1
|
Jiang SM, Li XJ, Wang ZL, Chen ZW, Liu ZL, Li Q, Chen XL. Role of autophagy in rejection after solid organ transplantation: A systematic review of the literature. World J Transplant 2025; 15:103163. [DOI: 10.5500/wjt.v15.i3.103163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 04/18/2025] Open
Abstract
Organ transplantation has long been recognized as an effective treatment for end-stage organ failure, metabolic diseases, and malignant tumors. However, graft rejection caused by major histocompatibility complex mismatch remains a significant challenge. While modern immunosuppressants have made significant strides in reducing the incidence and risk of rejection, they have not been able to eliminate it completely. The intricate mechanisms underlying transplant rejection have been the subject of intense investigation by transplant immunologists. Among these factors, autophagy has emerged as a key player. Autophagy is an evolutionarily conserved mechanism in eukaryotic cells that mediates autophagocytosis and cellular protection. This process is regulated by autophagy-related genes and their encoded protein families, which maintain the material and energetic balance within cells. Additionally, autophagy has been reported to play crucial roles in the development, maturation, differentiation, and responses of immune cells. In the complex immune environment following transplantation, the role and mechanisms of autophagy are gradually being revealed. In this review, we aim to explore the current understanding of the role of autophagy in solid organ rejection after transplantation. Furthermore, we delve into the therapeutic advancements achieved by targeting autophagy involved in the rejection process.
Collapse
Affiliation(s)
- Shu-Min Jiang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Xue-Jiao Li
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, Guangdong Province, China
| | - Zi-Lin Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Zhi-Wei Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Zhi-Long Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Qiang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Xiao-Long Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
2
|
Jajodia A, Mishra A, Doni Jayavelu N, Lambert K, Moss N, Yang Z, Cerosaletti K, Buckner JH, Hawkins RD. Functional dissection of noncoding variants associated with rheumatoid arthritis. Ann Rheum Dis 2025:S0003-4967(25)00890-8. [PMID: 40318978 DOI: 10.1016/j.ard.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 05/07/2025]
Abstract
OBJECTIVES Noncoding variants are critical to our understanding of the genetic basis of diseases and disorders such as rheumatoid arthritis (RA). While genome-wide association studies have identified regions of the genome associated with disease, functional studies are still lagging that can identify potentially causative variants. METHODS In order to functionally fine-map RA-associated variants, we identified variants at enhancers marked in primary activated T helper cells and conducted massively parallel reporter assay in these cells. RESULTS We found that combinations of functional variant genotypes are often exclusive to patients with RA. We leveraged 3-dimensional genome architecture and expression quantitative trait loci data to identify target genes of enhancers exhibiting allelic differences in activity. We confirmed enhancer activity and target gene interactions by Clustered Regularly Interpaced Short Palindromic Repeats Cas9 (CRISPR-Cas9) deletion in primary T cells. CONCLUSIONS The identification of functional enhancer variants suggests possible causal variants, and their target genes reveal known and novel genes as likely drivers of RA, as well as a means for therapeutic intervention.
Collapse
Affiliation(s)
- Ajay Jajodia
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Arpit Mishra
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Naresh Doni Jayavelu
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Nicholas Moss
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Zongchen Yang
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Jane H Buckner
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - R David Hawkins
- Division of Medical Genetics, Department of Medicine, Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA; Benaroya Research Institute at Virginia Mason, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
3
|
Al Sayegh R, Wan J, Caër C, Azoulai M, Gasperment M, Baweja S, Chouillard MA, Kandiah J, Cadoux M, Mabire M, Pignolet C, Thibault-Sogorb T, Hammoutene A, Paradis V, Saveanu L, Nicolle R, Gilgenkrantz H, Weiss E, Lotersztajn S. Defective autophagy in CD4 T cells drives liver fibrosis via type 3 inflammation. Nat Commun 2025; 16:3860. [PMID: 40274816 PMCID: PMC12022296 DOI: 10.1038/s41467-025-59218-y] [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: 06/20/2024] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Conventional CD4 T cells represent a major source of inflammatory mediators that drive progression of chronic liver disease to fibrosis and to end-stage cirrhosis. Identification of T cell pathways that limits the inflammatory response could thus have therapeutic relevance. Here we show, using both human samples and mouse models, that autophagy is deficient in CD4 T cells from patients with advanced fibrosis, and that loss of autophagy following genomic deletion of ATG5 in T cells is associated with the emergence of pathogenic IL-17A + IFN-γ + Th17 T cells that drive liver fibrosis in mice. Mechanistically, liver CD4 T cells lacking autophagy display a Th17 glycolytic phenotype associated with enhanced type 3 cytokine (i.e., IL-17A and GM-CSF) release, shifting hepatic myofibroblasts, hepatocytes and macrophages toward a proinflammatory phenotype. We also show that autophagy can be rescued in CD4 T cells from patients with extensive liver fibrosis, leading to decreased frequency of pathogenic Th17 cells and reduced GM-CSF levels; in addition, limited fibrosis is observed in mice in which Rubicon, a negative regulator of autophagy, is deleted specifically in their T cells. Our findings thus implicate autophagy in CD4 T cells as a key therapeutic target to control inflammation-driven fibrosis during chronic liver injury.
Collapse
Affiliation(s)
- Rola Al Sayegh
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Jinghong Wan
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Charles Caër
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Margot Azoulai
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Maxime Gasperment
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Sukriti Baweja
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Institute of Liver Biliary Sciences, Delhi, India
| | - Marc-Anthony Chouillard
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Département d'Anesthésie et Réanimation, Hôpital Beaujon, Assistance Publique- Hôpitaux de Paris, Clichy, France
| | - Janany Kandiah
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Inovarion, Paris, France
| | - Mathilde Cadoux
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Morgane Mabire
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Camille Pignolet
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Tristan Thibault-Sogorb
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Département d'Anesthésie et Réanimation, Hôpital Beaujon, Assistance Publique- Hôpitaux de Paris, Clichy, France
| | - Adel Hammoutene
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Valérie Paradis
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, Clichy, France
| | - Loredana Saveanu
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Rémy Nicolle
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Hélène Gilgenkrantz
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
| | - Emmanuel Weiss
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France
- Département d'Anesthésie et Réanimation, Hôpital Beaujon, Assistance Publique- Hôpitaux de Paris, Clichy, France
| | - Sophie Lotersztajn
- Université Paris Cité, INSERM, U1149, Centre de Recherche sur l'Inflammation (CRI), Paris, France.
| |
Collapse
|
4
|
Yibcharoenporn C, Muanprasat C, Moonwiriyakit A, Satitsri S, Pathomthongtaweechai N. AMPK in Intestinal Health and Disease: A Multifaceted Therapeutic Target for Metabolic and Inflammatory Disorders. Drug Des Devel Ther 2025; 19:3029-3058. [PMID: 40291159 PMCID: PMC12024487 DOI: 10.2147/dddt.s507489] [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: 11/27/2024] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
The intestines play essential roles in nutrient absorption and immune function and help maintain a protective barrier. Disruptions to its function can result in various diseases, including metabolic disorders, inflammation, and cancer. As a key regulator of cellular energy levels, 5'-adenosine monophosphate-activated protein kinase (AMPK) is essential for intestinal health. Beyond its established metabolic role, emerging evidence suggests that AMPK exerts profound effects on intestinal cell physiology, influencing cell proliferation and differentiation, inflammation, autophagy, barrier integrity, and smooth muscle contractility. Here, we explore the structure and regulation of AMPK, as well as its diverse roles in intestinal diseases and potential as a therapeutic target. Our findings reveal that AMPK is a multifaceted regulator of intestinal health, modulating various cellular processes and intestinal diseases. It plays a dual role in cancer, acting as both a tumor suppressor and promoter, and it regulates inflammatory pathways, autophagy, tight junction formation, and smooth muscle contractility. Both natural and synthetic AMPK activators offer promise as therapeutic agents. This review of AMPK's mechanisms and activators offers valuable insights for developing novel therapies for intestinal disorders. Further research is needed to fully define AMPK's roles and therapeutic potential.
Collapse
Affiliation(s)
- Chamnan Yibcharoenporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Aekkacha Moonwiriyakit
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Saravut Satitsri
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| | - Nutthapoom Pathomthongtaweechai
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, 10540, Thailand
| |
Collapse
|
5
|
Norton EG, Chapman NM, Shi H, Meng X, Huang H, KC A, Rankin S, Saravia J, Yuan S, Hu H, Vogel P, Chi H. Vps34-orchestrated lipid signaling processes regulate the transitional heterogeneity and functional adaptation of effector regulatory T cells. PLoS Biol 2025; 23:e3003074. [PMID: 40215232 PMCID: PMC11990774 DOI: 10.1371/journal.pbio.3003074] [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: 12/02/2024] [Accepted: 02/19/2025] [Indexed: 04/14/2025] Open
Abstract
Regulatory T cell (Treg) heterogeneity exists in lymphoid and non-lymphoid tissues, but we have limited understanding of context-dependent functions and spatiotemporal regulators of heterogenous Treg states, especially during perinatal life when immune tolerance is established. Here, we revealed that the class III PI3K Vps34 orchestrates effector Treg (eTreg) transitional heterogeneity during perinatal life. We found that loss of Vps34 reduced terminal eTreg accumulation in lymphoid tissues, associated with decreased Treg generation in non-lymphoid tissues and development of an early-onset autoimmune-like disease. After perinatal life, Vps34-deficient eTreg accumulation was further impaired due to reduced cell survival, highlighting temporal regulation of eTreg heterogeneity and maintenance by Vps34. Accordingly, inhibition of Vps34 in mature Tregs disrupted immune homeostasis but boosted anti-tumor immunity. Mechanistically, multiomics profiling approaches uncovered that Vps34-orchestrated transcriptional and epigenetic remodeling promotes terminal eTreg programming. Further, via genetic deletion of the Vps34-interacting proteins Atg14 or Uvrag in Tregs, we established that Atg14 but not Uvrag was required for the overall survival, but not terminal differentiation, of eTregs, suggesting that autophagy but not endocytosis partly contributed to Vps34-dependent effects. Accordingly, mice with Treg-specific loss of Atg14, but not Uvrag, had moderately disrupted immune homeostasis and reduced tumor growth, with Vps34- or Atg14-dependent gene signatures also being elevated in intratumoral Tregs from human cancer patients. Collectively, our study reveals distinct Vps34-orchestrated signaling events that regulate eTreg heterogeneity and functional adaptation and the pathophysiological consequences on autoimmunity versus anti-tumor immunity.
Collapse
Affiliation(s)
- Erienne G. Norton
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Nicole M. Chapman
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Xiaoxi Meng
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongling Huang
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Anil KC
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sherri Rankin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Jordy Saravia
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Sujing Yuan
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Haoran Hu
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Peter Vogel
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| | - Hongbo Chi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
| |
Collapse
|
6
|
Chen Y, Chen X, Lin S, Huang S, Li L, Hong M, Li J, Ma L, Ma J. Effects of psychological stress on inflammatory bowel disease via affecting the microbiota-gut-brain axis. Chin Med J (Engl) 2025; 138:664-677. [PMID: 39965932 PMCID: PMC11925421 DOI: 10.1097/cm9.0000000000003389] [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: 08/25/2024] [Indexed: 02/20/2025] Open
Abstract
ABSTRACT Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory condition with chronic and relapsing manifestations and is characterized by a disturbance in the interplay between the intestinal microbiota, the gut, and the brain. The microbiota-gut-brain axis involves interactions among the nervous system, the neuroendocrine system, the gut microbiota, and the host immune system. Increasing published data indicate that psychological stress exacerbates the severity of IBD due to its negative effects on the microbiota-gut-brain axis, including alterations in the stress response of the hypothalamic-pituitary-adrenal (HPA) axis, the balance between the sympathetic nervous system and vagus nerves, the homeostasis of the intestinal flora and metabolites, and normal intestinal immunity and permeability. Although the current evidence is insufficient, psychotropic agents, psychotherapies, and interventions targeting the microbiota-gut-brain axis show the potential to improve symptoms and quality of life in IBD patients. Therefore, further studies that translate recent findings into therapeutic approaches that improve both physical and psychological well-being are needed.
