLR8 synergistically activates TBK1 and IKKε, repurposes these kinases towards an inflammatory response via coupling with IRF5, and activates the NLRP3 inflammasome. CXCL4 signaling, in a cooperative and synergistic manner with TLR8, induces chromatin remodeling and activates de novo enhancers associated with inflammatory genes. Our findings thus identify new regulatory mechanisms of TLR responses relevant for cytokine storm, and suggest targeting the TBK1-IKKε-IRF5 axis may be beneficial in inflammatory diseases.
Collapse
Affiliation(s)
- Chao Yang
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Mahesh Bachu
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Caroline Brauner
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Ruoxi Yuan
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Giancarlo Chesi
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
| | - Franck J Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
| | - Lionel B Ivashkiv
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
41
|
Sreekantha RK, Mussari CP, Dodd DS, Pasunoori L, Hegde S, Posy SL, Critton D, Ruepp S, Subramanian M, Salter-Cid LM, Tagore DM, Sarodaya S, Dudhgaonkar S, Poss MA, Schieven GL, Carter PH, Macor JE, Dyckman AJ. Identification of 2-Pyridinylindole-Based Dual Antagonists of Toll-like Receptors 7 and 8 (TLR7/8). ACS Med Chem Lett 2022; 13:812-818. [PMID: 35586440 PMCID: PMC9109266 DOI: 10.1021/acsmedchemlett.2c00049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/20/2022] [Indexed: 11/30/2022] Open
Abstract
The toll-like receptors (TLRs) play key roles in activation of the innate immune system. Aberrant activation of TLR7 and TLR8 pathways can occur in the context of autoimmune disorders due to the elevated presence and recognition of self-RNA as activating ligands. Control of this unintended activation via inhibition of TLR7/8 signaling holds promise for the treatment of diseases such as psoriasis, arthritis, and lupus. Optimization of a 2-pyridinylindole series of compounds led to the identification of potent dual inhibitors of TLR7 and TLR8, which demonstrated good selectivity against TLR9 and other family members. The in vitro characterization and in vivo evaluation in rodent pharmacokinetic/pharmacodynamic and efficacy studies of BMS-905 is detailed, along with structural information obtained through X-ray cocrystallographic studies.
Collapse
Affiliation(s)
| | - Christopher P. Mussari
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Dharmpal S. Dodd
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Laxman Pasunoori
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Subramanya Hegde
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Shana L. Posy
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - David Critton
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Stefan Ruepp
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Murali Subramanian
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | - Luisa M. Salter-Cid
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | | | - Sanket Sarodaya
- Biocon Bristol Myers Squibb Research Center (BBRC), Bangalore 560099, India
| | | | - Michael A. Poss
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Gary L. Schieven
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Percy H. Carter
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - John E. Macor
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| | - Alaric J. Dyckman
- Bristol Myers Squibb Research and Development, P.O. Box 4000, Princeton, New Jersey 08543, United States
| |
Collapse
|
42
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022. [PMID: 35309296 DOI: 10.3389/fimmu.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
43
|
Betschart C, Faller M, Zink F, Hemmig R, Blank J, Vangrevelinghe E, Bourrel M, Glatthar R, Behnke D, Barker K, Heizmann A, Angst D, Nimsgern P, Jacquier S, Junt T, Zipfel G, Ruzzante G, Loetscher P, Limonta S, Hawtin S, Andre CB, Boulay T, Feifel R, Knoepfel T. Structure-Based Optimization of a Fragment-like TLR8 Binding Screening Hit to an In Vivo Efficacious TLR7/8 Antagonist. ACS Med Chem Lett 2022; 13:658-664. [PMID: 35450354 PMCID: PMC9014506 DOI: 10.1021/acsmedchemlett.1c00696] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/08/2022] [Indexed: 11/28/2022] Open
Abstract
Inappropriate activation of TLR7 and TLR8 is linked to several autoimmune diseases, such as lupus erythematosus. Here we report on the efficient structure-based optimization of the inhibition of TLR8, starting from a co-crystal structure of a small screening hit. Further optimization of the physicochemical properties for cellular potency and expansion of the structure-activity relationship for dual potency finally resulted in a highly potent TLR7/8 antagonist with demonstrated in vivo efficacy after oral dosing.
