1
|
Kim SH, White Z, Gainullina A, Kang S, Kim J, Dominguez JR, Choi Y, Cabrera I, Plaster M, Takahama M, Czepielewski RS, Yeom J, Gunzer M, Hay N, David O, Chevrier N, Sano T, Kim KW. IL-10 sensing by lung interstitial macrophages prevents bacterial dysbiosis-driven pulmonary inflammation and maintains immune homeostasis. Immunity 2025; 58:1306-1326.e7. [PMID: 40306274 DOI: 10.1016/j.immuni.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 10/02/2024] [Accepted: 04/03/2025] [Indexed: 05/02/2025]
Abstract
Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
Collapse
Affiliation(s)
- Seung Hyeon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Zachary White
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | | | - Soeun Kang
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA
| | - Jiseon Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Yeonwoo Choi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Ivan Cabrera
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Madison Plaster
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Michihiro Takahama
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Rafael S Czepielewski
- Immunology Center of Georgia, Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Jinki Yeom
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Nissim Hay
- Department of Biochemistry and Genetics, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Odile David
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA; Department of Pathology, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolas Chevrier
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA
| | - Teruyuki Sano
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| | - Ki-Wook Kim
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Bruqi K, Strappazzon F. NDP52 and its emerging role in pathogenesis. Cell Death Dis 2025; 16:359. [PMID: 40319017 PMCID: PMC12049512 DOI: 10.1038/s41419-025-07668-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
Autophagy is a pro-survival process that regulates the degradation and renewal of cellular components, making it a crucial mechanism for cellular homeostasis. There are selective forms of autophagy that are specific to a number of substrates, such as pathogens (bacteria or viruses), protein aggregates or excess/damaged organelles. These processes involve as key players autophagy receptors, that link the cargo to be degraded to the autophagic machinery. Among them, NDP52 (also known as CALCOCO2) has been described to act as a "bridge" between the autophagy machinery and (1) damaged mitochondria in the mitophagy process; (2) pathogens during xenophagy or (3) proteins in the process of aggrephagy. The aim of this review is to summarize the major functions of NDP52, and to highlight the existence of two human NDP52 variants that have been described as risk or protective factors for Crohn's disease or Multiple Sclerosis and Alzheimer's disease patients, respectively. As these three diseases share common pathological features that lead to inflammation, such as mitochondria or gut microbiota dysfunctions, but also pathogenic infections, it seems clear that NDP52 could be a key player at the crossroad by acting indirectly on inflammation, and therefore a potential target for clinical applications and benefits.
Collapse
Affiliation(s)
- Krenare Bruqi
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du muscle, UMR5261, U1315, Institut Neuromyogène, Lyon, France
| | - Flavie Strappazzon
- Univ Lyon, Univ Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du muscle, UMR5261, U1315, Institut Neuromyogène, Lyon, France.
| |
Collapse
|
3
|
Li C, Lu W, Zhang H. BTB domain and CNC homolog 2: A master regulator that controls immune response and cancer progression. Biochim Biophys Acta Rev Cancer 2025; 1880:189325. [PMID: 40252853 DOI: 10.1016/j.bbcan.2025.189325] [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: 12/08/2024] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
BTB domain and CNC homolog 2 (BACH2) is a transcription repressor from the basic region leucine zipper (bZIP) family. Although BACH2 is predominantly expressed in lymphoid cells, it plays pivotal roles throughout hematological development and differentiation, ranging from the regulation of hematopoietic stem and progenitor cell (HSPC) lineage commitment to the development of both innate and adaptive immune cells. Given its extensive regulation of immunity, it is not surprising that BACH2 has been implicated in cancer, particularly in hematological malignancies. While multiple findings indicate that BACH2 acts primarily as a tumor suppressor, other findings suggest that BACH2, whether within tumor cells or their surrounding microenvironment, may contribute to tumorigenesis and progression, highlighting the complexity of its roles and the diverse networks involved in different contexts. In this review, we provide a comprehensive overview of the evolving roles of BACH2 across various stages of hematopoiesis, with a particular focus on its associations with cancer and its therapeutic potential in a wide range of cancer types.
Collapse
Affiliation(s)
- Chenyang Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China
| | - Wei Lu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China; Kunming Medical University, Kunming, Yunnan 650500, China
| | - Han Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan 650118, China.
| |
Collapse
|
4
|
Rahaman MM, Wangchuk P, Sarker S. A systematic review on the role of gut microbiome in inflammatory bowel disease: Spotlight on virome and plant metabolites. Microb Pathog 2025; 205:107608. [PMID: 40250496 DOI: 10.1016/j.micpath.2025.107608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Inflammatory bowel diseases (IBD), including ulcerative colitis and Crohn's disease, arise from various factors such as dietary, genetic, immunological, and microbiological influences. The gut microbiota plays a crucial role in the development and treatment of IBD, though the exact mechanisms remain uncertain. Current research has yet to definitively establish the beneficial effects of the microbiome on IBD. Bacteria and viruses (both prokaryotic and eukaryotic) are key components of the microbiome uniquely related to IBD. Numerous studies suggest that dysbiosis of the microbiota, including bacteria, viruses, and bacteriophages, contributes to IBD pathogenesis. Conversely, some research indicates that bacteria and bacteriophages may positively impact IBD outcomes. Additionally, plant metabolites play a crucial role in alleviating IBD due to their anti-inflammatory and microbiome-modulating properties. This systematic review discusses the role of the microbiome in IBD pathogenesis and evaluates the potential connection between plant metabolites and the microbiome in the context of IBD pathophysiology.
Collapse
Affiliation(s)
- Md Mizanur Rahaman
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia
| | - Phurpa Wangchuk
- College of Science and Engineering, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD 4878, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Nguma Bada campus, McGregor Rd, Smithfield, Cairns, QLD, 4878, Australia
| | - Subir Sarker
- Biomedical Sciences and Molecular Biology, College of Medicine and Dentistry, James Cook University, Townsville, QLD, 4811, Australia; Australian Institute of Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia.
| |
Collapse
|
5
|
Yue X, Peng L, Liu S, Zhang B, Zhang X, Chang H, Pei Y, Li X, Liu J, Shui W, Wu L, Xu H, Liu ZJ, Hua T. Structural basis of stepwise proton sensing-mediated GPCR activation. Cell Res 2025:10.1038/s41422-025-01092-w. [PMID: 40211064 DOI: 10.1038/s41422-025-01092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/23/2025] [Indexed: 04/12/2025] Open
Abstract
The regulation of pH homeostasis is crucial in many biological processes vital for survival, growth, and function of life. The pH-sensing G protein-coupled receptors (GPCRs), including GPR4, GPR65 and GPR68, play a pivotal role in detecting changes in extracellular proton concentrations, impacting both physiological and pathological states. However, comprehensive understanding of the proton sensing mechanism is still elusive. Here, we determined the cryo-electron microscopy structures of GPR4 and GPR65 in various activation states across different pH levels, coupled with Gs, Gq or G13 proteins, as well as a small molecule NE52-QQ57-bound inactive GPR4 structure. These structures reveal the dynamic nature of the extracellular loop 2 and its signature conformations in different receptor states, and disclose the proton sensing mechanism mediated by networks of extracellular histidine and carboxylic acid residues. Notably, we unexpectedly captured partially active intermediate states of both GPR4-Gs and GPR4-Gq complexes, and identified a unique allosteric binding site for NE52-QQ57 in GPR4. By integrating prior investigations with our structural analysis and mutagenesis data, we propose a detailed atomic model for stepwise proton sensation and GPCR activation. These insights may pave the way for the development of selective ligands and targeted therapeutic interventions for pH sensing-relevant diseases.
Collapse
Affiliation(s)
- Xiaolei Yue
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Li Peng
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Shenhui Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Bingjie Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaodan Zhang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Hao Chang
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Yuan Pei
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Xiaoting Li
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Junlin Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Lijie Wu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
| | - Huji Xu
- Department of Rheumatology and Immunology, Changzheng Hospital, Second Military Medical University, Shanghai, China.
- School of Clinical Medicine, Tsinghua University, Beijing, China.
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, China.
| | - Zhi-Jie Liu
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Tian Hua
- iHuman Institute, ShanghaiTech University, Shanghai, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| |
Collapse
|
6
|
Wang WH, Xia MH, Liu XR, Lei SF, He P. A Bibliometric Analysis of GWAS on Rheumatoid Arthritis from 2002 to 2024. Hum Hered 2025; 90:18-32. [PMID: 40179854 DOI: 10.1159/000543947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/24/2025] [Indexed: 04/05/2025] Open
Abstract
INTRODUCTION Rheumatoid arthritis (RA) has become a serious threat to human health and quality of life worldwide. Previous studies have demonstrated that genetic factors play a crucial role in the onset and progression of RA. Due to the rapid development of genome-wide association study (GWAS) and large-scale genetic analysis, GWAS research on RA has received widespread attention in recent years. Therefore, we conducted a comprehensive visualization and bibliometric analysis of publications to identify hotspots and future trends in GWAS research on RA. METHODS Literature on RA and GWAS published between 2002 and 2024 was extracted from the Web of Science Core Collection database by strategic screening. Collected data were further analyzed by using VOSviewer, CiteSpace, and Excel. The collaborations networks of countries, authors, institutions, and the co-citation networks of publications were visualized. Finally, research hotspots and fronts were examined. RESULTS A total of 713 publications with 45,773 citations were identified. The number of publications and citations has had a significant surge since 2007. The United States contributed the most publications globally. Okada, Yukinori, was the most influential author. The most productive institution in this field was the University of Manchester. The analysis of keywords revealed that "mendelian randomization analysis", "association", "innate", "instruments", "bias", "pathogenesis", and "genome-wide association study" are likely to be the frontiers of research in this field. CONCLUSION This study can be used to predict future research advances in the fields of GWAS on RA and helps to promote academic collaboration among scholars.
Collapse
Affiliation(s)
- Wen-Hui Wang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China,
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China,
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China,
| | - Ming-Hui Xia
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Xin-Ru Liu
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| | - Shu-Feng Lei
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
- Changzhou Geriatric Hospital Affiliated to Soochow University, Changzhou, China
| | - Pei He
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, China
- Collaborative Innovation Center for Bone and Immunology between Sihong Hospital and Soochow University, Sihong, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Abacar K, Macleod T, Direskeneli H, McGonagle D. Takayasu arteritis: a geographically distant but immunologically proximal MHC-I-opathy. THE LANCET. RHEUMATOLOGY 2025; 7:e290-e302. [PMID: 39855247 DOI: 10.1016/s2665-9913(24)00307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 01/27/2025]
Abstract
Takayasu arteritis, a granulomatosis vasculitis with a pathogenesis that is poorly defined but known to be associated with HLA-B*52, shares many features with other MHC-I-opathies. In addition to the shared clinical features of inflammatory bowel diseases, cutaneous inflammation, and HLA-B*52, is shared association of an IL12B single- nucleotide polymorphism encoding the common IL-12 and IL-23 p40 subunit, which might affect not only type 17 cytokine responses, but also IFNγ and TNF production-the cardinal type 1 cytokines in granuloma formation. Considering the translational context of responses to TNF inhibition in Takayasu arteritis, in this Personal View we propose Takayasu arteritis as a type 1 MHC-I-opathy. Additionally, type 1 and type 17 T-cell immune responses show immune plasticity, which connects the overlapping features of Takayasu arteritis and spondyloarthritis spectrum disorders, providing a basis for shared anti-TNF responses, and points to p40 and IFNγ cytokine antagonism and potential selective CD8 T-cell repertoire ablation.
Collapse
Affiliation(s)
- Kerem Abacar
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK; Division of Rheumatology, Department of Internal Medicine, Marmara University School of Medicine, Istanbul, Türkiye
| | - Tom Macleod
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK
| | - Haner Direskeneli
- Division of Rheumatology, Department of Internal Medicine, Marmara University School of Medicine, Istanbul, Türkiye
| | - Dennis McGonagle
- Section of Musculoskeletal Disease, NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
8
|
Erozkan K, Costedio MM, DeRoss AL. Operative Management of Inflammatory Bowel Disease in Children. Surg Clin North Am 2025; 105:329-356. [PMID: 40015820 DOI: 10.1016/j.suc.2024.10.001] [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: 03/01/2025]
Abstract
Inflammatory bowel disease (IBD) encompasses a spectrum of chronic, idiopathic, and progressive inflammatory conditions of the gastrointestinal tract, with Crohn's disease and ulcerative colitis comprising the majority. Both conditions have distinct pathophysiological and clinical characteristics but share common immune-mediated mechanisms. Specific facets of IBD and its management in the pediatric population differ from adult parallels. This article explores the surgical treatments of pediatric IBD, focusing on indications for surgery and perioperative considerations.
Collapse
Affiliation(s)
- Kamil Erozkan
- Colorectal Division, Department of General Surgery, University Hospitals of Cleveland, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Meagan M Costedio
- Division of Colorectal Surgery, Department of General Surgery, University Hospitals of Cleveland, 11100 Euclid Avenue, Cleveland, OH 44106, USA
| | - Anthony L DeRoss
- Cleveland Clinic Children's, Section of Pediatric Surgery, 9500 Euclid Avenue, R3, Cleveland, OH 44195, USA.
| |
Collapse
|
9
|
Chauhan G, Rieder F. The Pathogenesis of Inflammatory Bowel Diseases. Surg Clin North Am 2025; 105:201-215. [PMID: 40015812 PMCID: PMC11868724 DOI: 10.1016/j.suc.2024.10.008] [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] [Indexed: 03/01/2025]
Abstract
Inflammatory bowel diseases (IBDs) are relapsing, remitting inflammatory diseases of the intestinal tract. Familial aggregation and genome-wide association studies revealed susceptibility variants that point toward a combination of innate immune and adaptive immune dysregulation that in concert with environmental factors, such as our microbiome, can initiate and perpetuate inflammation. Innate immune perturbations include functional abnormalities in the intestinal barrier, endoplasmic reticulum stress, and abnormal recognition of microbes. Adaptive immune changes include dysregulation of cytokines, regulatory T cells, and leukocyte migration. IBD is linked with an abnormal wound-healing response leading to fibrosis. This article summarizes key pathogenic mechanisms in the pathogenesis of IBDs.