Collapse
Affiliation(s)
- Yuhan Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xiaofen Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Suqin Lin
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shengjun Huang
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lijuan Li
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mingzhi Hong
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianzhou Li
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People’s Hospital of Xining, Xining, Qinghai 810003, China
| | - Lili Ma
- Department of Gastroenterology and Hepatology, Qinghai Provincial People’s Hospital, Xining, Qinghai 810007, China
| | - Juan Ma
- Department of Gastroenterology and Hepatology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People’s Hospital of Xining, Xining, Qinghai 810003, China
| |
Collapse
|
7
|
Yang S, Fan L, Yin L, Zhao Y, Li W, Zhao R, Jia X, Dong F, Zheng Z, Zhao D, Wang J. Ginseng exosomes modulate M1/M2 polarisation by activating autophagy and target IKK/IкB/NF-кB to alleviate inflammatory bowel disease. J Nanobiotechnology 2025; 23:198. [PMID: 40065319 PMCID: PMC11895377 DOI: 10.1186/s12951-025-03292-3] [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: 11/28/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Exosomes are involved in intercellular communication and regulation of the inflammatory microenvironment. In a previous study, we demonstrated that fresh ginseng exosomes (GEs) alleviated inflammatory bowel disease. However, the precise mechanism by which GEs activate the immune system and subsequently inhibit the formation of intestinal inflammatory microenvironment remains unknown. METHODS Herein, we investigated the effects of GEs on autophagy, macrophage polarisation, intestinal inflammation, and the epithelial barrier by means of transcriptome sequencing, network pharmacology, transmission electron microscopy, immunoblotting, flow cytometry and small molecule inhibitors. RESULTS GEs significantly activated autophagy and M2-like macrophage polarisation, which could be blocked by the autophagy inhibitor 3-methyladenine. In the co-culture system of macrophages and intestinal epithelial cells, macrophages treated with GEs secreted more interleukin-10 (IL-10) and significantly reduced Nitric oxide (NO) levels in intestinal epithelial cells in vitro. Furthermore, GEs acted directly on intestinal epithelial cells through the IKK/IкB/NF-кB signalling pathway to reduce inflammation and restore the intestinal barrier. Orally administered GEs could restore disrupted colonic barriers, alleviate inflammatory bowel responses, and regulate the polarisation of intestinal macrophages in vivo. CONCLUSION In summary, GEs may be a potential treatment for inflammatory bowel disease, and targeting autophagy and macrophage polarisation may help alleviate intestinal inflammation.
Collapse
Affiliation(s)
- Song Yang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Liangliang Fan
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Lijia Yin
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Yueming Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Wenjing Li
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Ronghua Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Xuxia Jia
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Fusong Dong
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Ze Zheng
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Daqing Zhao
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China
| | - Jiawen Wang
- Changchun University of Chinese Medicine, 1035 Boshuo Road, Changchun, 130117, Jilin Province, China.
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 17176, Stockholm, Sweden.
| |
Collapse
|
8
|
Lu Y, Man XY. Diversity and function of regulatory T cells in health and autoimmune diseases. J Autoimmun 2025; 151:103357. [PMID: 39805189 DOI: 10.1016/j.jaut.2025.103357] [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: 10/07/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/16/2025]
Abstract
Regulatory T cell (Treg) play a pivotal role in immune regulation and maintaining host immune homeostasis. Treg heterogeneity, characterized by diverse gene expression profiles and functional states, is complex in both health and disease. Research reveals that Tregs are not a uniform population but exhibit diversity based on their origin, location, and functional status. This heterogeneity is crucial for understanding Treg roles in various pathological conditions. Dysfunctional Tregs are closely linked to the pathogenesis of autoimmune diseases, although the precise mechanisms remain unclear. The phenotypic and functional heterogeneity of Tregs is particularly significant in diseases such as systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, type 1 diabetes, psoriasis and autoimmune liver diseases. This review explores Treg origins, classifications, and heterogeneity in these conditions, aiming to provide new perspectives and strategies for diagnosis and treatment. Understanding Treg heterogeneity and plasticity promises to reveal novel therapeutic targets and advance precision immunotherapy development.
Collapse
Affiliation(s)
- Yi Lu
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
9
|
Sharma R, Wu K, Han K, Russo AC, Dagur PK, Combs CA, Yao X, Levine SJ, Sack MN. BLOC1S1 Control of Vacuolar Organelle Fidelity Modulates Murine T H2 Cell Immunity and Allergy Susceptibility. Allergy 2024. [PMID: 39737471 DOI: 10.1111/all.16461] [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: 08/06/2024] [Revised: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/01/2025]
Abstract
BACKGROUND The levels of biogenesis of lysosome organelles complex 1 subunit 1 (BLOC1S1) control mitochondrial and endolysosome organelle homeostasis and function. Reduced fidelity of these vacuolar organelles is increasingly being recognized as important in instigating cell-autonomous immune cell activation. We reasoned that exploring the role of BLOC1S1 in CD4+ T cells may further advance our understanding of regulatory events linked to mitochondrial and/or endolysosomal function in adaptive immunity. METHODS CD4+ T cells were analyzed from control and CD4+ T-cell-specific BLOC1S1 knockout mice. Polarization profiles were assayed using biochemical and molecular signatures, and signaling pathways were disrupted pharmacologically or via siRNA. Mouse models of airway and skin inflammation were generated by Ovalbumin and MC903 exposure, respectively. RESULTS TH2 regulator GATA3 and phosphorylated STAT6 were preferentially induced in BLOC1S1-depleted primary CD4+ T (TKO) cells. The levels of IL-4, IL-5, and IL-13 were markedly induced in the absence of BLOC1S1. At the organelle level, mitochondrial DNA leakage evoked cGAS-STING and NF-κB pathway activation with subsequent TH2 polarization. The induction of autophagy with rapamycin reduced cytosolic mtDNA and reversed these TH2 signatures. Furthermore, genetic knockdown of STING and NF-κB inhibition ameliorated this immune regulatory cascade in TKO cells. Finally, at a functional level, TKO mice displayed an increased susceptibility to allergic conditions, including dermatitis and allergic asthma. CONCLUSIONS BLOC1S1 depletion in mouse CD4+ T cells mediated disruption of mitochondrial integrity to initiate a predominant TH2-responsive phenotype via STING-NF-κB-driven signaling of the canonical TH2 regulatory program.
Collapse
Affiliation(s)
- Rahul Sharma
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kaiyuan Wu
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| | - Kim Han
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | - Anna Chiara Russo
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
| | | | | | - Xianglan Yao
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Stewart J Levine
- Critical Care Medicine and Pulmonary Branch, Bethesda, Maryland, USA
| | - Michael N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, NIH, Bethesda, Maryland, USA
- Cardiovascular Branch, NHLBI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
10
|
Migliore L, Cianfanelli V, Zevolini F, Gesualdo M, Marzuoli L, Patrussi L, Ulivieri C, Marotta G, Cecconi F, Finetti F, Baldari CT. An AMBRA1, ULK1 and PP2A regulatory network regulates cytotoxic T cell differentiation via TFEB activation. Sci Rep 2024; 14:31838. [PMID: 39738384 PMCID: PMC11685475 DOI: 10.1038/s41598-024-82957-9] [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: 07/18/2024] [Accepted: 12/09/2024] [Indexed: 01/02/2025] Open
Abstract
The scaffold protein AMBRA1, which participates in the autophagy pathway, also promotes CD4+ T cell differentiation to Tregs independent of autophagy through its interactor PP2A. Here we have investigated the role of AMBRA1 in CD8+ T cell differentiation to cytotoxic T cells (CTL). AMBRA1 depletion in CD8+ T cells was associated with impaired expression of the transcription factors RUNX3 and T-BET that drive CTL differentiation and resulted in impaired acquisition of cytotoxic potential. These effects were recapitulated by pharmacological inhibition of the AMBRA1 activator ULK1 or its interactor PP2A. Based on the ability of PP2A to activate TFEB, we hypothesized a role for TFEB in the CTL differentiation program regulated by AMBRA1. We show that TFEB modulates RUNX3 and T-BET expression and the generation of killing-competent CTLs, and that AMBRA1 depletion, or ULK1 or PP2A inhibition, suppresses TFEB activity. These data highlight a role for AMBRA1, ULK1 and PP2A in CTL generation, mediated by TFEB, which we identify as a new pioneering transcription factor in the CTL differentiation program.
Collapse
Affiliation(s)
- Loredana Migliore
- Department of Life Sciences, University of Siena, Siena, Italy
- Department of Science, University "ROMA TRE", Rome, Italy
| | - Valentina Cianfanelli
- Department of Woman and Child Health and Public Health, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Science, University "ROMA TRE", Rome, Italy
| | | | - Monica Gesualdo
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | - Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy
| | | | | | - Francesco Cecconi
- Università Cattolica del Sacro Cuore and Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Cell Stress and Survival Group, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark
| | | | | |
Collapse
|
11
|
Tibbitt CA, Coquet JM. Metabolic requirements of type 2 lymphocytes in allergic disease. Curr Opin Immunol 2024; 91:102500. [PMID: 39471654 DOI: 10.1016/j.coi.2024.102500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/20/2024] [Accepted: 10/05/2024] [Indexed: 11/01/2024]
Abstract
Allergic diseases continue to increase in prevalence across the globe. Decades of research has uncovered the cytokines and transcription factors that are central to the allergic immune response, but only in the last few years have we begun to understand the metabolic requirements of allergic immunity. Here, we discuss the metabolic features of so-called 'type 2' lymphocytes, which are heavily implicated in allergy. We highlight the central role that nuclear receptors, such as peroxisome proliferator-activated receptor gamma, play in type 2 lymphocyte biology and explore the influence of dietary and microbial factors in allergic inflammation. In the future, targeting metabolic checkpoints may offer a meaningful way of treating patients with allergic disorders.
Collapse
Affiliation(s)
- Christopher A Tibbitt
- Department of Medicine Huddinge, Centre for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden; Clinical Lung and Allergy Research Medical Unit for Lung and Allergy Diseases, Karolinska University Hospital, Stockholm, Sweden.
| | - Jonathan M Coquet
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Leo Foundation Skin Immunology Research Centre, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Courreges CJF, Davenport ECM, Bilanges B, Rebollo-Gomez E, Hukelmann J, Schoenfelder P, Edgar JR, Sansom D, Scudamore CL, Roychoudhuri R, Garden OA, Vanhaesebroeck B, Okkenhaug K. Lack of phosphatidylinositol 3-kinase VPS34 in regulatory T cells leads to a fatal lymphoproliferative disorder without affecting their development. Front Immunol 2024; 15:1374621. [PMID: 39664390 PMCID: PMC11631860 DOI: 10.3389/fimmu.2024.1374621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/31/2024] [Indexed: 12/13/2024] Open
Abstract
Regulatory T (Treg) cells are essential for the maintenance of immunological tolerance, yet the molecular components required for their maintenance and effector functions remain incompletely defined. Inactivation of VPS34 in Treg cells led to an early, lethal phenotype, with massive effector T cell activation and inflammation, like mice lacking Treg cells completely. However, VPS34-deficient Treg cells developed normally, populated the peripheral lymphoid organs and effectively supressed conventional T cells in vitro. Our data suggest that VPS34 is required for the maintaining normal numbers of mature Treg. Functionally, we observed that lack of VPS34 activity impairs cargo processing upon transendocytosis, that defective autophagy may contribute to, but is not sufficient to explain this lethal phenotype, and that loss of VPS34 activity induces a state of heightened metabolic activity that may interfere with metabolic networks required for maintenance or suppressive functions of Treg cells.