Collapse
Affiliation(s)
- Claudia Betschart
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Michael Faller
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Florence Zink
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - René Hemmig
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Jutta Blank
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Eric Vangrevelinghe
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Marjorie Bourrel
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Ralf Glatthar
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Dirk Behnke
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Kerstin Barker
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Andreas Heizmann
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Daniela Angst
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Pierre Nimsgern
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Sébastien Jacquier
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Tobias Junt
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Géraldine Zipfel
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Giulia Ruzzante
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Pius Loetscher
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Sarah Limonta
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Stuart Hawtin
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Cedric Bernard Andre
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Thomas Boulay
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Roland Feifel
- Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| | - Thomas Knoepfel
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, CH-4002 Basel, Switzerland
| |
Collapse
|
44
|
Gerussi A, Caime C, Binatti E, Cristoferi L, Asselta R, Gershwin EM, Invernizzi P. X marks the spot in autoimmunity. Expert Rev Clin Immunol 2022; 18:429-437. [PMID: 35349778 DOI: 10.1080/1744666x.2022.2060203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Autoimmune diseases mostly affect females. Besides hormones, several factors related to chromosome X have been called in action to explain this sex predominance. AREAS COVERED This paper provides an overview on the role of chromosome X (chrX) in explaining why females have higher susceptibility to autoimmunity. The work outlines some essential concepts regarding chrX inactivation, escape from chrX inactivation and the evolutionary history of chrX. In addition, we will discuss the concept of gene escape in immune cells, with examples related to specific X-linked genes and autoimmune diseases. EXPERT OPINION There is growing evidence that many genes present on chrX escape inactivation, and some of them have significant immune-mediated functions. In immune cells of female individuals the escape of these genes is not constant, but the knowledge of the mechanisms controlling this plasticity are not completely understood. Future studies aimed at the characterization of these modifications at single-cell resolution, together with conformational 3D studies of the inactive X chromosome, will hopefully help to fill this gap of knowledge.
Collapse
Affiliation(s)
- Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Chiara Caime
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Eleonora Binatti
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Laura Cristoferi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy.,Humanitas Clinical and Research Center, IRCCS, Rozzano, Italy
| | - Eric M Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, Davis, CA, USA
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.,European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| |
Collapse
|
45
|
Bjornson-Hooper ZB, Fragiadakis GK, Spitzer MH, Chen H, Madhireddy D, Hu K, Lundsten K, McIlwain DR, Nolan GP. A Comprehensive Atlas of Immunological Differences Between Humans, Mice, and Non-Human Primates. Front Immunol 2022; 13:867015. [PMID: 35359965 PMCID: PMC8962947 DOI: 10.3389/fimmu.2022.867015] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/16/2022] [Indexed: 01/01/2023] Open
Abstract
Animal models are an integral part of the drug development and evaluation process. However, they are unsurprisingly imperfect reflections of humans, and the extent and nature of many immunological differences are unknown. With the rise of targeted and biological therapeutics, it is increasingly important that we understand the molecular differences in the immunological behavior of humans and model organisms. However, very few antibodies are raised against non-human primate antigens, and databases of cross-reactivity between species are incomplete. Thus, we screened 332 antibodies in five immune cell populations in blood from humans and four non-human primate species generating a comprehensive cross-reactivity catalog that includes cell type-specificity. We used this catalog to create large mass cytometry universal cross-species phenotyping and signaling panels for humans, along with three of the model organisms most similar to humans: rhesus and cynomolgus macaques and African green monkeys; and one of the mammalian models most widely used in drug development: C57BL/6 mice. As a proof-of-principle, we measured immune cell signaling responses across all five species to an array of 15 stimuli using mass cytometry. We found numerous instances of different cellular phenotypes and immune signaling events occurring within and between species, and detailed three examples (double-positive T cell frequency and signaling; granulocyte response to Bacillus anthracis antigen; and B cell subsets). We also explore the correlation of herpes simian B virus serostatus on the immune profile. Antibody panels and the full dataset generated are available online as a resource to enable future studies comparing immune responses across species during the evaluation of therapeutics.