Collapse
Affiliation(s)
- Gaurav Chauhan
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA; Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases Institute; Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
10
|
Liefferinckx C, Stern D, Perée H, Bottieau J, Mayer A, Dubussy C, Quertinmont E, Tafciu V, Minsart C, Petrov V, Kvasz A, Coppieters W, Karim L, Rahmouni S, Georges M, Franchimont D. The identification of blood-derived response eQTLs reveals complex effects of regulatory variants on inflammatory and infectious disease risk. PLoS Genet 2025; 21:e1011599. [PMID: 40208878 PMCID: PMC12013874 DOI: 10.1371/journal.pgen.1011599] [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: 09/19/2024] [Revised: 04/22/2025] [Accepted: 01/29/2025] [Indexed: 04/12/2025] Open
Abstract
Hundreds of risk loci for immune mediated inflammatory and infectious diseases have been identified by genome-wide association studies (GWAS). Yet, what causal variants and genes in risk loci underpin the observed associations remains poorly understood for most. The identification of colocalized cis-expression Quantitative Trait Loci (cis-eQTLs) is a promising way to identify candidate causative genes. The catalogue of cis-eQTLs of the immune system is likely incomplete as many cis-eQTLs may be context-specific. We built a large cohort of 406 healthy individuals and expanded the immune cis-regulome through their whole blood transcriptome obtained after stimulation with specific toll-like receptor (TLR) agonists and T-cell receptor (TCR) antagonist. We report three mechanisms that may explain why an eQTL could only be revealed after immune stimulation. More than half of the cis-eQTLs detected in this study would have been overlooked without specific immune stimulations. We then mined this new catalogue of response (r)eQTLs, with public GWAS summary statistics of three diseases through a colocalization approach: inflammatory bowel diseases, rheumatoid arthritis and COVID-19 disease. We identified reQTL-specific colocalizations for risk loci for which no matching eQTL were reported before, revealing interesting new candidate causal genes.
Collapse
Affiliation(s)
- Claire Liefferinckx
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - David Stern
- GIGA Bioinformatics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Hélène Perée
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Jérémie Bottieau
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
| | - Alice Mayer
- GIGA Bioinformatics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Christophe Dubussy
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Eric Quertinmont
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Vjola Tafciu
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Charlotte Minsart
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Vyacheslav Petrov
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Alex Kvasz
- Software development, University of Liège, Liège, Belgium
| | - Wouter Coppieters
- GIGA Genomics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Latifa Karim
- GIGA Genomics Platform, GIGA Institute, University of Liège, Liège, Belgium
| | - Souad Rahmouni
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
| | - Michel Georges
- Unit of Animal Genomics, GIGA Institute, University of Liège, Liège, Belgium
- WEL Research Institute & Faculty of Veterinary Medicine, Liège, Belgium
| | - Denis Franchimont
- Center for the study of IBD, Laboratory of Experimental Gastroenterology, Université libre de Bruxelles, Brussels, Belgium
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
11
|
Wei X, Tang D. Effect of Bacteroides on Crohn's disease. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:393-402. [PMID: 39586813 DOI: 10.1055/a-2435-2659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Crohn's disease (CD), also known as cicatrizing enteritis, is an inflammatory bowel disease that occurs in the distal ileum and right colon of unknown cause and is also called inflammatory bowel disease (IBD) with ulcerative colitis (UC). In recent years, intestinal biota have been confirmed to play a significant role in various gastrointestinal diseases. Studies have found that intestinal microbiota disorders are closely associated with the onset and progression of Crohn's disease. Bacteroidetes, the second largest microbiota in the intestine, are crucial for equilibrium in the microbiota and intestinal environment. Certain Bacteroides can induce the development of Crohn's disease and aggravate intestinal inflammation directly or through their metabolites. Conversely, certain Bacteroides can reduce intestinal inflammation and symptoms of Crohn's disease. This article reviews the effect of several intestinal Bacteroides in the onset and progression of Crohn's disease and their impact on its treatment.
Collapse
Affiliation(s)
- Xuanyu Wei
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People's Hospital, Nanjing University, Yangzhou, China
| |
Collapse
|
12
|
Li Y, Ascui G, Dicker M, Riffelmacher T, Chandra V, Schmiedel B, Chou TF, Vijayanand P, Kronenberg M. Crohn's Disease-associated variant in laccase domain containing 1 (LACC1) modulates T cell gene expression, metabolism and T cell function. Nat Commun 2025; 16:2577. [PMID: 40089498 PMCID: PMC11910630 DOI: 10.1038/s41467-025-57744-3] [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: 12/12/2023] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Genome wide association studies (GWAS) identify many risks for Crohn's disease (CD), including a site near the metabolism gene laccase domain containing 1 (LACC1). We previously found this site near LACC1 was associated with decreased LACC1 expression in T lymphocytes, yet the mechanism affecting gene expression and its links to T cell function and inflammatory disease were unknown. Here we identify variants in the promoter region that influence transcription of LACC1. Direct association of disease-risk variants with lower LACC1 pre-mRNA in human CD4+ T cells is confirmed by comparing transcripts from each allele from donors heterozygous for the LACC1 CD-risk allele. Using gene editing, we validate the function of this promoter region in LACC1 expression in T cells. Human CD4+ T cells with LACC1 gene knockdown show altered metabolism, including reduced oxygen consumption rate, and reduced in vitro regulatory T cell differentiation. Therefore, our study provides a mechanism linking these specific LACC1 variants to colitis by attributing promoter region variants to changes in T cell metabolism and function.
Collapse
Affiliation(s)
- Yingcong Li
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | - Gabriel Ascui
- La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | | | | | - Vivek Chandra
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Pandurangan Vijayanand
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Molecular and Clinical Cancer Medicine and NIHR and CRUK Liverpool Experimental Cancer Medicine Center, University of Liverpool, Liverpool, UK.
| | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
13
|
Andromaque L, Nancey S, Faure M, Rozières A. [Autophagy, dendritic cells, and Crohn's disease - "There is no point in running ; we must leave at the right time"]. Med Sci (Paris) 2025; 41:216-219. [PMID: 40117541 DOI: 10.1051/medsci/2025027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025] Open
Affiliation(s)
- Leslie Andromaque
- Centre international de recherche en infectiologie (CIRI), Univ Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, École normale supérieure de Lyon, Lyon France
| | - Stéphane Nancey
- Centre international de recherche en infectiologie (CIRI), Univ Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, École normale supérieure de Lyon, Lyon France
| | - Mathias Faure
- Centre international de recherche en infectiologie (CIRI), Univ Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, École normale supérieure de Lyon, Lyon France
| | - Aurore Rozières
- Centre international de recherche en infectiologie (CIRI), Univ Claude Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, École normale supérieure de Lyon, Lyon France
| |
Collapse
|
14
|
Foti F, Schuler C, Ruiz PA, Perren L, Malagola E, de Vallière C, Seuwen K, Hausmann M, Rogler G. The Simultaneous Deletion of pH-Sensing Receptors GPR4 and OGR1 (GPR68) Ameliorates Colitis with Additive Effects on Multiple Parameters of Inflammation. Int J Mol Sci 2025; 26:1552. [PMID: 40004018 PMCID: PMC11855581 DOI: 10.3390/ijms26041552] [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: 12/02/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
G protein-coupled receptors (GPRs), including pro-inflammatory GPR4 and ovarian cancer GPR1 (OGR1/GPR68), are involved in the pH sensing of the extracellular space and have been implicated in inflammatory bowel disease (IBD). Previous data show that a loss of GPR4 or OGR1 independently is associated with reduced intestinal inflammation in mouse models of experimental colitis. In the present manuscript, we investigated the impact of the simultaneous loss of GPR4 and OGR1 in animal models of IBD. To study the effects of combined loss of Gpr4 Ogr1 in IBD we used the well-established acute dextran sodium sulfate (DSS) and spontaneous Il10-/- murine colitis models. Disease severity was assessed using multiple clinical scores (e.g., body weight loss, disease activity score, murine endoscopic index of colitis severity (MEICS) and histological analyses). Real-time quantitative polymerase chain reaction (qPCR), Western blot, and flow cytometry were used to investigate changes in pro-inflammatory cytokines expression and immune cells infiltration. We found that a combined loss of GPR4 and OGR1 significantly reduces colon inflammation in IBD relative to single deficiencies as evidenced by reduced body weight loss, disease score, CD4/CD8 ratio, and Il1β, Il6, and Tnf in the colon. Similarly, in the II10 deficiency model, the inflammation was significantly ameliorated upon the simultaneous deletion of GPR4 and OGR1, evidenced by a reduction in the MEICS score, colon length, Tnf and Il1β measurements, and a decrease in the number of macrophages in the colon, as compared to single deletions. Importantly, hydroxyproline levels were decreased close to baseline in Il10-/- × Gpr4-/- × Ogr1-/- mice. Our findings demonstrate that the simultaneous loss of GRP4 and OGR1 functions exerts an additive effect on multiple parameters associated with colonic inflammation. These results further reinforce the hypothesis that chronic inflammatory acidosis is a driver of fibrosis and is dependent on GPR4 and OGR1 signaling. The inhibition of both GPR4 and OGR1 by pH-sensing receptor modulators may constitute as a potential therapeutic option for IBD, as both pH-sensing receptors appear to sustain inflammation by acting on complementary pro-inflammatory pathways.
Collapse
|
15
|
Tamburri S, Zucchelli C, Matafora V, Zapparoli E, Jevtic Z, Farris F, Iannelli F, Musco G, Bachi A. SP140 represses specific loci by recruiting polycomb repressive complex 2 and NuRD complex. Nucleic Acids Res 2025; 53:gkae1215. [PMID: 39718989 PMCID: PMC11879014 DOI: 10.1093/nar/gkae1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/26/2024] Open
Abstract
SP140, a lymphocytic-restricted protein, is an epigenetic reader working as a corepressor of genes implicated in inflammation and orchestrating macrophage transcriptional programs to maintain cellular identity. Reduced SP140 expression is associated both to autoimmune diseases and blood cancers. However, the molecular mechanisms that link SP140 altered protein levels to detrimental effects on the immune response and cellular growth, as well as the interactors through which SP140 promotes gene silencing, remain elusive. In this work, we have applied a multi-omics approach (i.e. interactomics, ChIP-seq and proteomics) in two Burkitt lymphoma cell lines to identify both interactors and target genes of endogenous SP140. We found that SP140 interacts with the PRC2 and NuRD complexes, and we showed that these interactions are functional as SP140 directs H3K27me3 deposition and NuRD binding on a set of target genes implicated in cellular growth and leukemia progression.
Collapse
Affiliation(s)
- Simone Tamburri
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Chiara Zucchelli
- Biomolecular NMR Laboratory, Division of Genetics and Cell biology, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Vittoria Matafora
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Ettore Zapparoli
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Zivojin Jevtic
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Francesco Farris
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Fabio Iannelli
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| | - Giovanna Musco
- Biomolecular NMR Laboratory, Division of Genetics and Cell biology, IRCCS Ospedale San Raffaele, Via Olgettina 58, 20132 Milano, Italy
| | - Angela Bachi
- IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 16039 Milano, Italy
| |
Collapse
|
16
|
Omori-Miyake M, Kawakami R, Kuwahara M, Okabe M, Muto J, Imamura T, Yamashita M. Loss of Bach2 in T cells causes prolonged allergic inflammation through accumulation of effector T cells and disruption of epidermal barrier. J Allergy Clin Immunol 2025:S0091-6749(25)00128-9. [PMID: 39924123 DOI: 10.1016/j.jaci.2025.01.036] [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: 02/15/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND Bach2 has been suggested to be a risk factor for allergic diseases in previous studies. Because type IV hypersensitivity reactions, including allergic contact dermatitis (ACD), develop through activated T cells, and because the expression of Bach2 is regulated in the development and functional differentiation of T cells, the expression of Bach2 in T cells may be involved in the onset of ACD. However, the role of Bach2 in T cells during ACD development has not yet been determined. OBJECTIVE We investigated the role of the appropriate expression of Bach2 in T cells in the development and prolongation of ACD. METHODS We induced ACD in mice by repeatedly applying a hapten and analyzed the expression of Bach2 in the T cells of lesional skin or skin-draining lymph nodes (sdLNs). We performed a phenotypic analysis of the skin and/or sdLNs by comparing mice with T cells overexpressing Bach2 or with Bach2 loss to the control mice. RESULTS We found that Bach2lo T cells accumulated in the skin and sdLNs as ACD developed. T-cell-specific Bach2-deficient mice showed more severe inflammatory responses to the hapten and had prolonged inflammation with T cells expressing higher levels of IL-13 in the skin and IFN-γ and IL-13 in the sdLNs. In contrast, the mice overexpressing Bach2 in T cells developed almost no symptoms of ACD. CONCLUSION The appropriate expression of Bach2 in T cells may be a key factor in the resolution of ACD.
Collapse
Affiliation(s)
- Miyuki Omori-Miyake
- Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Makoto Kuwahara
- Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masataka Okabe
- Department of Anatomy, The Jikei University School of Medicine, Tokyo, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masakatsu Yamashita
- Department of Infections and Host Defenses, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Immunology, Ehime University Graduate School of Medicine, Ehime, Japan.
| |
Collapse
|
17
|
Kayali S, Fantasia S, Gaiani F, Cavallaro LG, de’Angelis GL, Laghi L. NOD2 and Crohn's Disease Clinical Practice: From Epidemiology to Diagnosis and Therapy, Rewired. Inflamm Bowel Dis 2025; 31:552-562. [PMID: 38582044 PMCID: PMC11808579 DOI: 10.1093/ibd/izae075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Indexed: 04/08/2024]
Abstract
Crohn's disease (CD) is a chronic inflammatory bowel disease with a multifactorial pathogenesis involving environmental and genetic factors. Since the late 20th century, the discovery of the first susceptibility gene (NOD2, previously referred to as CARD15) for CD has paved the way for further investigations into the correlations between clinical features and genetics, and its potential impact on clinical practice has fueled the research in the last 2 decades. Recent therapeutic advancements involving novel biologic drugs and small molecules have shifted inflammatory bowel disease management from a disease-centered to a patient-centric approach. To date, the role of NOD2 has not been fully understood yet. Recent data suggest that its clinical impact may be greater than currently recognized. This review overviews the most common NOD2 variants' role in real-life clinical practice. These genetic variants increase the risk of developing the disease and can aid in tailoring diagnosis and treatment. They are associated with the stricturing phenotype and ileal involvement and increase the risk of steroid refractoriness. In the meantime, limited and inconclusive evidence exists regarding their predictive role in response to azathioprine, biologic drugs, and small molecules. Eventually, their role in increasing the risk for surgery is evident, especially in those with the L1007fs variant. If further trials will support the initial evidence reported so far, NOD2 genetic variants will emerge as possible candidates for developing precision medicine in CD.