Collapse
Affiliation(s)
- Christina J. F. Courreges
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
- Department of Pathology, The University of Cambridge, Cambridge, United Kingdom
| | - Elizabeth C. M. Davenport
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
- Royal Veterinary College, London, United Kingdom
| | - Benoit Bilanges
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Jens Hukelmann
- The School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Priya Schoenfelder
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
| | - James R. Edgar
- Department of Pathology, The University of Cambridge, Cambridge, United Kingdom
| | - David Sansom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | | | - Rahul Roychoudhuri
- Department of Pathology, The University of Cambridge, Cambridge, United Kingdom
| | | | | | - Klaus Okkenhaug
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, United Kingdom
- Department of Pathology, The University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
13
|
Feng P, Yang Q, Luo L, Guan Z, Fu J, Zhao M, Meng W, Wan S, He J, Li Z, Wang G, Sun G, Dong Z, Yang M. Vps34 sustains Treg cell survival and function via regulating intracellular redox homeostasis. Cell Death Differ 2024; 31:1519-1533. [PMID: 39117783 PMCID: PMC11519664 DOI: 10.1038/s41418-024-01353-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 07/28/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024] Open
Abstract
The survival and suppressive function of regulatory T (Treg) cells rely on various intracellular metabolic and physiological processes. Our study demonstrates that Vps34 plays a critical role in maintaining Treg cell homeostasis and function by regulating cellular metabolic activities. Disruption of Vps34 in Treg cells leads to spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number of Treg cells, particularly eTreg cells with highly immunosuppressive activity. Mechanistically, the poor survival of Vps34-deficient Treg cells is attributed to impaired endocytosis, intracellular vesicular trafficking and autophagosome formation, which further results in enhanced mitochondrial respiration and excessive ROS production. Removal of excessive ROS can effectively rescue the death of Vps34-deficient Treg cells. Functionally, acute deletion of Vps34 within established Treg cells enhances anti-tumor immunity in a malignant melanoma model by boosting T-cell-mediated anti-tumor activity. Overall, our results underscore the pivotal role played by Vps34 in orchestrating Treg cell homeostasis and function towards establishing immune homeostasis and tolerance.
Collapse
Affiliation(s)
- Peiran Feng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
| | - Liang Luo
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Zerong Guan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Jiamin Fu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Mingyue Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Wanqing Meng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China
| | - Shuo Wan
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Junming He
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China
| | - Zhizhong Li
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China
| | - Zhongjun Dong
- The First Affiliated Hospital of Anhui Medical University and Institute for Clinical Immunology, Anhui Medical University, Hefei, Anhui, China.
| | - Meixiang Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, China.
- Key Laboratory of Ministry of Education for Viral Pathogenesis & Infection Prevention and Control (Jinan University). Guangzhou Key Laboratory for Germ-Free Animals and Microbiota Application. Institute of Laboratory Animal Science, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
14
|
Camino-Mera A, Pardo-Seco J, Bello X, Argiz L, Boyle RJ, Custovic A, Herberg J, Kaforou M, Arasi S, Fiocchi A, Pecora V, Barni S, Mori F, Bracamonte T, Echeverria L, O'Valle-Aísa V, Hernández-Martínez NL, Carballeira I, García E, Garcia-Magan C, Moure-González JD, Gonzalez-Delgado P, Garriga-Baraut T, Infante S, Zambrano-Ibarra G, Tomás-Pérez M, Machinena A, Pascal M, Prieto A, Vázquez-Cortes S, Fernández-Rivas M, Vila L, Alsina L, Torres MJ, Mangone G, Quirce S, Martinón-Torres F, Vázquez-Ortiz M, Gómez-Carballa A, Salas A. Whole Exome Sequencing Identifies Epithelial and Immune Dysfunction-Related Biomarkers in Food Protein-Induced Enterocolitis Syndrome. Clin Exp Allergy 2024; 54:919-929. [PMID: 39348862 DOI: 10.1111/cea.14564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/01/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Food protein-induced enterocolitis syndrome (FPIES) is a food allergy primarily affecting infants, often leading to vomiting and shock. Due to its poorly understood pathophysiology and lack of specific biomarkers, diagnosis is frequently delayed. Understanding FPIES genetics can shed light on disease susceptibility and pathophysiology-key to developing diagnostic, prognostic, preventive and therapeutic strategies. Using a well-characterised cohort of patients we explored the potential genome-wide susceptibility factors underlying FPIES. METHODS Blood samples from 41 patients with oral food challenge-proven FPIES were collected for a comprehensive whole exome sequencing association study. RESULTS Notable genetic variants, including rs872786 (RBM8A), rs2241880 (ATG16L1) and rs2289477 (ATG16L1), were identified as significant findings in FPIES. A weighted SKAT model identified six other associated genes including DGKZ and SIRPA. DGKZ induces TGF-β signalling, crucial for epithelial barrier integrity and IgA production; RBM8A is associated with thrombocytopenia absent radius syndrome, frequently associated with cow's milk allergy; SIRPA is associated with increased neutrophils/monocytes in inflamed tissues as often observed in FPIES; ATG16L1 is associated with inflammatory bowel disease. Coexpression correlation analysis revealed a functional correlation between RBM8A and filaggrin gene (FLG) in stomach and intestine tissue, with filaggrin being a known key pathogenic and risk factor for IgE-mediated food allergy. A transcriptome-wide association study suggested genetic variability in patients impacted gene expression of RBM8A (stomach and pancreas) and ATG16L1 (transverse colon). CONCLUSIONS This study represents the first case-control exome association study of FPIES patients and marks a crucial step towards unravelling genetic susceptibility factors underpinning the syndrome. Our findings highlight potential factors and pathways contributing to FPIES, including epithelial barrier dysfunction and immune dysregulation. While these results are novel, they are preliminary and need further validation in a second cohort of patients.
Collapse
Affiliation(s)
- Alba Camino-Mera
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Laura Argiz
- Allergy Section, Clinica Universidad de Navarra, Madrid, Spain
| | - Robert J Boyle
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Adnan Custovic
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jethro Herberg
- Department of Infectious Disease, Imperial College London, London, UK
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London, UK
| | - Stefania Arasi
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alessandro Fiocchi
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Valentina Pecora
- Allergy Diseases Research Area, Pediatric Allergology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Simona Barni
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Francesca Mori
- Allergy Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Teresa Bracamonte
- Paediatric Allergy Section, Severo Ochoa University Hospital, Madrid, Spain
| | - Luis Echeverria
- Paediatric Allergy Section, Severo Ochoa University Hospital, Madrid, Spain
| | - Virginia O'Valle-Aísa
- Clinical Analysis and Clinical Biochemistry Service, Severo Ochoa University Hospital, Madrid, Spain
| | | | - Iria Carballeira
- Paediatric Allergy Section, Arquitecto Marcide Hospital, Ferrol, A Coruña in Galicia, Spain
| | - Emilio García
- Paediatric Allergy Section, Arquitecto Marcide Hospital, Ferrol, A Coruña in Galicia, Spain
| | - Carlos Garcia-Magan
- Paediatrics Department, Hospital Clínico Universitario de Santiago de Compostela, Coruña, Galicia, Spain
| | | | | | - Teresa Garriga-Baraut
- Paediatric Allergy Section, Vall D'Hebron University Hospital, Growth and Development Research Group, Vall d'Hebron Research Institute (VHIR), Barcelona, Spain
| | - Sonsoles Infante
- Pediatric Allergy Unit, Hospital General Universitario Gregorio Marañón, Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Gabriela Zambrano-Ibarra
- Pediatric Allergy Unit, Hospital General Universitario Gregorio Marañón, Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Margarita Tomás-Pérez
- Pediatric Allergy Unit, Hospital General Universitario Gregorio Marañón, Gregorio Marañón Health Research Institute (IiSGM), Madrid, Spain
| | - Adrianna Machinena
- Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mariona Pascal
- Immunology Department, CDB, Hospital Clínic de Barcelona, Barcelona, Spain
- IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - Ana Prieto
- Paediatric Allergy Section, General University Hospital, Malaga, Spain
| | - Sonia Vázquez-Cortes
- Allergy Department, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Montserrat Fernández-Rivas
- Allergy Department, Hospital Clinico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Universidad Complutense, Madrid, Spain
| | - Leticia Vila
- Paediatric Allergy Section, Teresa Herrera Hospital, Coruna, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Institut de Recerca Sant Joan de Déu and Universitat de Barcelona, Barcelona, Spain
| | - María José Torres
- Allergy Department, General University Hospital, Málaga, Spain
- Allergy Research Group, Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina-IBIMA Plataforma Bionand, Málaga, Spain
- Universidad de Málaga (UMA), Málaga, Spain
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Giusi Mangone
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Santiago Quirce
- Department of Allergy, La Paz University Hospital, IdiPAZ, Madrid, Spain
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Marta Vázquez-Ortiz
- Section of Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, London, UK
| | - Alberto Gómez-Carballa
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Antonio Salas
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| |
Collapse
|
15
|
Muñiz Pedrogo DA, Sears CL, Melia JMP. Colorectal Cancer in Inflammatory Bowel Disease: A Review of the Role of Gut Microbiota and Bacterial Biofilms in Disease Pathogenesis. J Crohns Colitis 2024; 18:1713-1725. [PMID: 38703073 DOI: 10.1093/ecco-jcc/jjae061] [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: 11/17/2023] [Revised: 01/31/2024] [Accepted: 05/03/2024] [Indexed: 05/06/2024]
Abstract
The risk of colorectal cancer [CRC] is increased in patients with inflammatory bowel disease [IBD], particularly in extensive ulcerative colitis [UC] and Crohn's colitis. Gut microbiota have been implicated in the pathogenesis of CRC via multiple mechanisms, including the release of reactive oxygen species and genotoxins, and induction of inflammation, as well as activation of the immune response. Gut microbiota can enhance their carcinogenic and proinflammatory properties by organising into biofilms, potentially making them more resistant to the host's immune system and to antibiotics. Colonic biofilms have the capacity to invade colonic tissue and accelerate tumorigenesis in tumour-prone models of mice. In the context of IBD, the prevalence of biofilms has been estimated to be up to 95%. Although the relationship between chronic inflammation and molecular mediators that contribute to IBD-associated CRC is well established, the role of gut microbiota and biofilms in this sequence is not fully understood. Because CRC can still arise in the absence of histological inflammation, there is a growing interest in identifying chemopreventive agents against IBD-associated CRC. Commonly used in the treatment of UC, 5-aminosalicylates have antimicrobial and anticarcinogenic properties that might have a role in the chemoprevention of CRC via the inhibition or modulation of carcinogenic gut microbiota and potentially of biofilm formation. Whether biologics and other IBD-targeted therapies can decrease the progression towards dysplasia and CRC, via mechanisms independent of inflammation, is still unknown. Further research is warranted to identify potential new microbial targets in therapy for chemoprevention of dysplasia and CRC in IBD.
Collapse
Affiliation(s)
- David A Muñiz Pedrogo
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Joanna M P Melia
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Jauregui W, Abarca YA, Ahmadi Y, Menon VB, Zumárraga DA, Rojas Gomez MC, Basri A, Madala RS, Girgis P, Nazir Z. Shared Pathophysiology of Inflammatory Bowel Disease and Psoriasis: Unraveling the Connection. Cureus 2024; 16:e68569. [PMID: 39364475 PMCID: PMC11449469 DOI: 10.7759/cureus.68569] [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] [Accepted: 09/02/2024] [Indexed: 10/05/2024] Open
Abstract
Psoriasis (PS) and inflammatory bowel disease (IBD) are immune-mediated chronic conditions that share pathophysiological processes, including immune system dysfunction, microbiome dysbiosis, and inflammatory pathways. These pathways result in increased turnover of epithelial cells and compromised barrier function. The assessment of the literature suggests that immunopathogenic mechanisms, such as tumor necrosis factor (TNF)-α signaling and IL-23/IL-17 axis dysregulation, are shared by PS and IBD. Clinical characteristics and diagnostic approaches overlap significantly, and advances in biomarker identification benefit both conditions. Current treatments, namely biologics that target TNF-α, IL-17, and IL-23, show promising results in decreasing inflammation and controlling symptoms. Precision medicine approaches are prioritized in prospective therapeutic procedures to tailor pharmaceuticals based on specific biomarkers, perhaps improving outcomes and minimizing side effects. This study thoroughly examines and evaluates the body of research on PS and IBD. Several papers were examined to compile data on clinical features, diagnosis, therapies, pathophysiology, epidemiology, and potential future therapeutic developments. The selection of articles was based on three methodological qualities: relevance and addition to the knowledge of IBD and PS. The retrieved data were combined to provide a coherent summary of the state of the knowledge and to spot new trends. The overview of the latest studies demonstrates that both PS and IBD share pathophysiological foundations and therapeutic approaches. With a spotlight on particular biomarkers, advances in precision medicine provide a promising path toward enhancing therapeutic effectiveness and minimizing side effects.