Collapse
Affiliation(s)
| | - Gabriela K. Fragiadakis
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
- Department of Medicine, Division of Rheumatology, University of California San Francisco, San Francisco, CA, United States
- Bakar ImmunoX Initiative, University of California San Francisco, San Francisco, CA, United States
- University of California, San Francisco (UCSF) Data Science CoLab and University of California, San Francisco (UCSF) Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Matthew H. Spitzer
- Immunology Program, Stanford University, Stanford, CA, United States
- Departments of Otolaryngology – Head and Neck Surgery and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, United States
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
| | - Han Chen
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Deepthi Madhireddy
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kevin Hu
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Kelly Lundsten
- BioLegend Inc, Advanced Cytometry, San Diego, CA, United States
| | - David R. McIlwain
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| | - Garry P. Nolan
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, United States
| |
Collapse
|
46
|
Duan T, Du Y, Xing C, Wang HY, Wang RF. Toll-Like Receptor Signaling and Its Role in Cell-Mediated Immunity. Front Immunol 2022; 13:812774. [PMID: 35309296 PMCID: PMC8927970 DOI: 10.3389/fimmu.2022.812774] [Citation(s) in RCA: 418] [Impact Index Per Article: 139.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
Innate immunity is the first defense system against invading pathogens. Toll-like receptors (TLRs) are well-defined pattern recognition receptors responsible for pathogen recognition and induction of innate immune responses. Since their discovery, TLRs have revolutionized the field of immunology by filling the gap between the initial recognition of pathogens by innate immune cells and the activation of the adaptive immune response. TLRs critically link innate immunity to adaptive immunity by regulating the activation of antigen-presenting cells and key cytokines. Furthermore, recent studies also have shown that TLR signaling can directly regulate the T cell activation, growth, differentiation, development, and function under diverse physiological conditions. This review provides an overview of TLR signaling pathways and their regulators and discusses how TLR signaling, directly and indirectly, regulates cell-mediated immunity. In addition, we also discuss how TLR signaling is critically important in the host's defense against infectious diseases, autoimmune diseases, and cancer.
Collapse
Affiliation(s)
- Tianhao Duan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Yang Du
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Changsheng Xing
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Helen Y. Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Rong-Fu Wang
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Department of Pediatrics, Children’s Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
47
|
Fejtkova M, Sukova M, Hlozkova K, Skvarova Kramarzova K, Rackova M, Jakubec D, Bakardjieva M, Bloomfield M, Klocperk A, Parackova Z, Sediva A, Aluri J, Novakova M, Kalina T, Fronkova E, Hrusak O, Malcova H, Sedlacek P, Liba Z, Kudr M, Stary J, Cooper MA, Svaton M, Kanderova V. TLR8/TLR7 dysregulation due to a novel TLR8 mutation causes severe autoimmune hemolytic anemia and autoinflammation in identical twins. Am J Hematol 2022; 97:338-351. [PMID: 34981838 DOI: 10.1002/ajh.26452] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022]
Abstract
Our study presents a novel germline c.1715G>T (p.G572V) mutation in the gene encoding Toll-like receptor 8 (TLR8) causing an autoimmune and autoinflammatory disorder in a family with monozygotic male twins, who suffer from severe autoimmune hemolytic anemia worsening with infections, and autoinflammation presenting as fevers, enteritis, arthritis, and CNS vasculitis. The pathogenicity of the mutation was confirmed by in vitro assays on transfected cell lines and primary cells. The p.G572V mutation causes impaired stability of the TLR8 protein, cross-reactivity to TLR7 ligands and reduced ability of TLR8 to attenuate TLR7 signaling. This imbalance toward TLR7-dependent signaling leads to increased pro-inflammatory responses, such as nuclear factor-κB (NF-κB) activation and production of pro-inflammatory cytokines IL-1β, IL-6, and TNFα. This unique TLR8 mutation with partial TLR8 protein loss and hyperinflammatory phenotype mediated by TLR7 ligands represents a novel inborn error of immunity with childhood-onset and a good response to TLR7 inhibition.