Collapse
Affiliation(s)
- Stefano Kayali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Stefano Fantasia
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Federica Gaiani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Gastroenterology and Endoscopy Unit, University Hospital of Parma, Parma, Italy
| | | | | | - Luigi Laghi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Laboratory of Molecular Gastroenterology, Humanitas Clinical and Research Centre, Rozzano, Italy
| |
Collapse
|
18
|
Witt KC, Dziulko A, An J, Pekovic F, Cheng AX, Liu GY, Lee OV, Turner DJ, Lari A, Gaidt MM, Chavez R, Fattinger SA, Abraham P, Dhaliwal H, Lee AY, Kotov DI, Coscoy L, Glaunsinger BA, Valkov E, Chuong EB, Vance RE. The SP140-RESIST pathway regulates interferon mRNA stability and antiviral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.28.610186. [PMID: 39974928 PMCID: PMC11838211 DOI: 10.1101/2024.08.28.610186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Type I interferons (IFN-Is) are essential for antiviral immunity but must be tightly regulated1-3. The conserved transcriptional repressor SP140 inhibits interferon beta (Ifnb1) expression via an unknown mechanism4,5. Here we report that SP140 does not directly repress Ifnb1 transcription. Instead, SP140 negatively regulates Ifnb1 mRNA stability by directly repressing the expression of a previously uncharacterized regulator we call RESIST (REgulated Stimulator of Interferon via Stabilization of Transcript, previously annotated as Annexin-2 Receptor). RESIST promotes Ifnb1 mRNA stability by counteracting Ifnb1 mRNA destabilization mediated by the Tristetraprolin (TTP) family of RNA-binding proteins and the CCR4-NOT deadenylase complex. SP140 localizes within nuclear bodies, punctate structures that play important roles in silencing DNA virus gene expression in the nucleus4. Consistent with this observation, we found that SP140 inhibits replication of the gammaherpesvirus MHV68. The antiviral activity of SP140 was independent of its ability to regulate Ifnb1. Our results establish dual antiviral and interferon regulatory functions for SP140. We propose that SP140 and RESIST participate in antiviral effector-triggered immunity6,7.
Collapse
Affiliation(s)
- Kristen C Witt
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Adam Dziulko
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Joohyun An
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Filip Pekovic
- National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Arthur Xiuyuan Cheng
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Grace Y Liu
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ophelia Vosshall Lee
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - David J Turner
- National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Azra Lari
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Moritz M Gaidt
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Current address: Research Institute of Molecular Pathology, Vienna BioCenter, Vienna, Austria
| | - Roberto Chavez
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Stefan A Fattinger
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Preethy Abraham
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | | | - Angus Y Lee
- Cancer Research Laboratory, University of California, Berkeley, CA, USA
| | - Dmitri I Kotov
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Laurent Coscoy
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Britt A Glaunsinger
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Department of Plant & Microbial Biology, University of California, Berkeley, CA, USA
| | - Eugene Valkov
- National Cancer Institute, National Institutes of Health, Frederick, MD, USA
| | - Edward B Chuong
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Russell E Vance
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA
- Division of Immunology and Molecular Medicine, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Cancer Research Laboratory, University of California, Berkeley, CA, USA
| |
Collapse
|
19
|
Stoolman JS, Grant RA, Billingham LK, Poor TA, Weinberg SE, Harding MC, Lu Z, Miska J, Szibor M, Budinger GRS, Chandel NS. Mitochondria complex III-generated superoxide is essential for IL-10 secretion in macrophages. SCIENCE ADVANCES 2025; 11:eadu4369. [PMID: 39841842 PMCID: PMC11753406 DOI: 10.1126/sciadv.adu4369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025]
Abstract
Mitochondrial electron transport chain (ETC) function modulates macrophage biology; however, mechanisms underlying mitochondria ETC control of macrophage immune responses are not fully understood. Here, we report that mutant mice with mitochondria ETC complex III (CIII)-deficient macrophages exhibit increased susceptibility to influenza A virus (IAV) and LPS-induced endotoxic shock. Cultured bone marrow-derived macrophages (BMDMs) isolated from these mitochondria CIII-deficient mice released less IL-10 than controls following TLR3 or TLR4 stimulation. Unexpectedly, restoring mitochondrial respiration without generating superoxide using alternative oxidase (AOX) was not sufficient to reverse LPS-induced endotoxic shock susceptibility or restore IL-10 release. However, activation of protein kinase A (PKA) rescued IL-10 release in mitochondria CIII-deficient BMDMs following LPS stimulation. In addition, mitochondria CIII deficiency did not affect BMDM responses to interleukin-4 (IL-4) stimulation. Thus, our results highlight the essential role of mitochondria CIII-generated superoxide in the release of anti-inflammatory IL-10 in response to TLR stimulation.
Collapse
Affiliation(s)
- Joshua S. Stoolman
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A. Grant
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Leah K. Billingham
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Taylor A. Poor
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Samuel E. Weinberg
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Madeline C. Harding
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyan Lu
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marten Szibor
- Department of Cardiothoracic Surgery, Center for Sepsis Control and Care (CSCC), Jena University Hospital, Friedrich Schiller University of Jena, Am Klinikum 1, 07747 Jena, Germany
- Faculty of Medicine and Health Technology, FI-33014 Tampere University, Tampere, Finland
| | - GR Scott Budinger
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Navdeep S. Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
20
|
Su Y, Li J, Chen Y, Bao J, Lei Z, Ma M, Zhang W, Liu Q, Xu B, Hu T, Hu Y. α-Methyl-Tryptophan Inhibits SLC6A14 Expression and Exhibits Immunomodulatory Effects in Crohn's Disease. J Inflamm Res 2025; 18:1127-1145. [PMID: 39877135 PMCID: PMC11774106 DOI: 10.2147/jir.s495855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/19/2025] [Indexed: 01/31/2025] Open
Abstract
Introduction Crohn's disease (CD) is a chronic inflammatory condition of the intestines with a rising global incidence. Traditional diagnostic and therapeutic methods have limitations, necessitating the exploration of more effective strategies. Methods In this study, we employed the Gene Expression Omnibus database to identify genes that are differentially expressed in CD. RT-PCR and immunohistochemical analysis were used to SLC6A14 RNA and protein expression in the colons of CD mice and CD tissues from patients. The mouse model of CD was induced by dextran sodium sulfate (DSS). Infiltrating immune cells in mouse model were screened by flow cytometry. Results We discovered that SLC6A14 is significantly overexpressed in CD samples, and its expression is positively correlated with the degree of infiltration by CD4+ and CD8+ T cells. The elevated levels of SLC6A14 RNA and protein were confirmed in clinical CD tissues. The SLC6A14 inhibitor α-methyl-tryptophan (α-MT) significantly decreased the expression of SLC6A14 RNA and protein in the colons of CD mice. The α-MT treatment group also exhibited reduced levels of cytokines involved in T cell differentiation (IFN-γ and TNF-α) and the expression of immune cell surface markers CXCR-3 and LAG-3. Flow cytometry analysis revealed a significant increase in the infiltration of CD4+ and CD8+ T cells in the DSS-treated group compared to the control group. Conversely, the α-MT treatment group showed a significant reduction in CD4+ and CD8+ T cell infiltration and the restoration of intestinal parameters in CD mice. These findings underscore the role of SLC6A14 in regulating intestinal immune cell infiltration during CD progression. Discussion Our findings suggest that SLC6A14 could serve as a potential diagnostic biomarker and therapeutic target for CD. Furthermore, α-MT offers a novel approach for the clinical diagnosis and treatment of CD by targeting SLC6A14 for therapeutic intervention.
Collapse
Affiliation(s)
- YongCheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
| | - Jiangquan Li
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
| | - Yijia Chen
- Department of Gastroenterology, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, People’s Republic of China
| | - Jiachen Bao
- Department of Gastroenterology, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, People’s Republic of China
| | - Ziyu Lei
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
| | - Miaomiao Ma
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
| | - Wenqing Zhang
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
| | - Qian Liu
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Beibei Xu
- Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518055, People’s Republic of China
| | - Tianhui Hu
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, 361102, People’s Republic of China
- Integrated Chinese and Western Medicine Institute for Children Health & Drug Innovation, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330004, People’s Republic of China
| | - Yiqun Hu
- Department of Gastroenterology, The National Key Clinical Specialty, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, 361004, People’s Republic of China
| |
Collapse
|
21
|
Huang Z, Yuan W. Exploring genetic structures and shared sites between alcohol, cheese intake, and inflammatory bowel disease. Front Nutr 2025; 12:1468457. [PMID: 39917747 PMCID: PMC11798781 DOI: 10.3389/fnut.2025.1468457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/09/2025] [Indexed: 02/09/2025] Open
Abstract
Background An association has been observed between alcohol and cheese intake and the onset of inflammatory bowel disease (IBD), necessitating further exploration from a genetic structural perspective. Methods The present analysis was focused on the intake of alcohol and cheese in conjunction with IBD genome-wide association study (GWAS) data, with the objective of exploring genetic correlations and identifying common loci. Initially, overall genetic correlations were assessed employing two methodologies: linkage disequilibrium score regression (LDSC) and genetic covariance analyzer (GNOVA). Subsequently, local correlations were examined through the SUPERGNOVA method. A genetic overlap analysis between various traits was then conducted based on the statistical theory of conditional/conjunctional false discovery rate (cond/conjFDR). Ultimately, shared loci between the two traits were identified via conjFDR analysis and multi-trait analysis of GWAS (MTAG). Results Substantial overall correlations were noted at the genome-wide level between alcohol and cheese intake and both IBD and Crohn's disease (CD), whereas the association with ulcerative colitis (UC) was of lesser significance. In the local genetic analysis, chromosome 16 emerged as a key region implicated in the relationship between alcohol and cheese intake and IBD (including both CD and UC). The conjFDR analysis confirmed the genetic overlap between the two diseases. Furthermore, both conjFDR and MTAG analyses identified multiple shared genetic loci, with nine genes (Y_RNA, DENND1B, GCKR, KPNA7, CLN3, SLC39A8, FUT2, ERAP2, and SMAD3) being. Conclusion The present study provides genetic evidence supporting the comorbidity of alcohol and cheese intake with IBD, offering novel insights into potential strategies for the prevention and treatment of IBD through the modulation of alcohol and cheese consumption.
Collapse
Affiliation(s)
- Zhifang Huang
- Department of Anorectal Surgery, Jiangmen Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, China
| | - Weichao Yuan
- Department of Anorectal Surgery, Affiliated Hospital of Jiujiang University, Jiujiang, China
| |
Collapse
|
22
|
Nicolle S, Barker M, Barrett J, Campbell M, Wojno-Picon J, Atkinson SJ, Aylott H, Kessedjian H, He Y, Messenger C, Roberts E, Spitzfaden C, Le J, Zinn N, Werner T, Dümpelfeld B, Bantscheff M, Somers DO, Reid H, Thang K, Gobbetti T, Lewis HD. Phenotype-Led Identification of IL-10 Upregulators in Human CD4 + T-cells and Elucidation of Their Pharmacology as Highly Selective CDK8/CDK19 Inhibitors. J Med Chem 2025; 68:1883-1900. [PMID: 39780505 DOI: 10.1021/acs.jmedchem.4c02630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Therapeutics promoting the endogenous production of IL-10 have the potential to restore homeostasis in inflammatory disorders such as inflammatory bowel disease (IBD). Here we describe the identification of a series of IL-10 upregulators based on a pyrimidyl-piperidine scaffold through a high throughput phenotypic CD4+ T-cell multiplex assay. In vitro optimization of the initial hit yielded a lead with good potency and an in vitro clearance profile, compound 3-7, which additionally demonstrated efficacy in a murine endotoxin challenge PK-PD mechanistic model. Target deconvolution efforts identified compound 3-7 as a highly selective CDK8/19 inhibitor, and crystallographic studies unveiled its binding mode to the CDK8/Cyclin-C complex, characterized by an unusual water-mediated hydrogen bond to the kinase hinge region.
Collapse
Affiliation(s)
- Simon Nicolle
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Mike Barker
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - John Barrett
- In Vitro In Vivo Translation, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Matthew Campbell
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Stephen J Atkinson
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Helen Aylott
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Yanan He
- Medicine Development and Supply, GlaxoSmithKline R&D Hub, 1250 South Collegeville Road, Upper Providence, Pennsylvania 19426, United States
| | - Cassie Messenger
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Emma Roberts
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Claus Spitzfaden
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Joelle Le
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Nico Zinn
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Thilo Werner
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Birgit Dümpelfeld
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Marcus Bantscheff
- Cellzome GmbH, GlaxoSmithKline, Meyerhofstrasse 1, Heidelberg 69117, Germany
| | - Don O Somers
- Medicine Design, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Heather Reid
- Immunology Research Unit GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Kevin Thang
- Immunology Research Unit GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Thomas Gobbetti
- Immunology Research Unit GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Huw D Lewis
- Immunology Research Unit GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
23
|
Kelly C, Sartor RB, Rawls JF. Early subclinical stages of the inflammatory bowel diseases: insights from human and animal studies. Am J Physiol Gastrointest Liver Physiol 2025; 328:G17-G31. [PMID: 39499254 PMCID: PMC11901386 DOI: 10.1152/ajpgi.00252.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 11/07/2024]
Abstract
The inflammatory bowel diseases (IBD) occur in genetically susceptible individuals that mount inappropriate immune responses to their microbiota leading to chronic intestinal inflammation. The natural history of IBD progression begins with early subclinical stages of disease that occur before clinical diagnosis. Improved understanding of those early subclinical stages could lead to new or improved strategies for IBD diagnosis, prognostication, or prevention. Here, we review our current understanding of the early subclinical stages of IBD in humans including studies from first-degree relatives of patients with IBD and members of the general population who go on to develop IBD. We also discuss representative mouse models of IBD that can be used to investigate disease dynamics and host-microbiota relationships during these early stages. In particular, we underscore how mouse models of IBD that develop disease later in life with variable penetrance may present valuable opportunities to discern early subclinical mechanisms of disease before histological inflammation and other severe symptoms become apparent.