Collapse
Affiliation(s)
- Walter Jauregui
- General Medicine, Universidad Nacional Autónoma de Honduras, Tegucigalpa, HND
| | - Yozahandy A Abarca
- Internal Medicine, Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Mexico City, MEX
| | - Yasmin Ahmadi
- School of Medicine, Royal College of Surgeons in Ireland - Medical University of Bahrain, Muharraq, BHR
| | - Vaishnavi B Menon
- Internal Medicine, Sri Ramachandra Institute of Higher Education and Research, Chennai, IND
| | | | | | - Aleeza Basri
- Internal Medicine, Liaquat University of Medical and Health Sciences, Hyderabad, PAK
| | | | - Peter Girgis
- Internal Medicine, Ross University School of Medicine, Bridgetown, BRB
| | - Zahra Nazir
- Internal Medicine, Combined Military Hospital, Quetta, PAK
| |
Collapse
|
17
|
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
|
18
|
Zhao X, Ma D, Yang B, Wang Y, Zhang L. Research progress of T cell autophagy in autoimmune diseases. Front Immunol 2024; 15:1425443. [PMID: 39104538 PMCID: PMC11298352 DOI: 10.3389/fimmu.2024.1425443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 08/07/2024] Open
Abstract
T cells, as a major lymphocyte population involved in the adaptive immune response, play an important immunomodulatory role in the early stages of autoimmune diseases. Autophagy is a cellular catabolism mediated by lysosomes. Autophagy maintains cell homeostasis by recycling degraded cytoplasmic components and damaged organelles. Autophagy has a protective effect on cells and plays an important role in regulating T cell development, activation, proliferation and differentiation. Autophagy mediates the participation of T cells in the acquired immune response and plays a key role in antigen processing as well as in the maintenance of T cell homeostasis. In autoimmune diseases, dysregulated autophagy of T cells largely influences the pathological changes. Therefore, it is of great significance to study how T cells play a role in the immune mechanism of autoimmune diseases through autophagy pathway to guide the clinical treatment of diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
19
|
Al-Kuraishy HM, Sulaiman GM, Jabir MS, Mohammed HA, Al-Gareeb AI, Albukhaty S, Klionsky DJ, Abomughaid MM. Defective autophagy and autophagy activators in myasthenia gravis: a rare entity and unusual scenario. Autophagy 2024; 20:1473-1482. [PMID: 38346408 PMCID: PMC11210922 DOI: 10.1080/15548627.2024.2315893] [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: 11/30/2023] [Revised: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 03/07/2024] Open
Abstract
Myasthenia gravis (MG) is an autoimmune disease of the neuromuscular junction (NMJ) that results from autoantibodies against nicotinic acetylcholine receptors (nAchRs) at NMJs. These autoantibodies are mainly originated from autoreactive B cells that bind and destroy nAchRs at NMJs preventing nerve impulses from activating the end-plates of skeletal muscle. Indeed, immune dysregulation plays a crucial role in the pathogenesis of MG. Autoreactive B cells are increased in MG due to the defect in the central and peripheral tolerance mechanisms. As well, autoreactive T cells are augmented in MG due to the diversion of regulatory T (Treg) cells or a defect in thymic anergy leading to T cell-mediated autoimmunity. Furthermore, macroautophagy/autophagy, which is a conserved cellular catabolic process, plays a critical role in autoimmune diseases by regulating antigen presentation, survival of immune cells and cytokine-mediated inflammation. Abnormal autophagic flux is associated with different autoimmune disorders. Autophagy regulates the connection between innate and adaptive immune responses by controlling the production of cytokines and survival of Tregs. As autophagy is involved in autoimmune disorders, it may play a major role in the pathogenesis of MG. Therefore, this mini-review demonstrates the potential role of autophagy and autophagy activators in MG.Abbreviations: Ach, acetylcholine; Breg, regulatory B; IgG, immunoglobulin G; MG, myasthenia gravis; NMJ, neuromuscular junction; ROS, reactive oxygen species; Treg, regulatory T; Ubl, ubiquitin-like.
Collapse
Affiliation(s)
- Hayder M. Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | | | - Majid S. Jabir
- Department of Applied Sciences, University of Technology, Baghdad, Iraq
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
- Department of Pharmacognosy and Medicinal Plants, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| | | | - Salim Albukhaty
- Department of Chemistry, College of Science, University of Misan, Maysan, Iraq
| | | | - Mosleh M. Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
20
|
Zhou L, Zhang L, Lv Y, Qian J, Huang L, Qu C. YTHDC1 inhibits autophagy-dependent NF-κB signaling by stabilizing Beclin1 mRNA in macrophages. J Inflamm (Lond) 2024; 21:22. [PMID: 38877444 PMCID: PMC11179287 DOI: 10.1186/s12950-024-00393-y] [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: 10/31/2023] [Accepted: 05/15/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND YTHDC1, a key m(6)A nuclear reader, plays a crucial role in regulating mRNA splicing, export, and stability. However, the functional significance and regulatory mechanisms of YTHDC1 in inflammatory bowel disease (IBD) remain to be explored. METHODS We established a dextran sulfate sodium (DSS)-induced murine colitis model in vivo and LPS/IFN-γ-stimulated macrophage inflammation in vitro. The expression of YTHDC1 was determined. Colocalization of YTHDC1 and macrophages was assayed by immunofluorescence staining. LV-YTHDC1 or shYTHDC1 lentiviruses were applied for YTHDC1 overexpression or inhibition. For NF-κB inhibition, JSH-23 was utilized. The interaction of YTHDC1 and Beclin1 mRNA was determined by RIP, and the m6A modification of Beclin1 was confirmed by MeRIP. RESULTS In DSS-induced colitis and LPS/IFN-γ-treated RAW264.7 macrophages, we observed a significant downregulation of YTHDC1. Overexpression of YTHDC1 resulted in decreased levels of iNOS, CD86, and IL-6 mRNA, along with inhibited NF-κB activation in LPS/IFN-γ-treated RAW264.7 cells. Conversely, downregulation of YTHDC1 promoted iNOS expression and inhibited autophagy. Additionally, the effect of YTHDC1 knockdown on CD86 and IL-6 mRNA induced by LPS/IFN-γ was abolished by the NF-κB inhibitor JSH-23. Mechanistically, YTHDC1 interacted with Beclin1 mRNA, thereby stabilizing Beclin1 mRNA and enhancing Beclin1 expression and autophagy. These effects ultimately led to the inhibition of NF-κB signaling in LPS/IFN-γ-challenged macrophages. CONCLUSIONS YTHDC1 inhibited the macrophage-mediated inflammatory response by stabilizing Beclin1 mRNA, which may be a potential therapeutic target for the treatment of IBD.
Collapse
Affiliation(s)
- Li Zhou
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Ling Zhang
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Yan Lv
- Center for Translational Medicine, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Jiasheng Qian
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China
| | - Long Huang
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China.
| | - Chenjiang Qu
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, No. 68 West Jiyang Road, Suzhou, 215600, China.
| |
Collapse
|
21
|
Schreurs RRCE, Koulis A, Booiman T, Boeser-Nunnink B, Cloherty APM, Rader AG, Patel KS, Kootstra NA, Ribeiro CMS. Autophagy-enhancing ATG16L1 polymorphism is associated with improved clinical outcome and T-cell immunity in chronic HIV-1 infection. Nat Commun 2024; 15:2465. [PMID: 38548722 PMCID: PMC10979031 DOI: 10.1038/s41467-024-46606-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 03/04/2024] [Indexed: 04/01/2024] Open
Abstract
Chronic HIV-1 infection is characterized by T-cell dysregulation that is partly restored by antiretroviral therapy. Autophagy is a critical regulator of T-cell function. Here, we demonstrate a protective role for autophagy in HIV-1 disease pathogenesis. Targeted analysis of genetic variation in core autophagy gene ATG16L1 reveals the previously unidentified rs6861 polymorphism, which correlates functionally with enhanced autophagy and clinically with improved survival of untreated HIV-1-infected individuals. T-cells carrying ATG16L1 rs6861(TT) genotype display improved antiviral immunity, evidenced by increased proliferation, revamped immune responsiveness, and suppressed exhaustion/immunosenescence features. In-depth flow-cytometric and transcriptional profiling reveal T-helper-cell-signatures unique to rs6861(TT) individuals with enriched regulation of pro-inflammatory networks and skewing towards immunoregulatory phenotype. Therapeutic enhancement of autophagy recapitulates the rs6861(TT)-associated T-cell traits in non-carriers. These data underscore the in vivo relevance of autophagy for longer-lasting T-cell-mediated HIV-1 control, with implications towards development of host-directed antivirals targeting autophagy to restore immune function in chronic HIV-1 infection.
Collapse
Affiliation(s)
- Renée R C E Schreurs
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Athanasios Koulis
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Thijs Booiman
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Brigitte Boeser-Nunnink
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Alexandra P M Cloherty
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Anusca G Rader
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Kharishma S Patel
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Neeltje A Kootstra
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands
| | - Carla M S Ribeiro
- Amsterdam UMC location University of Amsterdam, Experimental Immunology, Meibergdreef 9, Amsterdam, The Netherlands.
- Amsterdam institute for Immunology & Infectious Diseases, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Huang YJ, Ferrari MW, Lin S, Wang ZH. Recent advances on the Role of Gut Microbiota in the Development of Heart Failure by Mediating Immune Metabolism. Curr Probl Cardiol 2024; 49:102128. [PMID: 37802162 DOI: 10.1016/j.cpcardiol.2023.102128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 09/30/2023] [Indexed: 10/08/2023]
Abstract
The association between gut microbiota and the development of heart failure has become a research hotspot in recent years and the impact of gut microbiota on heart failure has attracted growing interest. From 2006 to 2021, the global research on gut microbiota and heart failure has gradually expanded, indicating a developed and promising research field. There were 40 countries, 196 institutions, and 257 authors involved in the publication on the relationship between gut microbiota and heart failure, respectively. In patients with heart failure, inadequate visceral perfusion leads to ischemia and intestinal edema, which compromise the gut barrier. This subsequently results in the translocation of bacteria and bacterial metabolites into the circulatory system and causes local and systemic inflammatory responses. The gastrointestinal tract contains the largest number of immune cells in the human body and gut microbiota play important roles in the immune system by promoting immune tolerance to symbiotic bacteria. Studies have shown that probiotics can act on gut microorganisms, thereby increasing choline metabolism and reducing plasma TMA and TMAO concentrations, thus inhibiting the development of heart failure. Meanwhile, probiotics induce the production of inflammatory suppressors to maintain gut immune stability and inhibit the progression of heart failure by reducing ventricular remodeling. Here, we review the current understanding of gut microbiota-driven immune dysfunction in experimental and clinical heart failure, as well as the therapeutic interventions that could be used to address these issues.