Collapse
Affiliation(s)
- Martina Fejtkova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Martina Sukova
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Katerina Hlozkova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Karolina Skvarova Kramarzova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Marketa Rackova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - David Jakubec
- Bioinformatics Group, Institute of Organic Chemistry and Biochemistry Czech Academy of Sciences Prague Czech Republic
- Department of Software Engineering, Faculty of Mathematics and Physics Charles University Prague Czech Republic
| | - Marina Bakardjieva
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Marketa Bloomfield
- Department of Paediatrics, First Faculty of Medicine Charles University and Thomayer University Hospital Prague Czech Republic
- Department of Immunology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Adam Klocperk
- Department of Immunology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Zuzana Parackova
- Department of Immunology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Anna Sediva
- Department of Immunology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Jahnavi Aluri
- Division of Rheumatology/Immunology, Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
| | - Michaela Novakova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Tomas Kalina
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Eva Fronkova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Ondrej Hrusak
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Hana Malcova
- Department of Paediatric and Adult Rheumatology University Hospital Motol Prague Czech Republic
| | - Petr Sedlacek
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Zuzana Liba
- Department of Paediatric Neurology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Martin Kudr
- Department of Paediatric Neurology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Jan Stary
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Megan A. Cooper
- Division of Rheumatology/Immunology, Department of Pediatrics Washington University School of Medicine St. Louis Missouri USA
| | - Michael Svaton
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| | - Veronika Kanderova
- CLIP ‐ Childhood Leukaemia Investigation Prague, Department of Paediatric Haematology and Oncology, Second Faculty of Medicine Charles University and University Hospital Motol Prague Czech Republic
| |
Collapse
|
48
|
Zhao J, Zhang X, Guan J, Su Y, Jiang J. Identification of key biomarkers in steroid-induced osteonecrosis of the femoral head and their correlation with immune infiltration by bioinformatics analysis. BMC Musculoskelet Disord 2022; 23:67. [PMID: 35042504 PMCID: PMC8767711 DOI: 10.1186/s12891-022-04994-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/28/2021] [Indexed: 11/17/2022] Open
Abstract
Objective This study aimed to identify key diagnostic markers and immune infiltration of (SONFH) by bioinformatics analysis. Methods Related SONFH datasets were downloaded from the Gene Expression Omnibus (GEO) database. First, we identified the differentially expressed genes (DEGs) and performed the functional enrichment analysis. Then weighted correlation network analysis (WGCNA) and the MCODE plug-in in Cytoscape were used to identify the diagnostic markers of SONFH. Finally, CIBERSORT was used to analyze the immune infiltration between SONFH and healthy controls, and the correlation between infiltrating immune cells and diagnostic markers was analyzed. Results TYROBP, TLR2, P2RY13, TLR8, HCK, MNDA, and NCF2 may be key diagnostic markers of SONFH. Immune cell infiltration analysis revealed that Memory B cells and activated dendritic cells may be related to the SONFH process. Moreover, HCK was negatively correlated with CD8 T cells, and neutrophils were positively correlated with those key diagnostic markers. Conclusions TYROBP, TLR2, P2RY13, TLR8, HCK, MNDA, and NCF2 may be used as diagnostic markers of SONFH, and immune-related mechanism of SONFH and the potential immunotherapy are worthy of further study. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-022-04994-7.
Collapse
|
49
|
Insights into Modern Therapeutic Approaches in Pediatric Acute Leukemias. Cells 2022; 11:cells11010139. [PMID: 35011701 PMCID: PMC8749975 DOI: 10.3390/cells11010139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 12/07/2021] [Accepted: 12/15/2021] [Indexed: 02/01/2023] Open
Abstract
Pediatric cancers predominantly constitute lymphomas and leukemias. Recently, our knowledge and awareness about genetic diversities, and their consequences in these diseases, have greatly expanded. Modern solutions are focused on mobilizing and impacting a patient’s immune system. Strategies to stimulate the immune system, to prime an antitumor response, are of intense interest. Amid those types of therapies are chimeric antigen receptor T (CAR-T) cells, bispecific antibodies, and antibody–drug conjugates (ADC), which have already been approved in the treatment of acute lymphoblastic leukemia (ALL)/acute myeloid leukemia (AML). In addition, immune checkpoint inhibitors (ICIs), the pattern recognition receptors (PRRs), i.e., NOD-like receptors (NLRs), Toll-like receptors (TLRs), and several kinds of therapy antibodies are well on their way to showing significant benefits for patients with these diseases. This review summarizes the current knowledge of modern methods used in selected pediatric malignancies and presents therapies that may hold promise for the future.
Collapse
|
50
|
Toll-Like Receptors as Drug Targets in the Intestinal Epithelium. Handb Exp Pharmacol 2021; 276:291-314. [PMID: 34783909 DOI: 10.1007/164_2021_563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Toll-like receptors (TLRs) receptors are responsible for initiation of inflammatory responses by their recognition of molecular patterns present in invading microorganisms (such as bacteria, viruses or fungi) or in molecules released following tissue damage in disease states. Expressed in the intestinal epithelium, they initiate an intracellular signalling cascade in response to molecular patterns resulting in the activation of transcription factors and the release of cytokines, chemokines and vasoactive molecules. Intestinal epithelial cells are exposed to microorganisms on a daily basis and form part of the primary defence against pathogens by using TLRs. TLRs and their accessory molecules are subject to tight regulation in these cells so as to not overreact or react in unnecessary circumstances. TLRs have more recently been associated with chronic inflammatory diseases as a result of inappropriate regulation, this can be damaging and lead to chronic inflammatory diseases such as inflammatory bowel disease (IBD). Targeting Toll-like receptors offers a potential therapeutic approach for IBD. In this review, the current knowledge on the TLRs is reviewed along with their association with intestinal diseases. Finally, compounds that target TLRs in animal models of IBD, clinic trials and their future merit as targets are discussed.
Collapse
|