Collapse
Affiliation(s)
- Cecelia Kelly
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| | - R Balfour Sartor
- Division of Gastroenterology and Hepatology, Department of Medicine, University of North Carolina, Chapel Hill, North Carolina, United States
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, United States
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine, Durham, North Carolina, United States
| |
Collapse
|
24
|
Huang Y, Xu L, Yang Q, Xiao X, Ye Z, Li R, Guan Y, Wu X. NLRP12 c.1382dup promotes the development of Crohn's disease through the ERK/NLRP3/ IL-1β pathway. Gene 2024; 931:148855. [PMID: 39181275 DOI: 10.1016/j.gene.2024.148855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Whole-genome sequencing was used to identify a dominant inherited NLRP12 c.1382dup mutation in refractory familial Crohn's disease (CD) patients. Additionally, we observed a T insertion at position 1382 in the third exon of NLRP12, leading to a frameshift mutation. Isolation of peripheral blood from mutation carriers and subsequent experiments demonstrated increased interleukin (IL)-1β in CD patients with the NLRP12 c.1382dup mutation. However, the mechanisms by which the NLRP12 c.1382dup mutation mediates IL-1β remain unclear. Our research findings reveal a close correlation between elevated p-ERK levels and increased expression of NLRP3 and IL-1β in the presence of the NLRP12 c.1382dup mutation. Further experiments demonstrate that inhibiting p-ERK with PD98059 effectively reduces the production of NLRP3 and IL-1β. This discovery provides new insights into the pathogenesis of CD, highlighting the significant role of the ERK/NLRP3/IL-1β pathway in the progression of CD. Not only does this offer novel therapeutic targets for treating CD, but it also lays the groundwork for the development of treatment strategies targeting this pathway.
Collapse
Affiliation(s)
- Yang Huang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006 Yancheng, Jiangsu, China
| | - Lincheng Xu
- Department of Pathology, Yancheng NO.1 People's Hospital., China
| | - Qingqing Yang
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006 Yancheng, Jiangsu, China
| | - Xueyi Xiao
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006 Yancheng, Jiangsu, China
| | - Zhenyu Ye
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Rongqing Li
- Department of Medical Genetics and Prenatal Diagnosis, The Affiliated Taizhou People's Hospital of Nanjing Medical University, 225399 Taizhou, Jiangsu, China.
| | - Yanyan Guan
- Department of Pathology, Yancheng NO.1 People's Hospital., China
| | - Xudong Wu
- Department of Gastroenterology, The Yancheng Clinical College of Xuzhou Medical University, 224006 Yancheng, Jiangsu, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China; Department of Gastroenterology, Yancheng NO.1 People's Hospital, China.
| |
Collapse
|
25
|
Demirkan A, van Dongen J, Finnicum CT, Westra HJ, Jankipersadsing S, Willemsen G, Ijzerman RG, Boomsma DI, Ehli EA, Bonder MJ, Fu J, Franke L, Wijmenga C, de Geus EJC, Kurilshikov A, Zhernakova A. Linking the gut microbiome to host DNA methylation by a discovery and replication epigenome-wide association study. BMC Genomics 2024; 25:1224. [PMID: 39702006 DOI: 10.1186/s12864-024-11136-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024] Open
Abstract
Microbiome influences multiple human systems, but its effects on gene methylation is unknown. We investigated the relations between gene methylation in blood and the abundance of common gut bacteria profiled by 16s rRNA gene sequencing in two population-based Dutch cohorts: LifeLines-Deep (LLD, n = 616, discovery) and the Netherlands Twin Register (NTR, n = 296, replication). In LLD, we also explored microbial pathways using data generated by shotgun metagenomic sequencing (n = 683). Methylation in both cohorts was profiled in blood samples using the Illumina 450K array. Discovery and replication analysis identified two independent CpGs associated with the genus Eggerthella: cg16586104 (Pmeta-analysis = 3.21 × 10-11) and cg12234533 (Pmeta-analysis = 4.29 × 10-10). We also show that microbiome can mediate the effect of environmental factors on host gene methylation. In this first association study linking epigenome to microbiome, we found and replicated the associations of two CpGs to the abundance of genus Eggerthella and identified microbiome as a mediator of the exposome. These associations are observational and suggest further investigation in larger and longitudinal set-ups.
Collapse
Affiliation(s)
- Ayşe Demirkan
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
- Department of Clinical and Experimental Medicine, Section of Statistical Multi-omics, School of Biosciences and Medicine & People-Centered AI institute University of Surrey, Guildford, United Kingdom.
| | - Jenny van Dongen
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Casey T Finnicum
- Avera Institute of Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, Sioux Falls, SD, USA
| | - Harm-Jan Westra
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Soesma Jankipersadsing
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Gonneke Willemsen
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Richard G Ijzerman
- Department of Endocrinology, Amsterdam University Medical Center, location AMC, Amsterdam, the Netherlands
| | - Dorret I Boomsma
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
- Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erik A Ehli
- Avera Institute of Human Genetics, Avera McKennan Hospital & University Health Center, Sioux Falls, Sioux Falls, SD, USA
| | - Marc Jan Bonder
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Eco J C de Geus
- Biological Psychology, Vrije Universiteit, Amsterdam, the Netherlands
- Amsterdam Public Health Research Institute, Amsterdam MC, Amsterdam, the Netherlands
| | - Alexander Kurilshikov
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| |
Collapse
|
26
|
Ghasempour S, Warner N, Guan R, Rodari MM, Ivanochko D, Whittaker Hawkins R, Marwaha A, Nowak JK, Liang Y, Mulder DJ, Stallard L, Li M, Yu DD, Pluthero FG, Batura V, Zhao M, Siddiqui I, Upton JE, Hulst JM, Kahr WH, Mendoza-Londono R, Charbit-Henrion F, Hoefsloot LH, Khiat A, Moreira D, Trindade E, Espinheira MDC, Pinto Pais I, Weerts MJ, Douben H, Kotlarz D, Snapper SB, Klein C, Dowling JJ, Julien JP, Joosten M, Cerf-Bensussan N, Freeman SA, Parlato M, van Ham TJ, Muise AM. Human ITGAV variants are associated with immune dysregulation, brain abnormalities, and colitis. J Exp Med 2024; 221:e20240546. [PMID: 39526957 PMCID: PMC11554753 DOI: 10.1084/jem.20240546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/16/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Integrin heterodimers containing an Integrin alpha V subunit are essential for development and play critical roles in cell adhesion and signaling. We identified biallelic variants in the gene coding for Integrin alpha V (ITGAV) in three independent families (two patients and four fetuses) that either caused abnormal mRNA and the loss of functional protein or caused mistargeting of the integrin. This led to eye and brain abnormalities, inflammatory bowel disease, immune dysregulation, and other developmental issues. Mechanistically, the reduction of functional Integrin αV resulted in the dysregulation of several pathways including TGF-β-dependent signaling and αVβ3-regulated immune signaling. These effects were confirmed using immunostaining, RNA sequencing, and functional studies in patient-derived cells. The genetic deletion of itgav in zebrafish recapitulated patient phenotypes including retinal and brain defects and the loss of microglia in early development as well as colitis in juvenile zebrafish with reduced SMAD3 expression and transcriptional regulation. Taken together, the ITGAV variants identified in this report caused a previously unknown human disease characterized by brain and developmental defects in the case of complete loss-of-function and atopy, neurodevelopmental defects, and colitis in cases of incomplete loss-of-function.
Collapse
Affiliation(s)
- Sina Ghasempour
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Neil Warner
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Rei Guan
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Marco M. Rodari
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Danton Ivanochko
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Canada
| | | | - Ashish Marwaha
- Division of Genetics, Department of Medical Genetics, University of Calgary, Alberta Children’s Hospital, Calgary, Canada
| | - Jan K. Nowak
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Yijing Liang
- Center for Computational Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Daniel J. Mulder
- Department of Pediatrics, Gastrointestinal Diseases Research Unit, Queen’s University, Kingston, Canada
| | - Lorraine Stallard
- National Centre for Pediatric Gastroenterology, Children’s Health Ireland, Dublin, Ireland
| | - Michael Li
- Center for Computational Medicine, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Daniel D. Yu
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Fred G. Pluthero
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Vritika Batura
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Mo Zhao
- Genetics and Genome Biology, Research Institute, Hospital for Sick Children, Toronto, Canada
| | - Iram Siddiqui
- Division of Pathology, Department of Pediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Canada
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Julia E.M. Upton
- Division of Immunology and Allergy, The Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Jessie M. Hulst
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, Canada
| | - Walter H.A. Kahr
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Roberto Mendoza-Londono
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Clinical and Metabolic Genetics, Department of Paediatrics, The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Fabienne Charbit-Henrion
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
- Genomic Medicine for Rare Diseases, Necker-Enfants Malades Hospital, Paris, France
| | - Lies H. Hoefsloot
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Anis Khiat
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Diana Moreira
- Consulta de Imunodeficiências Primárias, Serviço de Pediatria, Centro Hospitalar Vila Nova de Gaia e Espinho, Vila Nova de Gaia, Portugal
| | - Eunice Trindade
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Maria do Céu Espinheira
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Isabel Pinto Pais
- Department of Pediatrics, Unit of Pediatric Gastroenterology, Hepatology and Nutrition, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Marjolein J.A. Weerts
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Hannie Douben
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Daniel Kotlarz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Child and Adolescent Health, Munich Site, Munich, Germany
- Institute of Translational Genomics, Helmholtz Zentrum München German Research Center for Environmental Health, Neuherberg, Germany
| | - Scott B. Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston, Children’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- German Center for Child and Adolescent Health, Munich Site, Munich, Germany
| | - James J. Dowling
- Genetics and Genome Biology, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
| | - Jean-Philippe Julien
- Program in Molecular Medicine, The Hospital for Sick Children Research Institute, Toronto, Canada
| | - Marieke Joosten
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Spencer A. Freeman
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Marianna Parlato
- Laboratory of Intestinal Immunity, Université Paris-Cité, Institut Imagine, INSERM U1163, Paris, France
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Aleixo M. Muise
- Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
- Department of Paediatrics, University of Toronto, Toronto, Canada
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, Toronto, Canada
| |
Collapse
|
27
|
Jin Q, Ren F, Song P. Innovate therapeutic targets for autoimmune diseases: insights from proteome-wide mendelian randomization and Bayesian colocalization. Autoimmunity 2024; 57:2330392. [PMID: 38515381 DOI: 10.1080/08916934.2024.2330392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/10/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Despite growing knowledge regarding the pathogenesis of autoimmune diseases (ADs) onset, the current treatment remains unsatisfactory. This study aimed to identify innovative therapeutic targets for ADs through various analytical approaches. RESEARCH DESIGN AND METHODS Utilizing Mendelian randomization, Bayesian co-localization, phenotype scanning, and protein-protein interaction network, we explored potential therapeutic targets for 14 ADs and externally validated our preliminary findings. RESULTS This study identified 12 circulating proteins as potential therapeutic targets for six ADs. Specifically, IL12B was judged to be a risk factor for ankylosing spondylitis (p = 1.61E - 07). TYMP (p = 6.28E - 06) was identified as a protective factor for ulcerative colitis. For Crohn's disease, ERAP2 (p = 4.47E - 14), HP (p = 2.08E - 05), and RSPO3 (p = 6.52E - 07), were identified as facilitators, whereas FLRT3 (p = 3.42E - 07) had a protective effect. In rheumatoid arthritis, SWAP70 (p = 3.26E - 10), SIGLEC6 (p = 2.47E - 05), ISG15 (p = 3.69E - 05), and FCRL3 (p = 1.10E - 10) were identified as risk factors. B4GALT1 (p = 6.59E - 05) was associated with a lower risk of Type 1 diabetes (T1D). Interestingly, CTSH was identified as a protective factor for narcolepsy (p = 1.58E - 09) but a risk factor for T1D (p = 7.36E - 11), respectively. External validation supported the associations of eight of these proteins with three ADs. CONCLUSIONS Our integrated study identified 12 potential therapeutic targets for ADs and provided novel insights into future drug development for ADs.
Collapse
Affiliation(s)
- Qiubai Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Feihong Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate school, Beijing University of Chinese Medicine, Beijing, China
| | - Ping Song
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
28
|
Okumura R, Takeda K. The role of the mucosal barrier system in maintaining gut symbiosis to prevent intestinal inflammation. Semin Immunopathol 2024; 47:2. [PMID: 39589551 PMCID: PMC11599372 DOI: 10.1007/s00281-024-01026-5] [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: 10/10/2023] [Accepted: 09/29/2024] [Indexed: 11/27/2024]
Abstract
In the intestinal tract, where numerous intestinal bacteria reside, intestinal epithelial cells produce and release various antimicrobial molecules that form a complex barrier on the mucosal surface. These barrier molecules can be classified into two groups based on their functions: those that exhibit bactericidal activity through chemical reactions, such as antimicrobial peptides, and those that physically hinder bacterial invasion, like mucins, which lack bactericidal properties. In the small intestine, where Paneth cells specialize in producing antimicrobial peptides, the chemical barrier molecules primarily inhibit bacterial growth. In contrast, in the large intestine, where Paneth cells are absent, allowing bacterial growth, the primary defense mechanism is the physical barrier, mainly composed of mucus, which controls bacterial movement and prevents their invasion of intestinal tissues. The expression of these barrier molecules is regulated by metabolites produced by bacteria in the intestinal lumen and cytokines produced by immune cells in the lamina propria. This regulation establishes a defense mechanism that adapts to changes in the intestinal environment, such as alterations in gut microbial composition and the presence of pathogenic bacterial infections. Consequently, when the integrity of the gut mucosal barrier is compromised, commensal bacteria and pathogenic microorganisms from outside the body can invade intestinal tissues, leading to conditions such as intestinal inflammation, as observed in cases of inflammatory bowel disease.