Collapse
Affiliation(s)
- Yu-Jing Huang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China
| | - Markus W Ferrari
- Clinic of Internal Medicine 1, HSK, Clinic of the City of Wiesbaden and the HELIOS Group, Wiesbaden, Germany.
| | - Shu Lin
- Centre of Neurological and Metabolic Research, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China; Group of Neuroendocrinology, Garvan Institute of Medical Research, Sydney, Australia.
| | - Zhen-Hua Wang
- Department of Cardiology, Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, PR China.
| |
Collapse
|
23
|
Jiang X, Liu H, You Y, Zhong G, Ruan Z, Liao J, Zhang H, Pan J, Tang Z, Hu L. Multi-omics reveals the protective effects of curcumin against AFB1-induced oxidative stress and inflammatory damage in duckling intestines. Comp Biochem Physiol C Toxicol Pharmacol 2024; 276:109815. [PMID: 38061615 DOI: 10.1016/j.cbpc.2023.109815] [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: 10/07/2023] [Revised: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/03/2024]
Abstract
Aflatoxin B1 (AFB1) is the most prevalent and toxic class of aflatoxins, which is considered a significant risk factor for food safety. Curcumin, a phytoconstituent with anti-inflammatory and antioxidant properties, has potential therapeutic value for intestinal inflammatory diseases. In this study, the duckling model susceptible to AFB1 was selected for toxicity testing, aiming to explore the effect of curcumin on AFB1 enterotoxicity and its possible mechanism of action. The results showed that curcumin promoted the growth and development of ducklings and mitigated the changes in morphology and permeability serological index (DAO and D-LA) after AFB1 exposure. Curcumin also mitigated AFB1-induced oxidative stress by activating the Nrf2 pathway, and ameliorated intestinal inflammation by inhibiting the NF-κB/IκB signaling pathway and boosting intestinal autophagy. In terms of gut flora and their metabolites, we found that curcumin supplementation significantly increased the intestinal flora's abundance index and diversity index compared to the AFB1 group, mitigating the decline in the abundance of Actinobacteria and the rise in that of harmful bacteria Clostridia. Furthermore, untargeted metabolomic analysis revealed that the protective effect of curcumin on the intestine was mainly through the regulation of AFB1-induced disorders of lipid metabolism, involving linoleic acid metabolism, α-linolenic acid metabolism, and glycerolipid metabolism. Overall, the enteroprotective effects of curcumin may be of significant value in the future for treating chronic AFB1 poisoning and also provide new therapeutic ideas for other mycotoxicosis.
Collapse
Affiliation(s)
- Xuanxuan Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Medical Devices Research &Testing Center of South China University of Technology, Laboratory Animal Research Center of South China University of Technology, Guangzhou 510006, China
| | - Haiyan Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Yanli You
- College of Life Science, Yantai University, Yantai City 264005, Shandong Province, China
| | - Gaolong Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhiyan Ruan
- School of Pharmacy, Guangdong Food & Drug Vocational College, No. 321, Longdong North Road, Tianhe District, Guangzhou 510520, Guangdong Province, China
| | - Jianzhao Liao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Jiaqiang Pan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China
| | - Lianmei Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
24
|
Villa M, Sanin DE, Apostolova P, Corrado M, Kabat AM, Cristinzio C, Regina A, Carrizo GE, Rana N, Stanczak MA, Baixauli F, Grzes KM, Cupovic J, Solagna F, Hackl A, Globig AM, Hässler F, Puleston DJ, Kelly B, Cabezas-Wallscheid N, Hasselblatt P, Bengsch B, Zeiser R, Sagar, Buescher JM, Pearce EJ, Pearce EL. Prostaglandin E 2 controls the metabolic adaptation of T cells to the intestinal microenvironment. Nat Commun 2024; 15:451. [PMID: 38200005 PMCID: PMC10781727 DOI: 10.1038/s41467-024-44689-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Immune cells must adapt to different environments during the course of an immune response. Here we study the adaptation of CD8+ T cells to the intestinal microenvironment and how this process shapes the establishment of the CD8+ T cell pool. CD8+ T cells progressively remodel their transcriptome and surface phenotype as they enter the gut wall, and downregulate expression of mitochondrial genes. Human and mouse intestinal CD8+ T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We find that the intestinal microenvironment is rich in prostaglandin E2 (PGE2), which drives mitochondrial depolarization in CD8+ T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE2 sensing promotes CD8+ T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell pool. Thus, a PGE2-autophagy-glutathione axis defines the metabolic adaptation of CD8+ T cells to the intestinal microenvironment, to ultimately influence the T cell pool.
Collapse
Affiliation(s)
- Matteo Villa
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany.
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036, Graz, Austria.
| | - David E Sanin
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Petya Apostolova
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine I (Hematology and Oncology), University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Mauro Corrado
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Agnieszka M Kabat
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carmine Cristinzio
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Annamaria Regina
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Department of Life Sciences, University of Trieste, 34128, Trieste, Italy
| | - Gustavo E Carrizo
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Nisha Rana
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Michal A Stanczak
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Francesc Baixauli
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Katarzyna M Grzes
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Jovana Cupovic
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Francesca Solagna
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Alexandra Hackl
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Anna-Maria Globig
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Fabian Hässler
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Daniel J Puleston
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Beth Kelly
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | | | - Peter Hasselblatt
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I (Hematology and Oncology), University Medical Center Freiburg, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Sagar
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Joerg M Buescher
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Edward J Pearce
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Erika L Pearce
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany.
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany.
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
25
|
Kuczyńska M, Moskot M, Gabig-Cimińska M. Insights into Autophagic Machinery and Lysosomal Function in Cells Involved in the Psoriatic Immune-Mediated Inflammatory Cascade. Arch Immunol Ther Exp (Warsz) 2024; 72:aite-2024-0005. [PMID: 38409665 DOI: 10.2478/aite-2024-0005] [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: 10/06/2023] [Accepted: 12/08/2023] [Indexed: 02/28/2024]
Abstract
Impaired autophagy, due to the dysfunction of lysosomal organelles, contributes to maladaptive responses by pathways central to the immune system. Deciphering the immune-inflammatory ecosystem is essential, but remains a major challenge in terms of understanding the mechanisms responsible for autoimmune diseases. Accumulating evidence implicates a role that is played by a dysfunctional autophagy-lysosomal pathway (ALP) and an immune niche in psoriasis (Ps), one of the most common chronic skin diseases, characterized by the co-existence of autoimmune and autoinflammatory responses. The dysregulated autophagy associated with the defective lysosomal system is only one aspect of Ps pathogenesis. It probably cannot fully explain the pathomechanism involved in Ps, but it is likely important and should be seriously considered in Ps research. This review provides a recent update on discoveries in the field. Also, it sheds light on how the dysregulation of intracellular pathways, coming from modulated autophagy and endolysosomal trafficking, characteristic of key players of the disease, i.e., skin-resident cells, as well as circulating immune cells, may be responsible for immune impairment and the development of Ps.
Collapse
Affiliation(s)
- Martyna Kuczyńska
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | - Marta Moskot
- Department of Medical Biology and Genetics, University of Gdańsk, Gdańsk, Poland
| | | |
Collapse
|
26
|
Giansanti M, Theinert T, Boeing SK, Haas D, Schlegel PG, Vacca P, Nazio F, Caruana I. Exploiting autophagy balance in T and NK cells as a new strategy to implement adoptive cell therapies. Mol Cancer 2023; 22:201. [PMID: 38071322 PMCID: PMC10709869 DOI: 10.1186/s12943-023-01893-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/30/2023] [Indexed: 12/18/2023] Open
Abstract
Autophagy is an essential cellular homeostasis pathway initiated by multiple stimuli ranging from nutrient deprivation to viral infection, playing a key role in human health and disease. At present, a growing number of evidence suggests a role of autophagy as a primitive innate immune form of defense for eukaryotic cells, interacting with components of innate immune signaling pathways and regulating thymic selection, antigen presentation, cytokine production and T/NK cell homeostasis. In cancer, autophagy is intimately involved in the immunological control of tumor progression and response to therapy. However, very little is known about the role and impact of autophagy in T and NK cells, the main players in the active fight against infections and tumors. Important questions are emerging: what role does autophagy play on T/NK cells? Could its modulation lead to any advantages? Could specific targeting of autophagy on tumor cells (blocking) and T/NK cells (activation) be a new intervention strategy? In this review, we debate preclinical studies that have identified autophagy as a key regulator of immune responses by modulating the functions of different immune cells and discuss the redundancy or diversity among the subpopulations of both T and NK cells in physiologic context and in cancer.
Collapse
Affiliation(s)
- Manuela Giansanti
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Tobias Theinert
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Sarah Katharina Boeing
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Dorothee Haas
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paul-Gerhardt Schlegel
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Paola Vacca
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy
| | - Francesca Nazio
- Immunology Research Area, Innate Lymphoid Cells Unit, Bambino Gesù Children's Hospital (IRCCS), Rome, Italy.
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Ignazio Caruana
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
27
|
Lin H, Ao H, Guo G, Liu M. The Role and Mechanism of Metformin in Inflammatory Diseases. J Inflamm Res 2023; 16:5545-5564. [PMID: 38026260 PMCID: PMC10680465 DOI: 10.2147/jir.s436147] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 11/17/2023] [Indexed: 12/01/2023] Open
Abstract
Metformin is a classical drug used to treat type 2 diabetes. With the development of research on metformin, it has been found that metformin also has several advantages aside from its hypoglycemic effect, such as anti-inflammatory, anti-aging, anti-cancer, improving intestinal flora, and other effects. The prevention of inflammation is critical because chronic inflammation is associated with numerous diseases of considerable public health. Therefore, there has been growing interest in the role of metformin in treating various inflammatory conditions. However, the precise anti-inflammatory mechanisms of metformin were inconsistent in the reported studies. Thus, this review aims to summarize various currently known possible mechanisms of metformin involved in inflammatory diseases and provide references for the clinical application of metformin.
Collapse
Affiliation(s)
- Huan Lin
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Haiyong Ao
- Jiangxi Key Laboratory of Nanobiomaterials & School of Materials Science and Engineering, East China Jiaotong University, Nanchang, Jiangxi, People’s Republic of China
| | - Guanghua Guo
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| | - Mingzhuo Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
28
|
Gressler AE, Leng H, Zinecker H, Simon AK. Proteostasis in T cell aging. Semin Immunol 2023; 70:101838. [PMID: 37708826 PMCID: PMC10804938 DOI: 10.1016/j.smim.2023.101838] [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: 04/06/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
Aging leads to a decline in immune cell function, which leaves the organism vulnerable to infections and age-related multimorbidities. One major player of the adaptive immune response are T cells, and recent studies argue for a major role of disturbed proteostasis contributing to reduced function of these cells upon aging. Proteostasis refers to the state of a healthy, balanced proteome in the cell and is influenced by synthesis (translation), maintenance and quality control of proteins, as well as degradation of damaged or unwanted proteins by the proteasome, autophagy, lysosome and cytoplasmic enzymes. This review focuses on molecular processes impacting on proteostasis in T cells, and specifically functional or quantitative changes of each of these upon aging. Importantly, we describe the biological consequences of compromised proteostasis in T cells, which range from impaired T cell activation and function to enhancement of inflamm-aging by aged T cells. Finally, approaches to improve proteostasis and thus rejuvenate aged T cells through pharmacological or physical interventions are discussed.
Collapse
Affiliation(s)
- A Elisabeth Gressler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Houfu Leng
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Heidi Zinecker
- Ascenion GmbH, Am Zirkus 1, Bertold-Brecht-Platz 3, 10117 Berlin, Germany
| | - Anna Katharina Simon
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
29
|
Flati I, Di Vito Nolfi M, Dall’Aglio F, Vecchiotti D, Verzella D, Alesse E, Capece D, Zazzeroni F. Molecular Mechanisms Underpinning Immunometabolic Reprogramming: How the Wind Changes during Cancer Progression. Genes (Basel) 2023; 14:1953. [PMID: 37895302 PMCID: PMC10606647 DOI: 10.3390/genes14101953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Metabolism and the immunological state are intimately intertwined, as defense responses are bioenergetically expensive. Metabolic homeostasis is a key requirement for the proper function of immune cell subsets, and the perturbation of the immune-metabolic balance is a recurrent event in many human diseases, including cancer, due to nutrient fluctuation, hypoxia and additional metabolic changes occurring in the tumor microenvironment (TME). Although much remains to be understood in the field of immunometabolism, here, we report the current knowledge on both physiological and cancer-associated metabolic profiles of immune cells, and the main molecular circuits involved in their regulation, highlighting similarities and differences, and emphasizing immune metabolic liabilities that could be exploited in cancer therapy to overcome immune resistance.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L’Aquila, Via Vetoio, Coppito 2, 67100 L’Aquila, Italy; (I.F.); (M.D.V.N.); (F.D.); (D.V.); (D.V.); (E.A.); (F.Z.)
| | | |
Collapse
|
30
|
Chen S, Wang Q, Wang H, Xia S. Endoplasmic reticulum stress in T cell-mediated diseases. Scand J Immunol 2023; 98:e13307. [PMID: 38441291 DOI: 10.1111/sji.13307] [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: 01/17/2023] [Revised: 05/23/2023] [Accepted: 06/18/2023] [Indexed: 03/07/2024]
Abstract
T cells synthesize a large number of proteins during their development, activation, and differentiation. The build-up of misfolded and unfolded proteins in the endoplasmic reticulum, however, causes endoplasmic reticulum (ER) stress. Thus, T cells can maintain ER homeostasis via endoplasmic reticulum-associated degradation, unfolded protein response, and autophagy. In T cell-mediated diseases, such as rheumatoid arthritis, systemic lupus erythematosus, Sjogren's syndrome, type 1 diabetes and vitiligo, ER stress caused by changes in the internal microenvironment can cause disease progression by affecting T cell homeostasis. This review discusses ER stress in T cell formation, activation, differentiation, and T cell-mediated illnesses, and may offer new perspectives on the involvement of T cells in autoimmune disorders and cancer.