Collapse
Affiliation(s)
- Ryu Okumura
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan
- Institute for Open and Transdisciplinary Research Initiative, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Kiyoshi Takeda
- Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan.
- WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, 565-0871, Japan.
- Institute for Open and Transdisciplinary Research Initiative, Osaka University, Suita, Osaka, 565-0871, Japan.
- Center for Infectious Disease Education and Research, Osaka University, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
29
|
Veerasubramanian PK, Wynn TA, Quan J, Karlsson FJ. Targeting TNF/TNFR superfamilies in immune-mediated inflammatory diseases. J Exp Med 2024; 221:e20240806. [PMID: 39297883 PMCID: PMC11413425 DOI: 10.1084/jem.20240806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/19/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Dysregulated signaling from TNF and TNFR proteins is implicated in several immune-mediated inflammatory diseases (IMIDs). This review centers around seven IMIDs (rheumatoid arthritis, systemic lupus erythematosus, Crohn's disease, ulcerative colitis, psoriasis, atopic dermatitis, and asthma) with substantial unmet medical needs and sheds light on the signaling mechanisms, disease relevance, and evolving drug development activities for five TNF/TNFR signaling axes that garner substantial drug development interest in these focus conditions. The review also explores the current landscape of therapeutics, emphasizing the limitations of the approved biologics, and the opportunities presented by small-molecule inhibitors and combination antagonists of TNF/TNFR signaling.
Collapse
Affiliation(s)
| | - Thomas A. Wynn
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | - Jie Quan
- Inflammation and Immunology Research Unit, Pfizer, Inc., Cambridge, MA, USA
| | | |
Collapse
|
30
|
Casanova JL, Abel L. The Microbe, the Infection Enigma, and the Host. Annu Rev Microbiol 2024; 78:103-124. [PMID: 38986133 PMCID: PMC11956784 DOI: 10.1146/annurev-micro-092123-022855] [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] [Indexed: 07/12/2024]
Abstract
Human infectious diseases are unique in that the discovery of their environmental trigger, the microbe, was sufficient to drive the development of extraordinarily effective principles and tools for their prevention or cure. This unique medical prowess has outpaced, and perhaps even hindered, the development of scientific progress of equal magnitude in the biological understanding of infectious diseases. Indeed, the hope kindled by the germ theory of disease was rapidly subdued by the infection enigma, in need of a host solution, when it was realized that most individuals infected with most infectious agents continue to do well. The root causes of disease and death in the unhappy few remained unclear. While canonical approaches in vitro (cellular microbiology), in vivo (animal models), and in natura (clinical studies) analyzed the consequences of infection with a microbe, considered to be the cause of disease, in cells, tissues, or organisms seen as a uniform host, alternative approaches searched for preexisting causes of disease, particularly human genetic and immunological determinants in populations of diverse individuals infected with a trigger microbe.
Collapse
Affiliation(s)
- Jean-Laurent Casanova
- Howard Hughes Medical Institute, New York, NY, USA
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
| | - Laurent Abel
- Paris Cité University, Imagine Institute, Paris, France
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA;
| |
Collapse
|
31
|
Nandi S, Varotariya K, Luhana S, Kyada AD, Saha A, Roy N, Sharma N, Rambabu D. GWAS for identification of genomic regions and candidate genes in vegetable crops. Funct Integr Genomics 2024; 24:203. [PMID: 39470821 DOI: 10.1007/s10142-024-01477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 09/24/2024] [Accepted: 10/14/2024] [Indexed: 11/01/2024]
Abstract
Genome-wide association Studies (GWAS), initially developed for human genetics, have been highly effective in plant research, particularly for vegetable crops. GWAS is a robust tool for identifying genes associated with key traits such as yield, nutritional value, disease resistance, adaptability, and bioactive compound biosynthesis. Unlike traditional methods, GWAS does not require prior biological knowledge and can accurately pinpoint loci, minimizing false positives. The process involves developing a diverse panel, rigorous phenotyping and genotyping, and sophisticated statistical analysis using various models and software tools. By scanning the entire genome, GWAS identifies specific loci or single nucleotide polymorphisms (SNPs) linked to target traits. When a causal SNP variant is not directly genotyped, GWAS identifies SNPs in linkage disequilibrium (LD) with the causal variant, mapping the genetic interval. The method begins with careful panel selection, phenotyping, and genotyping, controlling for environmental effects and utilizing Best Linear Unbiased Prediction (BLUP). High-correlation, high-heritability traits are prioritized. Various genotyping methods address confounders like population structure and kinship. Bonferroni correction (BC) prevents false positives, and significant associations are shown in Manhattan plots. Candidate genes are identified through LD analysis and fine mapping, followed by functional validation. GWAS offers critical insights for enhancing vegetable crop breeding efficiency and precision, driving breakthroughs through advanced methods.
Collapse
Affiliation(s)
- Swagata Nandi
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Kishor Varotariya
- Division of Vegetable Science, ICAR-Indian Institute of Horticultural Research, Bengaluru, 560089, India.
| | - Sohamkumar Luhana
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Amitkumar D Kyada
- Division of Genetics, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Ankita Saha
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Nabanita Roy
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Neha Sharma
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| | - Dharavath Rambabu
- Division of Vegetable Science, ICAR-Indian Agricultural Research Institute, New Delhi, 110012, India
| |
Collapse
|
32
|
Agrifoglio O, Kasprick A, Gross N, Wahlig M, Kauffold E, Woitas A, Vorobyev A, Ehlers L, Ludwig RJ, Bieber K, Jaster R. Dexamethasone's Clinical Efficacy in Experimental Autoimmune Pancreatitis Correlates with a Unique Transcriptomic Signature, Whilst Kinase Inhibitors Are Not Effective. Biomedicines 2024; 12:2480. [PMID: 39595046 PMCID: PMC11591683 DOI: 10.3390/biomedicines12112480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/14/2024] [Accepted: 10/26/2024] [Indexed: 11/28/2024] Open
Abstract
(1) Background: Autoimmune pancreatitis (AIP) is mainly treated with steroids. Using an AIP mouse model, we investigated two potential alternatives, the transforming growth factor-β-activated kinase 1 inhibitor, takinib, and the Janus kinase inhibitor, tofacitinib. (2) Methods: In a multicenter preclinical study, MRL/MpJ mice were injected with polyinosinic/polycytidylic acid (poly I:C) for two weeks to induce AIP. They were then treated for four weeks with either takinib (25, 50, or 75 mg/kg body weight), tofacitinib (5, 10 or 15 mg/kg), dexamethasone (1 mg/kg), or solvent, while the poly I:C injections were continued. The severity of AIP was assessed histopathologically. Flow cytometry was used to examine lymphocyte subtypes in the spleen and mesenteric lymph nodes. The pancreatic gene expression profiles were analyzed by RNA sequencing. (3) Results: Poly I:C-treated mice developed severe AIP with inflammation, destruction of acinar tissue, and fibrosis. Dexamethasone significantly attenuated the disease, while takinib or tofacitinib had no effects. Dexamethasone also antagonized the effects of poly I:C on the relative frequencies of the AIP-associated lymphocyte subtypes CD4/CD69, CD8/CD44high, and CD4/CD25/FoxP3 in the spleen. In the principal component analysis of pancreatic transcriptomics, poly I:C-injected mice treated with tofacitinib, takinib, or solvent clustered together, while untreated and dexamethasone-treated mice formed separate, unique clusters. (4) Conclusions: Dexamethasone effectively reduced AIP severity, while takinib and tofacitinib were ineffective. The unique gene expression profile in dexamethasone-treated mice may provide a basis for identifying new drug targets for AIP treatment.
Collapse
Affiliation(s)
- Ottavia Agrifoglio
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Anika Kasprick
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, 23562 Lübeck, Germany
| | - Natalie Gross
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, 23562 Lübeck, Germany
| | - Marc Wahlig
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Emilia Kauffold
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Aline Woitas
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Artem Vorobyev
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Luise Ehlers
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Ralf J. Ludwig
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, 23562 Lübeck, Germany
- Department of Dermatology, University of Lübeck, 23562 Lübeck, Germany
| | - Katja Bieber
- Lübeck Institute of Experimental Dermatology and Center for Research on Inflammation of the Skin, University of Lübeck, 23562 Lübeck, Germany
| | - Robert Jaster
- Department of Medicine II, Division of Gastroenterology and Endocrinology, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
33
|
Huang Z, Liu B, Xiao L, Liao M, Huang L, Zhao X, Ma K, Wang R, Ji F, Li W, Huang L, Xie L. Effects of breast-fed infants-derived Limosilactobacillus reuteri and Bifidobacterium breve ameliorate DSS-induced colitis in mice. iScience 2024; 27:110902. [PMID: 39351200 PMCID: PMC11439849 DOI: 10.1016/j.isci.2024.110902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/11/2024] [Accepted: 09/04/2024] [Indexed: 10/04/2024] Open
Abstract
Studies have shown that breastfeeding can reduce the risk and severity of inflammatory bowel disease (IBD) in children and adults. Probiotics in breast milk have also been isolated and their effects on IBD have been studied. However, based on current evidence, the exact efficacy and mechanisms of probiotics in the treatment of IBD cannot be determined. In this study, Bifidobacterium breve FPHC4024 (BB FPHC4024) and Limosilactobacillus reuteri FPHC2951 (LR FPHC2951) were isolated from feces of exclusively breastfed healthy infants and administered by gavage to dextran sulfate sodium (DSS)-induced IBD mice. The results showed that LR FPHC2951 improved the symptoms of DSS-induced IBD, increased the expression of interleukin (IL)-10 mRNA and upregulated the abundance of Verrucomicrobiaceae Akkermansia. Combined with Kyoto Encyclopedia of Genes and Genomes (KEGG)-based Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) function prediction results, we hypothesized that LR FPHC2951 improved DSS-induced colitis symptoms in mice by increasing of IL-10 mRNA, altering the structure of intestinal flora, and reducing proinflammatory pathways and enhancing pathways associated with anti-inflammatory and intestinal protection.
Collapse
Affiliation(s)
- Zhipeng Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Bingdong Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lanlin Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Miaomiao Liao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
| | - Liujing Huang
- Medical Affairs Department, Guangzhou Betrue Technology Co., Ltd. Guangzhou 510700, China
| | - Xiaogan Zhao
- Nanjing Agricultural University College of Food Science and Technology, Nanjing, Jiangsu, China
| | - Kai Ma
- Jiangsu New-bio Biotechnology Co., Ltd., Jiangyin, China
| | - Runxin Wang
- Jiangsu New-bio Biotechnology Co., Ltd., Jiangyin, China
| | - Feng Ji
- Jiangsu New-bio Biotechnology Co., Ltd., Jiangyin, China
| | - Wei Li
- Nanjing Agricultural University College of Food Science and Technology, Nanjing, Jiangsu, China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Liwei Xie
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China
- College of Life and Health Sciences, Guangdong Industry Polytechnic University, Guangzhou, Guangdong 510300, China
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Internal Medicine, Shunde Women and Children’s Hospital (Maternity and Child Healthcare Hospital of Shunde Foshan), Guangdong Medical University, Foshan, Guangdong, China
| |
Collapse
|
34
|
Chen R, Huang Q, Rao Y, Wang J, Yu R, Peng S, Huang K, Huang Y, Zhu X, Tang D, Zhang X, Lin T, Chen T, Yan A. Genomic and Transcriptional Analysis of the Necroptosis Pathway Elements RIPK and MLKL in Sea Cucumber, Holothuria leucospilota. Genes (Basel) 2024; 15:1297. [PMID: 39457421 PMCID: PMC11507063 DOI: 10.3390/genes15101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Receptor-interacting protein kinases (RIPKs) and mixed-lineage kinase domain-like protein (MLKL) are crucial in regulating innate immune responses and cell death signaling (necroptosis and apoptosis), and are potential candidates for genetic improvement in breeding programs. Knowledge about the RIPK family and MLKL in sea cucumber remains limited. Methods: We searched the genomes of sea cucumber Holothuria leucospilota for genes encoding RIPKs and MLKL, performed phylogenetic tree, motif and functional domain analyses, and examined tissue distribution and embryonic development patterns using qPCR. Results: RIPK5 (Hl-RIPK5), RIPK7 (Hl-RIPK7) and MLKL (Hl-MLKL) were identified in sea cucumber H. leucospilota. Hl-RIPK5 and Hl-RIPK7 were mainly expressed in coelomocytes, suggesting that they play a role in innate immunity, whereas Hl-MLKL exhibited relatively low expression across tissues. During embryonic development, Hl-MLKL was highly expressed from the 2-cell stage to the morula stage, while Hl-RIPK5 and Hl-RIPK7 were primarily expressed after the morula stage, indicating different roles in embryonic development. In primary coelomocytes, Hl-RIPK5 transcriptional activity was significantly depressed by LPS, poly(I:C), or pathogen Vibrio harveyi. Hl-RIPK7 expression levels were unchanged following the same challenges. Hl-MLKL mRNA levels were significantly decreased with poly(I:C) or V. harveyi, but did not change with LPS. Conclusions: These findings provide valuable insights into the evolutionary tree and characterization of RIPK and MLKL genes in sea cucumber, contributing to the broader understanding of the RIPK gene family and MLKL in ancient echinoderms.