Collapse
Affiliation(s)
- Shaodan Chen
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Qiulei Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
31
|
Gan T, Qu S, Zhang H, Zhou X. Modulation of the immunity and inflammation by autophagy. MedComm (Beijing) 2023; 4:e311. [PMID: 37405276 PMCID: PMC10315166 DOI: 10.1002/mco2.311] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/12/2023] [Accepted: 05/26/2023] [Indexed: 07/06/2023] Open
Abstract
Autophagy, a highly conserved cellular self-degradation pathway, has emerged with novel roles in the realms of immunity and inflammation. Genome-wide association studies have unveiled a correlation between genetic variations in autophagy-related genes and heightened susceptibility to autoimmune and inflammatory diseases. Subsequently, substantial progress has been made in unraveling the intricate involvement of autophagy in immunity and inflammation through functional studies. The autophagy pathway plays a crucial role in both innate and adaptive immunity, encompassing various key functions such as pathogen clearance, antigen processing and presentation, cytokine production, and lymphocyte differentiation and survival. Recent research has identified novel approaches in which the autophagy pathway and its associated proteins modulate the immune response, including noncanonical autophagy. This review provides an overview of the latest advancements in understanding the regulation of immunity and inflammation through autophagy. It summarizes the genetic associations between variants in autophagy-related genes and a range of autoimmune and inflammatory diseases, while also examining studies utilizing transgenic animal models to uncover the in vivo functions of autophagy. Furthermore, the review delves into the mechanisms by which autophagy dysregulation contributes to the development of three common autoimmune and inflammatory diseases and highlights the potential for autophagy-targeted therapies.
Collapse
Affiliation(s)
- Ting Gan
- Renal DivisionPeking University First HospitalBeijingChina
- Peking University Institute of NephrologyBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)Ministry of EducationBeijingChina
| | - Shu Qu
- Renal DivisionPeking University First HospitalBeijingChina
- Peking University Institute of NephrologyBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)Ministry of EducationBeijingChina
| | - Hong Zhang
- Renal DivisionPeking University First HospitalBeijingChina
- Peking University Institute of NephrologyBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)Ministry of EducationBeijingChina
| | - Xu‐jie Zhou
- Renal DivisionPeking University First HospitalBeijingChina
- Peking University Institute of NephrologyBeijingChina
- Key Laboratory of Renal DiseaseMinistry of Health of ChinaBeijingChina
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University)Ministry of EducationBeijingChina
| |
Collapse
|
32
|
Liu Q, Zheng Y, Goronzy JJ, Weyand CM. T cell aging as a risk factor for autoimmunity. J Autoimmun 2023; 137:102947. [PMID: 36357240 PMCID: PMC10164202 DOI: 10.1016/j.jaut.2022.102947] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 10/23/2022] [Indexed: 11/09/2022]
Abstract
Immune aging is a complex process rendering the host susceptible to cancer, infection, and insufficient tissue repair. Many autoimmune diseases preferentially occur during the second half of life, counterintuitive to the concept of excess adaptive immunity driving immune-mediated tissue damage. T cells are particularly susceptible to aging-imposed changes, as they are under extreme proliferative pressure to fulfill the demands of clonal expansion and of homeostatic T cell repopulation. T cells in older adults have a footprint of genetic and epigenetic changes, lack mitochondrial fitness, and fail to maintain proteostasis, diverging them from host protection to host injury. Here, we review recent progress in understanding how the human T-cell system ages and the evidence detailing how T cell aging contributes to autoimmune conditions. T cell aging is now recognized as a risk determinant in two prototypic autoimmune syndromes; rheumatoid arthritis and giant cell arteritis. The emerging concept adds susceptibility to autoimmune and autoinflammatory disease to the spectrum of aging-imposed adaptations and opens new opportunities for immunomodulatory therapy by restoring the functional intactness of aging T cells.
Collapse
Affiliation(s)
- Qingxiang Liu
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yanyan Zheng
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA
| | - Jorg J Goronzy
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA
| | - Cornelia M Weyand
- Department of Medicine, Mayo Clinic Alix School of Medicine, Rochester, MN 55905, USA; Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA; Department of Cardiovascular Medicine, Mayo Alix School of Medicine, Rochester, MN, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94306, USA.
| |
Collapse
|
33
|
Kabat AM, Pearce EL, Pearce EJ. Metabolism in type 2 immune responses. Immunity 2023; 56:723-741. [PMID: 37044062 PMCID: PMC10938369 DOI: 10.1016/j.immuni.2023.03.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
The immune response is tailored to the environment in which it takes place. Immune cells sense and adapt to changes in their surroundings, and it is now appreciated that in addition to cytokines made by stromal and epithelial cells, metabolic cues provide key adaptation signals. Changes in immune cell activation states are linked to changes in cellular metabolism that support function. Furthermore, metabolites themselves can signal between as well as within cells. Here, we discuss recent progress in our understanding of how metabolic regulation relates to type 2 immunity firstly by considering specifics of metabolism within type 2 immune cells and secondly by stressing how type 2 immune cells are integrated more broadly into the metabolism of the organism as a whole.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Erika L Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Edward J Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| |
Collapse
|
34
|
Ye P, Feng L, Zhang D, Li R, Wen Y, Tong X, Shi S, Dong C. Metformin Ameliorates D-Galactose-Induced Senescent Human Bone Marrow-Derived Mesenchymal Stem Cells by Enhancing Autophagy. Stem Cells Int 2023; 2023:1429642. [PMID: 37035446 PMCID: PMC10079386 DOI: 10.1155/2023/1429642] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/16/2023] [Accepted: 03/11/2023] [Indexed: 04/04/2023] Open
Abstract
Human bone marrow-derived mesenchymal stem cells (hBMSCs) are promising candidates for stem cell therapy in clinical trials. Applications of hBMSCs in clinical therapy are limited by cellular senescence due to long-term ex vivo expansion. Metformin, an oral hypoglycemic drug for type 2 diabetes, has been shown to have antiaging effects. However, the mechanisms of metformin in antiaging treatment remain controversial. Here, we used D-galactose (D-gal) to establish an appropriate model of senescent hBMSCs to explore the antiaging effects of metformin. Following metformin treatment with a low concentration range, senescence phenotypes induced by D-gal significantly changed, including generation of reactive oxygen species (ROS), loss of mitochondrial membrane potential (MMP), and cell cycle arrest. In contrast, no apparent change was found in unsenescent hBMSCs. Furthermore, the results show that activation of 5'AMP-activated protein kinase (AMPK) by metformin enhances cell autophagy in senescent hBMSCs. These findings suggest that metformin exerts antiaging function within the low concentration range by enhancing autophagy and exhibits potential benefits for clinical stem cell therapy by ameliorating the ex vivo replicative senescence of hBMSCs.
Collapse
Affiliation(s)
- Pingting Ye
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Lei Feng
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Dan Zhang
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Ruihao Li
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Yixuan Wen
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Xiaohan Tong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Shuo Shi
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| | - Chunyan Dong
- Department of Oncology, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200120, China
| |
Collapse
|
35
|
Richter FC, Friedrich M, Kampschulte N, Piletic K, Alsaleh G, Zummach R, Hecker J, Pohin M, Ilott N, Guschina I, Wideman SK, Johnson E, Borsa M, Hahn P, Morriseau C, Hammock BD, Schipper HS, Edwards CM, Zechner R, Siegmund B, Weidinger C, Schebb NH, Powrie F, Simon AK. Adipocyte autophagy limits gut inflammation by controlling oxylipin and IL-10. EMBO J 2023; 42:e112202. [PMID: 36795015 PMCID: PMC10015370 DOI: 10.15252/embj.2022112202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 02/17/2023] Open
Abstract
Lipids play a major role in inflammatory diseases by altering inflammatory cell functions, either through their function as energy substrates or as lipid mediators such as oxylipins. Autophagy, a lysosomal degradation pathway that limits inflammation, is known to impact on lipid availability, however, whether this controls inflammation remains unexplored. We found that upon intestinal inflammation visceral adipocytes upregulate autophagy and that adipocyte-specific loss of the autophagy gene Atg7 exacerbates inflammation. While autophagy decreased lipolytic release of free fatty acids, loss of the major lipolytic enzyme Pnpla2/Atgl in adipocytes did not alter intestinal inflammation, ruling out free fatty acids as anti-inflammatory energy substrates. Instead, Atg7-deficient adipose tissues exhibited an oxylipin imbalance, driven through an NRF2-mediated upregulation of Ephx1. This shift reduced secretion of IL-10 from adipose tissues, which was dependent on the cytochrome P450-EPHX pathway, and lowered circulating levels of IL-10 to exacerbate intestinal inflammation. These results suggest an underappreciated fat-gut crosstalk through an autophagy-dependent regulation of anti-inflammatory oxylipins via the cytochrome P450-EPHX pathway, indicating a protective effect of adipose tissues for distant inflammation.
Collapse
Affiliation(s)
| | - Matthias Friedrich
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Nadja Kampschulte
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Klara Piletic
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Ghada Alsaleh
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Julia Hecker
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Mathilde Pohin
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Nicholas Ilott
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | | | - Sarah Karin Wideman
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe HospitalUniversity of OxfordOxfordUK
| | - Errin Johnson
- The Dunn School of PathologyUniversity of OxfordOxfordUK
| | - Mariana Borsa
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Paula Hahn
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Christophe Morriseau
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Bruce D Hammock
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer CenterUniversity of CaliforniaDavisCAUSA
| | - Henk Simon Schipper
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Claire M Edwards
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
- Nuffield Department of Surgical Sciences, Botnar Research CentreUniversity of OxfordOxfordUK
| | - Rudolf Zechner
- Institute of Molecular BiosciencesUniversity of GrazGrazAustria
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Carl Weidinger
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin and Berlin Institute of HealthBerlinGermany
- Department of Gastroenterology, Infectious Diseases and RheumatologyCampus Benjamin FranklinBerlinGermany
| | - Nils Helge Schebb
- Faculty of Mathematics and Natural SciencesUniversity of WuppertalWuppertalGermany
| | - Fiona Powrie
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
| | - Anna Katharina Simon
- Kennedy Institute of RheumatologyUniversity of OxfordOxfordUK
- Max Delbrück CenterBerlinGermany
| |
Collapse
|
36
|
Villa M, Sanin DE, Apostolova P, Corrado M, Kabat AM, Cristinzio C, Regina A, Carrizo GE, Rana N, Stanczak MA, Baixauli F, Grzes KM, Cupovic J, Solagna F, Hackl A, Globig AM, Hässler F, Puleston DJ, Kelly B, Cabezas-Wallscheid N, Hasselblatt P, Bengsch B, Zeiser R, Sagar, Buescher JM, Pearce EJ, Pearce EL. Prostaglandin E 2 controls the metabolic adaptation of T cells to the intestinal microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532431. [PMID: 36993703 PMCID: PMC10054978 DOI: 10.1101/2023.03.13.532431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Immune cells must adapt to different environments during the course of an immune response. We studied the adaptation of CD8 + T cells to the intestinal microenvironment and how this process shapes their residency in the gut. CD8 + T cells progressively remodel their transcriptome and surface phenotype as they acquire gut residency, and downregulate expression of mitochondrial genes. Human and mouse gut-resident CD8 + T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We found that the intestinal microenvironment is rich in prostaglandin E 2 (PGE 2 ), which drives mitochondrial depolarization in CD8 + T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE 2 sensing promotes CD8 + T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell population. Thus, a PGE 2 -autophagy-glutathione axis defines the metabolic adaptation of CD8 + T cells to the intestinal microenvironment, to ultimately influence the T cell pool.