Collapse
Affiliation(s)
- Rong Chen
- Mangrove Rare and Endangered Species Protection and Utilization Engineering Technology Research Center, Institute of Applied Biotechnology, School of Life Science and Technolog, Lingnan Normal University, Zhanjiang 528048, China; (R.C.); (Y.R.)
| | - Qianying Huang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Yingzhu Rao
- Mangrove Rare and Endangered Species Protection and Utilization Engineering Technology Research Center, Institute of Applied Biotechnology, School of Life Science and Technolog, Lingnan Normal University, Zhanjiang 528048, China; (R.C.); (Y.R.)
| | - Junyan Wang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Ruiming Yu
- School of Global Public Health, New York University, New York, NY 10012, USA;
| | - Shuangxin Peng
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Kaiyi Huang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Yihang Huang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Xiangxing Zhu
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Dongsheng Tang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Xiaoli Zhang
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| | - Tiehao Lin
- Guangdong Institute for Drug Control, Guangzhou 5106630, China;
| | - Ting Chen
- Key Laboratory of Breeding Biotechnology and Sustainable Aquaculture (CAS), Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China;
- Research Centre on Aquaculture Nutrition and Environmental Ecology of the Ministry of Agriculture and Rural Affair, Shanghai Ocean University, Shanghai 201306, China
| | - Aifen Yan
- School of Medicine, Foshan University, Foshan 528000, China; (Q.H.); (J.W.); (S.P.); (K.H.); (Y.H.); (X.Z.); (D.T.); (X.Z.)
| |
Collapse
|
35
|
Deng L, He K, Zhang X. Joint mirror procedure: controlling false discovery rate for identifying simultaneous signals. Biometrics 2024; 80:ujae142. [PMID: 39671277 PMCID: PMC11639532 DOI: 10.1093/biomtc/ujae142] [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: 11/03/2023] [Revised: 08/02/2024] [Accepted: 11/11/2024] [Indexed: 12/15/2024]
Abstract
In many applications, the process of identifying a specific feature of interest often involves testing multiple hypotheses for their joint statistical significance. Examples include mediation analysis, which simultaneously examines the existence of the exposure-mediator and the mediator-outcome effects, and replicability analysis, aiming to identify simultaneous signals that exhibit statistical significance across multiple independent studies. In this work, we present a new approach called the joint mirror (JM) procedure that effectively detects such features while maintaining false discovery rate (FDR) control in finite samples. The JM procedure employs an iterative method that gradually shrinks the rejection region based on progressively revealed information until a conservative estimate of the false discovery proportion is below the target FDR level. Additionally, we introduce a more stringent error measure known as the composite FDR (cFDR), which assigns weights to each false discovery based on its number of null components. We use the leave-one-out technique to prove that the JM procedure controls the cFDR in finite samples. To implement the JM procedure, we propose an efficient algorithm that can incorporate partial ordering information. Through extensive simulations, we show that our procedure effectively controls the cFDR and enhances statistical power across various scenarios, including the case that test statistics are dependent across the features. Finally, we showcase the utility of our method by applying it to real-world mediation and replicability analyses.
Collapse
Affiliation(s)
- Linsui Deng
- School of Data Science, The Chinese University of Hong Kong, Shenzhen 518172, China
- Center for Applied Statistics, Institute of Statistics and Big Data, Renmin University of China, Beijing 100872, China
| | - Kejun He
- Center for Applied Statistics, Institute of Statistics and Big Data, Renmin University of China, Beijing 100872, China
| | - Xianyang Zhang
- Department of Statistics, Texas A&M University, College Station, TX 77843, United States
| |
Collapse
|
36
|
Nakamura H, Hikichi H, Seto S, Hijikata M, Keicho N. Transcriptional regulators SP110 and SP140 modulate inflammatory response genes in Mycobacterium tuberculosis-infected human macrophages. Microbiol Spectr 2024; 12:e0010124. [PMID: 39162523 PMCID: PMC11448263 DOI: 10.1128/spectrum.00101-24] [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: 01/15/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
Understanding the functions of human transcriptional regulatory genes SP110 and SP140 during Mycobacterium tuberculosis infection is crucial; in a mouse model, homologous genes Sp110 and Sp140 have been shown to negatively regulate inflammatory response genes, including the type I interferon (IFN) response. The reduction of these genes in mice is associated with susceptibility to M. tuberculosis infection and the development of necrotizing granulomatous lesions. To investigate the involvement of SP110 and SP140 in human inflammatory response, we analyzed their regulatory manner in THP-1 macrophages infected with M. tuberculosis. Genome-wide transcriptional profiling revealed that the depletion of SP110 and/or SP140 impaired the induction of gene expression associated with inflammatory responses, including IFN response genes, although it had little effect on the intracellular proliferation of M. tuberculosis. By contrast, genes related to phosphorylation were upregulated in infected macrophages with SP110 and/or SP140 knockdown, but downregulated in infected control macrophages without their knockdown. Reverse transcription-quantitative PCR and ELISA further confirmed the impairment of the induction of IFN response genes by the depletion of SP110 and/or SP140 in M. tuberculosis-infected macrophages. These findings suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses in M. tuberculosis-infected macrophages. IMPORTANCE Tuberculosis (TB) is one of the most serious infectious diseases, with high morbidity and mortality worldwide. C3HeB/FeJ mice are widely utilized for evaluating anti-TB drugs because their drug sensitivity and pathology during M. tuberculosis infection resemble those of human TB, including the development of necrotizing granulomas. Downregulation of the transcriptional regulatory genes Sp110 and Sp140 in C3HeB/FeJ mice has been demonstrated to activate gene expression associated with inflammatory responses during M. tuberculosis infection, resulting in susceptibility to the infection. Here, we examined the regulatory manner of SP110 and SP140 using transcriptomic analysis in M. tuberculosis-infected human macrophages. Depletion of SP110 and/or SP140 in M. tuberculosis-infected THP-1 macrophages impaired the induction of gene expression associated with inflammatory responses, including interferon response genes, compared with that in control macrophages. These results suggest that human SP110 and SP140 act as positive regulators for genes associated with inflammatory responses upon M. tuberculosis infection.
Collapse
Affiliation(s)
- Hajime Nakamura
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Haruka Hikichi
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shintaro Seto
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Minako Hijikata
- Department of Pathophysiology and Host Defense, The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| | - Naoto Keicho
- Department of Basic Mycobacteriosis, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- The Research Institute of Tuberculosis, Japan Anti-Tuberculosis Association, Tokyo, Japan
| |
Collapse
|
37
|
Hitomi Y, Ueno K, Aiba Y, Nishida N, Kono M, Sugihara M, Kawai Y, Kawashima M, Khor SS, Sugi K, Kouno H, Kohno H, Naganuma A, Iwamoto S, Katsushima S, Furuta K, Nikami T, Mannami T, Yamashita T, Ario K, Komatsu T, Makita F, Shimada M, Hirashima N, Yokohama S, Nishimura H, Sugimoto R, Komura T, Ota H, Kojima M, Nakamuta M, Fujimori N, Yoshizawa K, Mano Y, Takahashi H, Hirooka K, Tsuruta S, Sato T, Yamasaki K, Kugiyama Y, Motoyoshi Y, Suehiro T, Saeki A, Matsumoto K, Nagaoka S, Abiru S, Yatsuhashi H, Ito M, Kawata K, Takaki A, Arai K, Arinaga-Hino T, Abe M, Harada M, Taniai M, Zeniya M, Ohira H, Shimoda S, Komori A, Tanaka A, Ishigaki K, Nagasaki M, Tokunaga K, Nakamura M. A genome-wide association study identified PTPN2 as a population-specific susceptibility gene locus for primary biliary cholangitis. Hepatology 2024; 80:776-790. [PMID: 38652555 DOI: 10.1097/hep.0000000000000894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 02/22/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND AND AIMS Previous genome-wide association studies (GWAS) have indicated the involvement of shared (population-nonspecific) and nonshared (population-specific) susceptibility genes in the pathogenesis of primary biliary cholangitis (PBC) among European and East-Asian populations. Although a meta-analysis of these distinct populations has recently identified more than 20 novel PBC susceptibility loci, analyses of population-specific genetic architecture are still needed for a more comprehensive search for genetic factors in PBC. APPROACH AND RESULTS Protein tyrosine phosphatase nonreceptor type 2 ( PTPN2) was identified as a novel PBC susceptibility gene locus through GWAS and subsequent genome-wide meta-analysis involving 2181 cases and 2699 controls from the Japanese population (GWAS-lead variant: rs8098858, p = 2.6 × 10 -8 ). In silico and in vitro functional analyses indicated that the risk allele of rs2292758, which is a primary functional variant, decreases PTPN2 expression by disrupting Sp1 binding to the PTPN2 promoter in T follicular helper cells and plasmacytoid dendritic cells. Infiltration of PTPN2-positive T-cells and plasmacytoid dendritic cells was confirmed in the portal area of the PBC liver by immunohistochemistry. Furthermore, transcriptomic analysis of PBC-liver samples indicated the presence of a compromised negative feedback loop in vivo between PTPN2 and IFNG in patients carrying the risk allele of rs2292758. CONCLUSIONS PTPN2 , a novel susceptibility gene for PBC in the Japanese population, may be involved in the pathogenesis of PBC through an insufficient negative feedback loop caused by the risk allele of rs2292758 in IFN-γ signaling. This suggests that PTPN2 could be a potential molecular target for PBC treatment.
Collapse
Affiliation(s)
- Yuki Hitomi
- Department of Human Genetics, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kazuko Ueno
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yoshihiro Aiba
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Nao Nishida
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
- Department of Genomic Function and Diversity, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Michihiro Kono
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Mitsuki Sugihara
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yosuke Kawai
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Seik-Soon Khor
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Kazuhiro Sugi
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Hirotaka Kouno
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Hiroshi Kohno
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Atsushi Naganuma
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Satoru Iwamoto
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Shinji Katsushima
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kiyoshi Furuta
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Toshiki Nikami
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Tomohiko Mannami
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Tsutomu Yamashita
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Keisuke Ario
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Tatsuji Komatsu
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Fujio Makita
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Masaaki Shimada
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Noboru Hirashima
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Shiro Yokohama
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Hideo Nishimura
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Rie Sugimoto
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Takuya Komura
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Hajime Ota
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Motoyuki Kojima
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Makoto Nakamuta
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Naoyuki Fujimori
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kaname Yoshizawa
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Yutaka Mano
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Hironao Takahashi
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kana Hirooka
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Satoru Tsuruta
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Takeaki Sato
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kazumi Yamasaki
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Yuki Kugiyama
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | | | - Tomoyuki Suehiro
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Akira Saeki
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kosuke Matsumoto
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Shinya Nagaoka
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Seigo Abiru
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | | | - Masahiro Ito
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Akinobu Takaki
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kuniaki Arai
- Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan
| | - Teruko Arinaga-Hino
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Masanori Abe
- Department of Gastroenterology and Metabology, Ehime University Graduate School of Medicine, Matsuyama, Japan
| | - Masaru Harada
- The Third Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Makiko Taniai
- Department of Medicine and Gastroenterology, Tokyo Women's Medical University, Tokyo, Japan
| | - Mikio Zeniya
- Department of Gastroenterology and Hepatology, Tokyo Jikei University School of Medicine, Tokyo, Japan
| | - Hiromasa Ohira
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Japan
| | - Shinji Shimoda
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Hirakata, Japan
| | - Atsumasa Komori
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
- Department of Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Omura, Japan
| | - Atsushi Tanaka
- Department of Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Masao Nagasaki
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Human Biosciences Unit for the Top Global Course Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Minoru Nakamura
- Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Headquarters of PBC Research in NHO Study Group for Liver Disease in Japan (NHOSLJ), Clinical Research Center, NHO Nagasaki Medical Center, Omura, Japan
- Department of Hepatology, Nagasaki University Graduate School of Biomedical Sciences, Omura, Japan
| |
Collapse
|
38
|
Saha S, Chatterjee P, Nasipuri M, Basu S, Chakraborti T. Computational drug repurposing for viral infectious diseases: a case study on monkeypox. Brief Funct Genomics 2024; 23:570-578. [PMID: 38183212 DOI: 10.1093/bfgp/elad058] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
The traditional method of drug reuse or repurposing has significantly contributed to the identification of new antiviral compounds and therapeutic targets, enabling rapid response to developing infectious illnesses. This article presents an overview of how modern computational methods are used in drug repurposing for the treatment of viral infectious diseases. These methods utilize data sets that include reviewed information on the host's response to pathogens and drugs, as well as various connections such as gene expression patterns and protein-protein interaction networks. We assess the potential benefits and limitations of these methods by examining monkeypox as a specific example, but the knowledge acquired can be applied to other comparable disease scenarios.
Collapse
Affiliation(s)
- Sovan Saha
- Department of Computer Science and Engineering (Artificial Intelligence and Machine Learning), Techno Main Salt Lake, EM-4/1, Sector V, Bidhannagar, Kolkata, West Bengal 700091, India
| | - Piyali Chatterjee
- Department of Computer Science and Engineering, Netaji Subhash Engineering College, Garia, Kolkata-700152, India
| | - Mita Nasipuri
- Department of Computer Science and Engineering, Jadavpur University, Kolkata - 700032, India
| | - Subhadip Basu
- Department of Computer Science and Engineering, Jadavpur University, Kolkata - 700032, India
| | - Tapabrata Chakraborti
- Department of Medical Physics and Biomedical Engineering, University College London, UK
- Health Science Programme, The Alan Turing Institute, London, UK
- Linacre College, University of Oxford, UK
| |
Collapse
|
39
|
Baumgart DC, Stallmach A, Grunert P, Schubert S, Howaldt S, von Arnim U, Ochsenkühn T, Stein J, Lügering A, Schmidt D, Schulz M, Fischer A. Induction and maintenance of mucosal healing in Crohn's disease with ustekinumab in clinical practice across all care levels in Germany (MUCUS). Sci Rep 2024; 14:20502. [PMID: 39227642 PMCID: PMC11371836 DOI: 10.1038/s41598-024-70241-9] [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: 06/06/2024] [Accepted: 08/14/2024] [Indexed: 09/05/2024] Open
Abstract
The impact of ustekinumab (UST) on mucosal- and fistula healing and extraintestinal manifestations (EIM) in Crohn's disease (CD) were not fully elucidated in the registration trials. In this prospective, multicenter study (EudraCT number: 2017-005151-83) we evaluated the German label real-world-effectiveness of UST to achieve the primary endpoint of combined clinical and endoscopic response at week 52 and several secondary endpoints. Of 79 screened we enrolled 52 patients (female n = 28, bionaïve n = 13, biologic n = 39). At week 52 (per protocol analysis), 52% (n = 13/25) of patients achieved the primary endpoint [50% (n = 3/6) in the bionaïve, 45.5% (n = 5/11) biologic, 62.5% (n = 5/8 ) multiple biologics cohorts, respectively with age as independent predictor [OR 95% CI 0.933 (0.873, 0.998) p = 0.043], 60% (n = 15/25) achieved endoscopic response [50% (n = 3/6) in the bionaïve, 54.5% (n = 6/11) biologic, 75% (n = 6/8) multiple biologics cohorts, respectively], 36% (n = 9/25) achieved endoscopic remission [50% (n = 3/6) in the bionaïve, 27.3% (n = 3/11) biologic, 37.5% (n = 3/8) multiple biologics cohorts, respectively], 48% (n = 12/25) achieved mucosal healing [50% (n = 3/6) in the bionaïve, 36.4% (n = 4/11) biologic, 62.5% (n = 5/8) multiple biologics cohorts, respectively]. All achieved a fistula response and 33.3% (n = 1/3) in the multiple biologics group fistula remission at week 52. EIM decreased (week 0 28.2% vs. week 52 8%). CRP, FCP, PRO-2, EQ-5D-5L improved throughout. 36 patients (69.2%) experienced ≥ 1 treatment emergent adverse event, in 8 (15.4%) cases rated as severe and in 5 (9.6%) leading to UST discontinuation, but no very severe events or deaths. The effectiveness of UST was better than in the registration trials.
Collapse
Affiliation(s)
- Daniel C Baumgart
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| | - Andreas Stallmach
- Klinik für Innere Medizin IV Gastroenterologie, Hepatologie, Infektiologie, Zentrale Endoskopie, Universitätsklinikum Jena, Jena, Germany
| | - Philip Grunert
- Klinik für Innere Medizin IV Gastroenterologie, Hepatologie, Infektiologie, Zentrale Endoskopie, Universitätsklinikum Jena, Jena, Germany
| | - Stefan Schubert
- Gastroenterologie am Bayerischen Platz, Gastroenterologie, Berlin, Germany
| | - Stefanie Howaldt
- Hamburgisches Forschungsinstitut für chronisch entzündliche Darmerkrankungen, Immunologie, Hamburg, Germany
| | - Ulrike von Arnim
- Universitätsklinikum Magdeburg, Universitätsklinik für Gastroenterologie, Hepatologie und Infektiologie, Magdeburg, Germany
| | - Thomas Ochsenkühn
- Isarklinikum, Klinik für Gastroenterologie, Hepatologie und Gastroenterologische Onkologie, München, Germany
| | - Jürgen Stein
- DGD Kliniken Frankfurt Sachsenhausen, Abteilung Gastroenterologie/Ernährungsmedizin, Frankfurt am Main, Germany
| | - Andreas Lügering
- MVZ Portal 10, Zentrum für Leber-, Magen- und Darmerkrankungen, Münster, Germany
| | - Daniel Schmidt
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marten Schulz
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Andreas Fischer
- Medizinische Klinik m.S. Hepatologie und Gastroenterologie, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
40
|
Lee YM, Vucic D. The role of autophagy in RIP1 mediated cell death and intestinal inflammation. Adv Immunol 2024; 163:1-20. [PMID: 39271257 DOI: 10.1016/bs.ai.2024.07.003] [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: 09/15/2024]
Abstract
Autophagy, a highly conserved catabolic process that targets various types of cellular cargoes to lysosomal degradation, is one of the most important biological mechanisms critical for cellular homeostasis. Components of these cellular cargoes can range from individual proteins to invading pathogens, and degrading these materials is important for maintaining organismal health and survival. The process of autophagy is carried out by complex molecular mechanisms, and a growing body of evidence indicates that these mechanisms intersect with those involved in the cell death pathways. In this review, we examine several emerging studies elucidating the role of autophagy in RIP1-mediated cell death signaling, with particular emphasis on impaired autophagy caused by ATG16L1 deficiency. We also discuss how autophagy in RIP1-mediated cell death affects intestinal homeostasis in preclinical models, and the implications of the intersection between RIP1 and autophagy for understanding the intestinal pathologies associated with inflammatory bowel disease (IBD). Finally, we highlight the potential benefits of therapeutic targeting of RIP1 and autophagy proteins, while also proposing areas of research that will likely elucidate new links between autophagy and cell death signaling.
Collapse
Affiliation(s)
| | - Domagoj Vucic
- Immunology Discovery, Genentech, South San Francisco, CA, United States.
| |
Collapse
|
41
|
Akkoç T. Epithelial barrier dysfunction and microbial dysbiosis: exploring the pathogenesis and therapeutic strategies for Crohn's disease. Tissue Barriers 2024:2390705. [PMID: 39185541 DOI: 10.1080/21688370.2024.2390705] [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: 06/13/2024] [Revised: 08/05/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Crohn's disease (CD), a chronic gastrointestinal inflammatory disease, is becoming more widespread worldwide. Crohn's disease is caused by gut microbiota changes, genetics, environmental stresses, and immunological responses. Current treatments attempt to achieve long-term remission and avoid complications, delaying disease progression. Immunosuppressive measures and combination medicines should be started early for high-risk patients. These medicines monitor inflammatory indicators and adjust as needed. The epithelial barrier helps defend against physical, chemical, and immunological threats. When tissues' protective barrier breaks down, the microbiome may reach the layer underneath. Unbalanced microbial populations and inflammation impair healing and adjustment. Inflammatory cells infiltrating sensitive tissues aggravate the damage and inflammation. This approach promotes chronic inflammatory diseases. The epithelial barrier hypothesis states that hereditary and environmental variables cause epithelial tissue inflammation. This review focuses on how epithelial barrier break-down and microbial dysbiosis cause Crohn's disease and current advances in understanding the epithelial barrier, immune system, and microbiome. Additionally, investigate treatments that restore barrier integrity and promote microbial balance. Overall, it stresses the role of epithelial barrier failure and microbial dysbiosis in Crohn's disease development and discusses current advances in understanding the barrier, immunological responses, and microbiota.
Collapse
Affiliation(s)
- Tunç Akkoç
- Department of Immunology, Marmara University School of Medicine, İstanbul, Türkiye
- Division of Pediatric Allergy and Immunology, Marmara University School of Medicine, İstanbul, Türkiye
| |
Collapse
|
42
|
Sæterstad S, Østvik AE, Hansen MD, Bruland T, van Beelen Granlund A. The effect of rs2910686 on ERAP2 expression in IBD and epithelial inflammatory response. J Transl Med 2024; 22:750. [PMID: 39123229 PMCID: PMC11316291 DOI: 10.1186/s12967-024-05532-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: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND ERAP2 is an aminopeptidase involved in antigen processing and presentation, and harbor genetic variants linked to several inflammatory diseases such as Inflammatory Bowel Disease (IBD). The lack of an ERAP2 gene homologue in mice has hampered functional studies, and most human studies have focused on cells of hematopoietic origin. Using an IBD biobank as vantage point, this study explores how genetic variation in ERAP2 affects gene expression in human-derived epithelial organoids upon proinflammatory stimulation. METHODS An IBD patient cohort was genotyped with regards to two single nucleotide polymorphisms (SNP) (rs2910686/rs2248374) associated with ERAP2 expression levels, and we examined the correlation between colon gene expression and genotype, specifically aiming to establish a relationship with ERAP2 expression proficiency. Human-derived colon organoids (colonoids) with known ERAP2 genotype were established and used to explore differences in whole genome gene expression between ERAP2-deficient (n = 4) and -proficient (n = 4) donors upon pro-inflammatory encounter. RESULTS When taking rs2910686 genotype into account, ERAP2 gene expression is upregulated in the inflamed colon of IBD patients. Colonoids upregulate ERAP2 upon IFNɣ stimulation, and ERAP2 expression proficiency is dependent on rs2910686 genotype. Colonoid genotyping confirms that mechanisms independent of the frequently studied SNP rs2248374 can cause ERAP2-deficiency. A total of 586 genes involved in various molecular mechanisms are differentially expressed between ERAP2 proficient- and deficient colonoids upon proinflammatory stimulation, including genes encoding proteins with the following molecular function: catalytic activity (AOC1, CPE, ANPEP and MEP1A), regulator activity (TNFSF9, MDK, GDF15, ILR6A, LGALS3 and FLNA), transmembrane transporter activity (SLC40A1 and SLC5A1), and extracellular matrix structural constituents (FGL2, HMCN2, and MUC17). CONCLUSIONS ERAP2 is upregulated in the inflamed IBD colon mucosa, and expression proficiency is highly correlated with genotype of rs2910686. While the SNP rs2248374 is commonly used to determine ERAP2 expressional proficiency, our data confirms that mechanisms independent of this SNP can lead to ERAP2 deficiency. Our data demonstrates that epithelial ERAP2 presence affects the inflammatory response in colonoids, suggesting a pleiotropic role of ERAP2 beyond MHC class I antigen processing.
Collapse
Affiliation(s)
- Siri Sæterstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann Elisabeth Østvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Marianne Doré Hansen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway.
- Department of Pathology, St. Olav's University Hospital, Trondheim, Norway.
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
43
|
Dziulko AK, Allen H, Chuong EB. An endogenous retrovirus regulates tumor-specific expression of the immune transcriptional regulator SP140. Hum Mol Genet 2024; 33:1454-1464. [PMID: 38751339 PMCID: PMC11305685 DOI: 10.1093/hmg/ddae084] [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: 02/27/2024] [Revised: 04/24/2024] [Accepted: 05/07/2024] [Indexed: 07/26/2024] Open
Abstract
Speckled Protein 140 (SP140) is a chromatin reader with critical roles regulating immune cell transcriptional programs, and SP140 splice variants are associated with immune diseases including Crohn's disease, multiple sclerosis, and chronic lymphocytic leukemia. SP140 expression is currently thought to be restricted to immune cells. However, by analyzing human transcriptomic datasets from a wide range of normal and cancer cell types, we found recurrent cancer-specific expression of SP140, driven by an alternative intronic promoter derived from an intronic endogenous retrovirus (ERV). The ERV belongs to the primate-specific LTR8B family and is regulated by oncogenic mitogen-activated protein kinase (MAPK) signaling. The ERV drives expression of multiple cancer-specific isoforms, including a nearly full-length isoform that retains all the functional domains of the full-length canonical isoform and is also localized within the nucleus, consistent with a role in chromatin regulation. In a fibrosarcoma cell line, silencing the cancer-specific ERV promoter of SP140 resulted in increased sensitivity to interferon-mediated cytotoxicity and dysregulation of multiple genes. Our findings implicate aberrant ERV-mediated SP140 expression as a novel mechanism contributing to immune gene dysregulation in a wide range of cancer cells.
Collapse
Affiliation(s)
- Adam K Dziulko
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, JSC Biotech Bldg, Boulder, Colorado 80303, USA
| | - Holly Allen
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, JSC Biotech Bldg, Boulder, Colorado 80303, USA
| | - Edward B Chuong
- Department of Molecular, Cellular, and Developmental Biology and BioFrontiers Institute, University of Colorado Boulder, 3415 Colorado Ave, JSC Biotech Bldg, Boulder, Colorado 80303, USA
| |
Collapse
|
44
|
Shehab M, De Marco D, Lakatos PL, Bessissow T. The potential for medical therapies to address fistulizing Crohn's disease: a state-of-the-art review. Expert Opin Biol Ther 2024; 24:733-746. [PMID: 39045643 DOI: 10.1080/14712598.2024.2383882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/25/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Crohn's disease (CD) is a chronic, relapsing immune mediated disease, which is one of the two major types of inflammatory bowel disease (IBD). Fistulizing CD poses a significant clinical challenge for physicians. Effective management of CD requires a multidisciplinary approach, involving a gastroenterologist and a GI surgeon while tailoring treatment to each patient's unique risk factors, clinical representations, and preferences. AREAS COVERED This comprehensive review explores the intricacies of fistulizing CD including its manifestations, types, impact on quality of life, management strategies, and novel therapies under investigation. EXPERT OPINION Antibiotics are often used as first-line therapy to treat symptoms. Biologics that selectively target TNF-α, such infliximab (IFX), have shown high efficacy in randomized controlled trials. However, more than 50% of patients lose response to IFX, prompting them to explore alternative strategies. Current options include adalimumab and certolizumab pegol combination therapies, as well as small-molecule drugs targeting Janus kinases such as Upadacitinib. Furthermore, a promising treatment for complex fistulas is mesenchymal stem cells such as Darvadstrocel (Alofisel), an allogeneic stem cell-based therapy. However, surgical interventions are necessary for complex cases or intra-abdominal complications. Setons and LIFT procedures are the most common surgical options.