Collapse
|
37
|
Hasan KMM, Haque MA. Autophagy and Its Lineage-Specific Roles in the Hematopoietic System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:8257217. [PMID: 37180758 PMCID: PMC10171987 DOI: 10.1155/2023/8257217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 02/26/2023] [Accepted: 03/17/2023] [Indexed: 05/16/2023]
Abstract
Autophagy is a dynamic process that regulates the selective and nonselective degradation of cytoplasmic components, such as damaged organelles and protein aggregates inside lysosomes to maintain tissue homeostasis. Different types of autophagy including macroautophagy, microautophagy, and chaperon-mediated autophagy (CMA) have been implicated in a variety of pathological conditions, such as cancer, aging, neurodegeneration, and developmental disorders. Furthermore, the molecular mechanism and biological functions of autophagy have been extensively studied in vertebrate hematopoiesis and human blood malignancies. In recent years, the hematopoietic lineage-specific roles of different autophagy-related (ATG) genes have gained more attention. The evolution of gene-editing technology and the easy access nature of hematopoietic stem cells (HSCs), hematopoietic progenitors, and precursor cells have facilitated the autophagy research to better understand how ATG genes function in the hematopoietic system. Taking advantage of the gene-editing platform, this review has summarized the roles of different ATGs at the hematopoietic cell level, their dysregulation, and pathological consequences throughout hematopoiesis.
Collapse
Affiliation(s)
- Kazi Md Mahmudul Hasan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
- Department of Neurology, David Geffen School of Medicine, The University of California, 710 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Md Anwarul Haque
- Department of Biotechnology and Genetic Engineering, Islamic University, Kushtia 7003, Bangladesh
| |
Collapse
|
38
|
Li Y, Law HKW. Deciphering the role of autophagy in the immunopathogenesis of inflammatory bowel disease. Front Pharmacol 2022; 13:1070184. [DOI: 10.3389/fphar.2022.1070184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a typical immune-mediated chronic inflammatory disorder. Following the industrialization and changes in lifestyle, the incidence of IBD in the world is rising, which makes health concerns and heavy burdens all over the world. However, the pathogenesis of IBD remains unclear, and the current understanding of the pathogenesis involves dysregulation of mucosal immunity, gut microbiome dysbiosis, and gut barrier defect based on genetic susceptibility and environmental triggers. In recent years, autophagy has emerged as a key mechanism in IBD development and progression because Genome-Wide Association Study revealed the complex interactions of autophagy in IBD, especially immunopathogenesis. Besides, autophagy markers are also suggested to be potential biomarkers and target treatment in IBD. This review summarizes the autophagy-related genes regulating immune response in IBD. Furthermore, we explore the evolving evidence that autophagy interacts with intestinal epithelial and immune cells to contribute to the inflammatory changes in IBD. Finally, we discuss how novel discovery could further advance our understanding of the role of autophagy and inform novel therapeutic strategies in IBD.
Collapse
|
39
|
Pant A, Yao X, Lavedrine A, Viret C, Dockterman J, Chauhan S, Chong-Shan Shi, Manjithaya R, Cadwell K, Kufer TA, Kehrl JH, Coers J, Sibley LD, Faure M, Taylor GA, Chauhan S. Interactions of Autophagy and the Immune System in Health and Diseases. AUTOPHAGY REPORTS 2022; 1:438-515. [PMID: 37425656 PMCID: PMC10327624 DOI: 10.1080/27694127.2022.2119743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Autophagy is a highly conserved process that utilizes lysosomes to selectively degrade a variety of intracellular cargo, thus providing quality control over cellular components and maintaining cellular regulatory functions. Autophagy is triggered by multiple stimuli ranging from nutrient starvation to microbial infection. Autophagy extensively shapes and modulates the inflammatory response, the concerted action of immune cells, and secreted mediators aimed to eradicate a microbial infection or to heal sterile tissue damage. Here, we first review how autophagy affects innate immune signaling, cell-autonomous immune defense, and adaptive immunity. Then, we discuss the role of non-canonical autophagy in microbial infections and inflammation. Finally, we review how crosstalk between autophagy and inflammation influences infectious, metabolic, and autoimmune disorders.
Collapse
Affiliation(s)
- Aarti Pant
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Xiaomin Yao
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Aude Lavedrine
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Christophe Viret
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Jake Dockterman
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
| | - Swati Chauhan
- Cell biology and Infectious diseases, Institute of Life Sciences, Bhubaneswar, India
| | - Chong-Shan Shi
- Laboratory of Immunoregulation, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, India
| | - Ken Cadwell
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University Grossman School of Medicine, New York, New York, United States of America
- Department of Microbiology, New York University Grossman School of Medicine, New York, New York, United States of America
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States of America
| | - Thomas A. Kufer
- Department of Immunology, Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany
| | - John H. Kehrl
- Laboratory of Immunoregulation, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jörn Coers
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina, USA
| | - L. David Sibley
- Department of Molecular Microbiology, Washington University Sch. Med., St Louis, MO, 63110, USA
| | - Mathias Faure
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, F-69007, Lyon, France
- Equipe Labellisée par la Fondation pour la Recherche Médicale, FRM
| | - Gregory A Taylor
- Department of Immunology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Genetics and Microbiology, Duke University, Medical Center, Durham, North Carolina, USA
- Department of Molecular Microbiology, Washington University Sch. Med., St Louis, MO, 63110, USA
- Geriatric Research, Education, and Clinical Center, VA Health Care Center, Durham, North Carolina, USA
- Departments of Medicine, Division of Geriatrics, and Center for the Study of Aging and Human Development, Duke University, Medical Center, Durham, North Carolina, USA
| | - Santosh Chauhan
- Cell biology and Infectious diseases, Institute of Life Sciences, Bhubaneswar, India
- CSIR–Centre For Cellular And Molecular Biology (CCMB), Hyderabad, Telangana
| |
Collapse
|
40
|
Prospect of ULK1 modulators in targeting regulatory T cells. Bioorg Chem 2022; 129:106141. [PMID: 36137312 DOI: 10.1016/j.bioorg.2022.106141] [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: 05/17/2022] [Revised: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022]
Abstract
Regulatory T (Treg) cells play an instrumental role in coordinating immune homeostasis via potent inhibitory effects. Defects in Treg cells lead to autoimmunity, but an overwhelming proportion of Treg cells encourages cancer progression. Hence, targeting Treg cells has emerged as a promising approach for mitigating disease severity. Recent studies have revealed that kinases are a critical component for tuning the fate of Treg cells, but the entire network of Treg-modulating kinases is still unclear. Here, we propose that the autophagy-activating UNC-51-like kinase 1 (ULK1) is a candidate for Treg cell modulation. While accumulating evidence has highlighted the role of autophagy-related kinases in Treg cells, the ULK1-Treg cell axis is yet to be examined. In this review, we predicted the potential role of ULK1 in Treg cell modulation. Furthermore, we summarized current ULK1 activators and inhibitors that can be investigated as Treg-targeting strategies, which might have beneficial outcomes in autoimmunity and cancer.
Collapse
|
41
|
Transcription factor EB coordinates environmental cues to regulate T regulatory cells' mitochondrial fitness and function. Proc Natl Acad Sci U S A 2022; 119:e2205469119. [PMID: 35895684 PMCID: PMC9351480 DOI: 10.1073/pnas.2205469119] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
T regulatory (Treg) cells are essential for self-tolerance whereas they are detrimental for dampening the host anti-tumor immunity. How Treg cells adapt to environmental signals to orchestrate their homeostasis and functions remains poorly understood. Here, we identified that transcription factor EB (TFEB) is induced by host nutrition deprivation or interleukin (IL)-2 in CD4+ T cells. The loss of TFEB in Treg cells leads to reduced Treg accumulation and impaired Treg function in mouse models of cancer and autoimmune disease. TFEB intrinsically regulates genes involved in Treg cell differentiation and mitochondria function while it suppresses expression of proinflammatory cytokines independently of its established roles in autophagy. This coordinated action is required for mitochondria integrity and appropriate lipid metabolism in Treg cells. These findings identify TFEB as a critical regulator for orchestrating Treg generation and function, which may contribute to the adaptive responses of T cells to local environmental cues.
Collapse
|
42
|
Caza T, Wijewardena C, Al-Rabadi L, Perl A. Cell type-specific mechanistic target of rapamycin-dependent distortion of autophagy pathways in lupus nephritis. Transl Res 2022; 245:55-81. [PMID: 35288362 PMCID: PMC9240418 DOI: 10.1016/j.trsl.2022.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/07/2022] [Accepted: 03/07/2022] [Indexed: 01/02/2023]
Abstract
Pro-inflammatory immune system development, metabolomic defects, and deregulation of autophagy play interconnected roles in driving the pathogenesis of systemic lupus erythematosus (SLE). Lupus nephritis (LN) is a leading cause of morbidity and mortality in SLE. While the causes of SLE have not been clearly delineated, skewing of T and B cell differentiation, activation of antigen-presenting cells, production of antinuclear autoantibodies and pro-inflammatory cytokines are known to contribute to disease development. Underlying this process are defects in autophagy and mitophagy that cause the accumulation of oxidative stress-generating mitochondria which promote necrotic cell death. Autophagy is generally inhibited by the activation of the mammalian target of rapamycin (mTOR), a large protein kinase that underlies abnormal immune cell lineage specification in SLE. Importantly, several autophagy-regulating genes, including ATG5 and ATG7, as well as mitophagy-regulating HRES-1/Rab4A have been linked to lupus susceptibility and molecular pathogenesis. Moreover, genetically-driven mTOR activation has been associated with fulminant lupus nephritis. mTOR activation and diminished autophagy promote the expansion of pro-inflammatory Th17, Tfh and CD3+CD4-CD8- double-negative (DN) T cells at the expense of CD8+ effector memory T cells and CD4+ regulatory T cells (Tregs). mTOR activation and aberrant autophagy also involve renal podocytes, mesangial cells, endothelial cells, and tubular epithelial cells that may compromise end-organ resistance in LN. Activation of mTOR complexes 1 (mTORC1) and 2 (mTORC2) has been identified as biomarkers of disease activation and predictors of disease flares and prognosis in SLE patients with and without LN. This review highlights recent advances in molecular pathogenesis of LN with a focus on immuno-metabolic checkpoints of autophagy and their roles in pathogenesis, prognosis and selection of targets for treatment in SLE.