Collapse
Affiliation(s)
- Mohammad Shehab
- Division of Gastroenterology, Department of Internal Medicine, Mubarak Al-Kabeer University Hospital, Kuwait University, Kuwait City, Kuwait
| | - Davide De Marco
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
| | - Peter L Lakatos
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
- 1st Department of Medicine, Semmelweis University, Budapest, Hungary
| | - Talat Bessissow
- Division of Gastroenterology and Hepatology, Department of Medicine, McGill University Health Center, Montreal, Canada
| |
Collapse
|
45
|
Quiniou G, Andromaque L, Duclaux-Loras R, Dinet O, Cervantes O, Verdet M, Meunier C, Boschetti G, Viret C, Nancey S, Faure M, Rozières A. Impaired reprogramming of the autophagy flux in maturing dendritic cells from crohn disease patients with core autophagy gene-related polymorphisms. Autophagy 2024; 20:1837-1853. [PMID: 38615686 PMCID: PMC11262231 DOI: 10.1080/15548627.2024.2338574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/16/2024] [Accepted: 03/29/2024] [Indexed: 04/16/2024] Open
Abstract
Crohn disease (CD) is an inflammatory bowel disease whose pathogenesis involves inappropriate immune responses toward gut microbiota on genetically predisposed backgrounds. Notably, CD is associated with single-nucleotide polymorphisms affecting several genes involved in macroautophagy/autophagy, the catabolic process that ensures the degradation and recycling of cytosolic components and microorganisms. In a clinical translation perspective, monitoring the autophagic activity of CD patients will require some knowledge on the intrinsic functional status of autophagy. Here, we focused on monocyte-derived dendritic cells (DCs) to characterize the intrinsic quantitative features of the autophagy flux. Starting with DCs from healthy donors, we documented a reprogramming of the steady state flux during the transition from the immature to mature status: both the autophagosome pool size and the flux were diminished at the mature stage while the autophagosome turnover remained stable. At the cohort level, DCs from CD patients were comparable to control in term of autophagy flux reprogramming capacity. However, the homozygous presence of ATG16L1 rs2241880 A>G (T300A) and ULK1 rs12303764 (G/T) polymorphisms abolished the capacity of CD patient DCs to reprogram their autophagy flux during maturation. This effect was not seen in the case of CD patients heterozygous for these polymorphisms, revealing a gene dose dependency effect. In contrast, the NOD2 rs2066844 c.2104C>T (R702W) polymorphism did not alter the flux reprogramming capacity of DCs. The data, opening new clinical translation perspectives, indicate that polymorphisms affecting autophagy-related genes can differentially influence the capacity of DCs to reprogram their steady state autophagy flux when exposed to proinflammatory challenges.Abbreviation: BAFA1: bafilomycin A1, CD: Crohn disease; DC: dendritic cells; HD: healthy donor; iDCs: immature DCs; IL: interleukin; J: autophagosome flux; LPS: lipopolysaccharide; MHC: major histocompatibility complex; nA: autophagosome pool size; SNPs: single-nucleotide polymorphisms; PCA: principal component analysis; TLR: toll like receptor; τ: transition time; TNF: tumor necrosis factor.
Collapse
Affiliation(s)
- Gaëlle Quiniou
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Leslie Andromaque
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Rémi Duclaux-Loras
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Pediatric Hepatology, Gastroenterology and Nutrition, Femme-Mère-Enfant Hospital, Hospices Civils de Lyon, Bron, France
| | - Océane Dinet
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Ornella Cervantes
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Mallorie Verdet
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Camille Meunier
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Gilles Boschetti
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| | - Stéphane Nancey
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Department of Gastroenterology, Lyon-Sud university hospital, Lyon, France
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM, France
| | - Aurore Rozières
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, Lyon, France
| |
Collapse
|
46
|
Fougiaxis V, He B, Khan T, Vatinel R, Koutroumpa NM, Afantitis A, Lesire L, Sierocki P, Deprez B, Deprez-Poulain R. ERAP Inhibitors in Autoimmunity and Immuno-Oncology: Medicinal Chemistry Insights. J Med Chem 2024; 67:11597-11621. [PMID: 39011823 DOI: 10.1021/acs.jmedchem.4c00840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Endoplasmic reticulum aminopeptidases ERAP1 and 2 are intracellular aminopeptidases that trim antigenic precursors and generate antigens presented by major histocompatibility complex class I (MHC-I) molecules. They thus modulate the antigenic repertoire and drive the adaptive immune response. ERAPs are considered as emerging targets for precision immuno-oncology or for the treatment of autoimmune diseases, in particular MHC-I-opathies. This perspective covers the structural and biological characterization of ERAP, their relevance to these diseases and the ongoing research on small-molecule inhibitors. We describe the chemical and pharmacological space explored by medicinal chemists to exploit the potential of these targets given their localization, biological functions, and family depth. Specific emphasis is put on the binding mode, potency, selectivity, and physchem properties of inhibitors featuring diverse scaffolds. The discussion provides valuable insights for the future development of ERAP inhibitors and analysis of persisting challenges for the translation for clinical applications.
Collapse
Affiliation(s)
- Vasileios Fougiaxis
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
| | - Ben He
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
| | - Tuhina Khan
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
- European Genomic Institute for Diabetes, EGID, University of Lille, F-59000 Lille, France
| | - Rodolphe Vatinel
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
| | | | | | - Laetitia Lesire
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
- European Genomic Institute for Diabetes, EGID, University of Lille, F-59000 Lille, France
| | - Pierre Sierocki
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
- European Genomic Institute for Diabetes, EGID, University of Lille, F-59000 Lille, France
| | - Benoit Deprez
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
- European Genomic Institute for Diabetes, EGID, University of Lille, F-59000 Lille, France
| | - Rebecca Deprez-Poulain
- U1177 - Drugs and Molecules for Living Systems, Univ. Lille, Inserm, Institut Pasteur de Lille, F-59000 Lille, France
- European Genomic Institute for Diabetes, EGID, University of Lille, F-59000 Lille, France
| |
Collapse
|
47
|
Muro P, Zhang L, Li S, Zhao Z, Jin T, Mao F, Mao Z. The emerging role of oxidative stress in inflammatory bowel disease. Front Endocrinol (Lausanne) 2024; 15:1390351. [PMID: 39076514 PMCID: PMC11284038 DOI: 10.3389/fendo.2024.1390351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/19/2024] [Indexed: 07/31/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated condition that affects the digestive system and includes Crohn's disease (CD) and ulcerative colitis (UC). Although the exact etiology of IBD remains uncertain, dysfunctional immunoregulation of the gut is believed to be the main culprit. Amongst the immunoregulatory factors, reactive oxygen species (ROS) and reactive nitrogen species (RNS), components of the oxidative stress event, are produced at abnormally high levels in IBD. Their destructive effects may contribute to the disease's initiation and propagation, as they damage the gut lining and activate inflammatory signaling pathways, further exacerbating the inflammation. Oxidative stress markers, such as malondialdehyde (MDA), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and serum-free thiols (R-SH), can be measured in the blood and stool of patients with IBD. These markers are elevated in patients with IBD, and their levels correlate with the severity of the disease. Thus, oxidative stress markers can be used not only in IBD diagnosis but also in monitoring the response to treatment. It can also be targeted in IBD treatment through the use of antioxidants, including vitamin C, vitamin E, glutathione, and N-acetylcysteine. In this review, we summarize the role of oxidative stress in the pathophysiology of IBD, its diagnostic targets, and the potential application of antioxidant therapies to manage and treat IBD.
Collapse
Affiliation(s)
- Peter Muro
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Li Zhang
- Nanjing Lishui People’s Hospital, Zhongda Hospital, Southeast University, Nanjing, China
| | - Shuxuan Li
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zihan Zhao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Tao Jin
- Department of Gastrointestinal and Endoscopy, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Fei Mao
- Key Laboratory of Medical Science and Laboratory Medicine of Jiangsu Province, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhenwei Mao
- The Key Lab of Precision Diagnosis and Treatment in Hematologic Malignancies of Zhenjiang City, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
48
|
Nakamura A, Jo S, Nakamura S, Aparnathi MK, Boroojeni SF, Korshko M, Park YS, Gupta H, Vijayan S, Rockel JS, Kapoor M, Jurisica I, Kim TH, Haroon N. HIF-1α and MIF enhance neutrophil-driven type 3 immunity and chondrogenesis in a murine spondyloarthritis model. Cell Mol Immunol 2024; 21:770-786. [PMID: 38839914 PMCID: PMC11214626 DOI: 10.1038/s41423-024-01183-5] [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: 10/29/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
The hallmarks of spondyloarthritis (SpA) are type 3 immunity-driven inflammation and new bone formation (NBF). Macrophage migration inhibitory factor (MIF) was found to be a key driver of the pathogenesis of SpA by amplifying type 3 immunity, yet MIF-interacting molecules and networks remain elusive. Herein, we identified hypoxia-inducible factor-1 alpha (HIF1A) as an interacting partner molecule of MIF that drives SpA pathologies, including inflammation and NBF. HIF1A expression was increased in the joint tissues and synovial fluid of SpA patients and curdlan-injected SKG (curdlan-SKG) mice compared to the respective controls. Under hypoxic conditions in which HIF1A was stabilized, human and mouse neutrophils exhibited substantially increased expression of MIF and IL-23, an upstream type 3 immunity-related cytokine. Similar to MIF, systemic overexpression of IL-23 induced SpA pathology in SKG mice, while the injection of a HIF1A-selective inhibitor (PX-478) into curdlan-SKG mice prevented or attenuated SpA pathology, as indicated by a marked reduction in the expression of MIF and IL-23. Furthermore, genetic deletion of MIF or HIF1A inhibition with PX-478 in IL-23-overexpressing SKG mice did not induce evident arthritis or NBF, despite the presence of psoriasis-like dermatitis and blepharitis. We also found that MIF- and IL-23-expressing neutrophils infiltrated areas of the NBF in curdlan-SKG mice. These neutrophils potentially increased chondrogenesis and cell proliferation via the upregulation of STAT3 in periosteal cells and ligamental cells during endochondral ossification. Together, these results provide supporting evidence for an MIF/HIF1A regulatory network, and inhibition of HIF1A may be a novel therapeutic approach for SpA by suppressing type 3 immunity-mediated inflammation and NBF.
Collapse
Affiliation(s)
- Akihiro Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
- Department of Medicine, Division of Rheumatology, Queen's University, Kingston, ON, K7L, 2V6, Canada.
- Translational Institute of Medicine, School of Medicine, Queen's University, Kingston, ON, K7L 2V6, Canada.
- Division of Rheumatology, Kingston Health Science Centre, Kingston, ON, K7L 2V6, Canada.
| | - Sungsin Jo
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, 04763, Republic of Korea
- Department of Biology, College of Natural Sciences, Soonchunhyang University, Asan, 31538, Republic of Korea
| | - Sayaka Nakamura
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Mansi K Aparnathi
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Shaghayegh Foroozan Boroojeni
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mariia Korshko
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Ye-Soo Park
- Department of Orthopedic Surgery, Guri Hospital, Hanyang University College of Medicine, Guri, 11293, Republic of Korea
| | - Himanshi Gupta
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Sandra Vijayan
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Jason S Rockel
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
| | - Mohit Kapoor
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Department of Surgery and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5T 1P5, Canada
| | - Igor Jurisica
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada
- Departments of Medical Biophysics and Comp. Science and Faculty of Dentistry, University of Toronto, Toronto, ON, M5G 1L7, Canada
- Institute of Neuroimmunology, Slovak Academy of Sciences, 85410, Bratislava, Slovakia
| | - Tae-Hwan Kim
- Hanyang University Institute for Rheumatology Research (HYIRR), Seoul, 04763, Republic of Korea
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, 04763, Republic of Korea
| | - Nigil Haroon
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Krembil Research Institute, University Health Network, Toronto, ON, M5T 0S8, Canada.
- Institute of Medical Science, Temerty Faculty of Medicine of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
49
|
Esrefoglu M. Harnessing autophagy: A potential breakthrough in digestive disease treatment. World J Gastroenterol 2024; 30:3036-3043. [PMID: 38983959 PMCID: PMC11230060 DOI: 10.3748/wjg.v30.i24.3036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/30/2024] [Accepted: 06/04/2024] [Indexed: 06/25/2024] Open
Abstract
Autophagy, a conserved cellular degradation process, is crucial for various cellular processes such as immune responses, inflammation, metabolic and oxidative stress adaptation, cell proliferation, development, and tissue repair and remodeling. Dysregulation of autophagy is suspected in numerous diseases, including cancer, neurodegenerative diseases, digestive disorders, metabolic syndromes, and infectious and inflammatory diseases. If autophagy is disrupted, for example, this can have serious consequences and lead to chronic inflammation and tissue damage, as occurs in diseases such as Chron's disease and ulcerative colitis. On the other hand, the influence of autophagy on the development and progression of cancer is not clear. Autophagy can both suppress and promote the progression and metastasis of cancer at various stages. From inflammatory bowel diseases to gastrointestinal cancer, researchers are discovering the intricate role of autophagy in maintaining gut health and its potential as a therapeutic target. Researchers should carefully consider the nature and progression of diseases such as cancer when trying to determine whether inhibiting or stimulating autophagy is likely to be beneficial. Multidisciplinary approaches that combine cutting-edge research with clinical expertise are key to unlocking the full therapeutic potential of autophagy in digestive diseases.
Collapse
Affiliation(s)
- Mukaddes Esrefoglu
- Department of Histology and Embryology, Bezmialem Vakif University Medical Faculty, Istanbul 34093, Türkiye
| |
Collapse
|
50
|
Graça M, Nobre R, Sousa L, Ilic A. Distributed transformer for high order epistasis detection in large-scale datasets. Sci Rep 2024; 14:14579. [PMID: 38918413 PMCID: PMC11199512 DOI: 10.1038/s41598-024-65317-5] [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: 02/24/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
Understanding the genetic basis of complex diseases is one of the most important challenges in current precision medicine. To this end, Genome-Wide Association Studies aim to correlate Single Nucleotide Polymorphisms (SNPs) to the presence or absence of certain traits. However, these studies do not consider interactions between several SNPs, known as epistasis, which explain most genetic diseases. Analyzing SNP combinations to detect epistasis is a major computational task, due to the enormous search space. A possible solution is to employ deep learning strategies for genomic prediction, but the lack of explainability derived from the black-box nature of neural networks is a challenge yet to be addressed. Herein, a novel, flexible, portable, and scalable framework for network interpretation based on transformers is proposed to tackle any-order epistasis. The results on various epistasis scenarios show that the proposed framework outperforms state-of-the-art methods for explainability, while being scalable to large datasets and portable to various deep learning accelerators. The proposed framework is validated on three WTCCC datasets, identifying SNPs related to genes known in the literature that have direct relationships with the studied diseases.
Collapse
Affiliation(s)
- Miguel Graça
- INESC-ID, Instituto Superior Técnico, 1000-029, Lisbon, Portugal.
| | - Ricardo Nobre
- INESC-ID, Instituto Superior Técnico, 1000-029, Lisbon, Portugal
| | - Leonel Sousa
- INESC-ID, Instituto Superior Técnico, 1000-029, Lisbon, Portugal
| | - Aleksandar Ilic
- INESC-ID, Instituto Superior Técnico, 1000-029, Lisbon, Portugal
| |
Collapse
|