Collapse
Affiliation(s)
| | - Chathura Wijewardena
- Departments of Medicine, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York
| | - Laith Al-Rabadi
- Department of Medicine, University of Utah, Salt Lake City, Utah
| | - Andras Perl
- Departments of Medicine, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York; Biochemistry and Molecular Biology, Neuroscience and Physiology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York; Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York.
| |
Collapse
|
43
|
Jeong J, Choi YJ, Lee HK. The Role of Autophagy in the Function of CD4 + T Cells and the Development of Chronic Inflammatory Diseases. Front Pharmacol 2022; 13:860146. [PMID: 35392563 PMCID: PMC8981087 DOI: 10.3389/fphar.2022.860146] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/07/2022] [Indexed: 12/29/2022] Open
Abstract
Uncontrolled acute inflammation progresses to persistent inflammation that leads to various chronic inflammatory diseases, including asthma, Crohn’s disease, rheumatoid arthritis, multiple sclerosis, and systemic lupus erythematosus. CD4+ T cells are key immune cells that determine the development of these chronic inflammatory diseases. CD4+ T cells orchestrate adaptive immune responses by producing cytokines and effector molecules. These functional roles of T cells vary depending on the surrounding inflammatory or anatomical environment. Autophagy is an important process that can regulate the function of CD4+ T cells. By lysosomal degradation of cytoplasmic materials, autophagy mediates CD4+ T cell-mediated immune responses, including cytokine production, proliferation, and differentiation. Furthermore, through canonical processes involving autophagy machinery, autophagy also contributes to the development of chronic inflammatory diseases. Therefore, a targeted intervention of autophagy processes could be used to treat chronic inflammatory diseases. This review focuses on the role of autophagy via CD4+ T cells in the pathogenesis and treatment of such diseases. In particular, we explore the underlying mechanisms of autophagy in the regulation of CD4+ T cell metabolism, survival, development, proliferation, differentiation, and aging. Furthermore, we suggest that autophagy-mediated modulation of CD4+ T cells is a promising therapeutic target for treating chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jiung Jeong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Young Joon Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| |
Collapse
|
44
|
Chen PM, Tsokos GC. Mitochondria in the Pathogenesis of Systemic Lupus Erythematosus. Curr Rheumatol Rep 2022; 24:88-95. [PMID: 35290598 DOI: 10.1007/s11926-022-01063-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2022] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by autoantibody production and inflammation in multiple organs. In this article, we present data on how various mitochondria pathologies are involved in the pathogenesis of the disease including the fact that they serve as a reservoir of autoantigens which contribute to the upending of lymphocyte tolerance. RECENT FINDINGS Mitochondrial DNA from various cell sources, including neutrophil extracellular traps, platelets, and red blood cells, elicits the production of type I interferon which contributes to breaking of peripheral tolerance. Mitochondrial DNA also serves as autoantigen targeted by autoantibodies. Mutations of mitochondrial DNA triggered by reactive oxygen species induce T cell cross-reactivity against self-antigens. Selective gene polymorphisms that regulate mitochondrial apoptosis in autoreactive B and T cells represent another key aspect in the induction of autoimmunity. Various mitochondrial abnormalities, including changes in mitochondrial function, oxidative stress, genetic polymorphism, mitochondrial DNA mutations, and apoptosis pathways, are each linked to different aspects of lupus pathogenesis. However, whether targeting these mitochondrial pathologies can be used to harness autoimmunity remains to be explored.
Collapse
Affiliation(s)
- Ping-Min Chen
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
45
|
González-Osuna L, Sierra-Cristancho A, Cafferata EA, Melgar-Rodríguez S, Rojas C, Carvajal P, Cortez C, Vernal R. Senescent CD4 +CD28 - T Lymphocytes as a Potential Driver of Th17/Treg Imbalance and Alveolar Bone Resorption during Periodontitis. Int J Mol Sci 2022; 23:ijms23052543. [PMID: 35269683 PMCID: PMC8910032 DOI: 10.3390/ijms23052543] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 01/10/2023] Open
Abstract
Senescent cells express a senescence-associated secretory phenotype (SASP) with a pro-inflammatory bias, which contributes to the chronicity of inflammation. During chronic inflammatory diseases, infiltrating CD4+ T lymphocytes can undergo cellular senescence and arrest the surface expression of CD28, have a response biased towards T-helper type-17 (Th17) of immunity, and show a remarkable ability to induce osteoclastogenesis. As a cellular counterpart, T regulatory lymphocytes (Tregs) can also undergo cellular senescence, and CD28− Tregs are able to express an SASP secretome, thus severely altering their immunosuppressive capacities. During periodontitis, the persistent microbial challenge and chronic inflammation favor the induction of cellular senescence. Therefore, senescence of Th17 and Treg lymphocytes could contribute to Th17/Treg imbalance and favor the tooth-supporting alveolar bone loss characteristic of the disease. In the present review, we describe the concept of cellular senescence; particularly, the one produced during chronic inflammation and persistent microbial antigen challenge. In addition, we detail the different markers used to identify senescent cells, proposing those specific to senescent T lymphocytes that can be used for periodontal research purposes. Finally, we discuss the existing literature that allows us to suggest the potential pathogenic role of senescent CD4+CD28− T lymphocytes in periodontitis.
Collapse
Affiliation(s)
- Luis González-Osuna
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
- Correspondence: (L.G.-O.); (R.V.)
| | - Alfredo Sierra-Cristancho
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
- Faculty of Dentistry, Universidad Andres Bello, Santiago 8370035, Chile
| | - Emilio A. Cafferata
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
- Department of Periodontology, School of Dentistry, Universidad Científica del Sur, Lima 15067, Peru
| | - Samanta Melgar-Rodríguez
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile;
| | - Carolina Rojas
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
| | - Paola Carvajal
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile;
| | - Cristian Cortez
- Center for Genomics and Bioinformatics, Faculty of Sciences, Universidad Mayor, Santiago 8580745, Chile;
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile; (A.S.-C.); (E.A.C.); (S.M.-R.); (C.R.)
- Department of Conservative Dentistry, Faculty of Dentistry, Universidad de Chile, Santiago 8380492, Chile;
- Correspondence: (L.G.-O.); (R.V.)
| |
Collapse
|
46
|
Effects of Herb-Partitioned Moxibustion on Autophagy and Immune Activity in the Colon Tissue of Rats with Crohn’s Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3534874. [PMID: 35126598 PMCID: PMC8816589 DOI: 10.1155/2022/3534874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/11/2022] [Indexed: 11/18/2022]
Abstract
Objective To investigate the mechanism of action of herb-partitioned moxibustion on CD from the perspective of autophagy and immunity. Methods The expression of microtubule-associated protein LC3II and SQSTM1/p62 in the colon tissues was detected by immunohistochemistry. Western blot was used to detect the expression of autophagic and immune-related proteins in the colon, such as LC3II, SQSTM1/p62, Beclin1, ATG16L1, NOD2, IRGM, IL-1β, IL-17, and TNF-β. mRNA levels of immune factors, such as IL-1β, IL-17, and TNF-β, and autophagy signaling molecules, such as PI3KC, AKT1, LKB1, and mTOR, were detected by RT-qPCR. Results Herb-partitioned moxibustion reduced the protein levels of ATG16L1, NOD2, IRGM, LC3II, and Beclin1 (P < 0.01) and both the protein and mRNA levels of IL-1β, IL-17, and TNF-β in CD rats (P < 0.01 or P < 0.05), and it also increased the expression of SQSTM1/p62 protein (P < 0.01). The modulatory effects of herb-partitioned moxibustion on ATG16L1, NOD2, IRGM, LC3II, TNF-β, and IL-17 protein and IL-1β protein and mRNA were better than those of mesalazine (P < 0.01 or P < 0.05). Herb-partitioned moxibustion also reduced colon PI3KC, AKT1, and LKB1 mRNA expressions in CD rats (P < 0.01 or P < 0.05) and increased mTOR protein expression (P < 0.05). And the modulatory effect of herb-partitioned moxibustion on AKT1 mRNA was better than that of mesalazine (P < 0.05). Conclusion Herb-partitioned moxibustion may inhibit excessively activated autophagy and modulate the expression of immune-related factors by regulating the LKB1-mTOR-PI3KC signal transduction networks, thereby alleviating intestinal inflammation in CD rats.
Collapse
|
47
|
Pimentel-Muiños FX. Autophagy in the gastrointestinal system and cross talk with microbiota. AUTOPHAGY IN HEALTH AND DISEASE 2022:321-333. [DOI: 10.1016/b978-0-12-822003-0.00016-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
48
|
El-Sahhar S, Varga-Weisz P. The gut microbiome in health and disease: Inflammatory bowel diseases. ADV ECOL RES 2022. [DOI: 10.1016/bs.aecr.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
49
|
Bai Y, Guan F, Zhu F, Jiang C, Xu X, Zheng F, Liu W, Lei J. IL-33/ST2 Axis Deficiency Exacerbates Hepatic Pathology by Regulating Treg and Th17 Cells in Murine Schistosomiasis Japonica. J Inflamm Res 2021; 14:5981-5998. [PMID: 34815688 PMCID: PMC8604654 DOI: 10.2147/jir.s336404] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 10/27/2021] [Indexed: 12/22/2022] Open
Abstract
Purpose Schistosoma japonicum-infected IL-33 and ST2 gene deficiency (IL-33−/− and ST2−/−, respectively) mice were used to explore the role of the IL-33/ST2 axis in liver pathology targeting regulatory T cells (Treg)/T helper 17 cells (Th17). Materials and Methods Each mouse was infected percutaneously with 20 S. japonicum cercariae. Hepatic mass index (HMI), liver egg granulomas, hepatic fibrosis biomarkers and serum levels of alanine aminotransferase (ALT) were investigated. Treg and Th17 frequency was determined by flow cytometry. Expressions of Foxp3, ST2, TGF-β1, IL-10, RORγt, and IL-17A were measured via quantitative real-time polymerase chain reaction (qRT-PCR). Concentrations of TGF-β1, IL-10 and IL-17A were tested with ELISA. In vitro experiments, mRNA expressions of Foxp3, TGF-β1, IL-10, Atg5, Beclin-1 and p62 associated with polarization of Treg by recombinant mouse IL-33 (rmIL-33) were detected by qRT-PCR. Results An increased expression of IL-33/ST2 was shown in S. japonicum-infected mice. Deficiency of IL-33 or ST2 gene led to an aggravated liver pathology, which was evidenced by elevated hepatic granuloma volume, HMI and ALT levels and fibrosis, which was demonstrated by increased hepatic collagen deposition in the infected mice. Injection of rmIL-33 into the infected IL-33−/− mice strongly abrogated the liver pathology and fibrosis, whereas no detectable effect with injecting rmIL-33 into the infected ST2−/− mice. Furthermore, depletion of the IL-33/ST2 axis inhibited Treg, accompanied by increased Th17. rmIL-33 treatment upregulated Treg and downregulated Th17 in the infected IL-33−/− mice, while no effect in the infected ST2−/− mice. rmIL-33 led to elevated expressions of Atg5, Beclin-1 and inhibited expression of p62 in expansion of Treg. Conclusion The IL-33/ST2 axis plays a protective role in S. japonicum infected mice, which is closely related to increasing Treg responses as well as suppressing Th17 responses. Expansion of Treg by IL-33 may be associated with its regulation of autophagy.
Collapse
Affiliation(s)
- Yang Bai
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Feifan Zhu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chunjie Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - XiaoXiao Xu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Wenqi Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| |
Collapse
|
50
|
Daneshmandi S, Cassel T, Higashi RM, Fan TWM, Seth P. 6-Phosphogluconate dehydrogenase (6PGD), a key checkpoint in reprogramming of regulatory T cells metabolism and function. eLife 2021; 10:e67476. [PMID: 34709178 PMCID: PMC8553335 DOI: 10.7554/elife.67476] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 10/17/2021] [Indexed: 12/29/2022] Open
Abstract
Cellular metabolism has key roles in T cells differentiation and function. CD4+ T helper-1 (Th1), Th2, and Th17 subsets are highly glycolytic while regulatory T cells (Tregs) use glucose during expansion but rely on fatty acid oxidation for function. Upon uptake, glucose can enter pentose phosphate pathway (PPP) or be used in glycolysis. Here, we showed that blocking 6-phosphogluconate dehydrogenase (6PGD) in the oxidative PPP resulted in substantial reduction of Tregs suppressive function and shifts toward Th1, Th2, and Th17 phenotypes which led to the development of fetal inflammatory disorder in mice model. These in turn improved anti-tumor responses and worsened the outcomes of colitis model. Metabolically, 6PGD blocked Tregs showed improved glycolysis and enhanced non-oxidative PPP to support nucleotide biosynthesis. These results uncover critical role of 6PGD in modulating Tregs plasticity and function, which qualifies it as a novel metabolic checkpoint for immunotherapy applications.
Collapse
Affiliation(s)
- Saeed Daneshmandi
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
- Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
| | - Teresa Cassel
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Richard M Higashi
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, University of KentuckyLexingtonUnited States
| | - Pankaj Seth
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
- Division of Interdisciplinary Medicine, Beth Israel Deaconess Medical Center, Harvard Medical SchoolBostonUnited States
| |
Collapse
|