1
|
Chen Q, Gao Y, Li F, Yuan L. The role of gut-islet axis in pancreatic islet function and glucose homeostasis. Diabetes Obes Metab 2025; 27:1676-1692. [PMID: 39916498 PMCID: PMC11885102 DOI: 10.1111/dom.16225] [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: 09/03/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 03/08/2025]
Abstract
The gastrointestinal tract plays a vital role in the occurrence and treatment of metabolic diseases. Recent studies have convincingly demonstrated a bidirectional axis of communication between the gut and islets, enabling the gut to influence glucose metabolism and energy homeostasis in animals strongly. The 'gut-islet axis' is an essential endocrine signal axis that regulates islet function through the dialogue between intestinal microecology and endocrine metabolism. The discovery of glucagon-like peptide-1 (GLP-1), gastric inhibitory peptide (GIP) and other gut hormones has initially set up a bridge between gut and islet cells. However, the influence of other factors remains largely unknown, such as the homeostasis of the gut microbiota and the integrity of the gut barrier. Although gut microbiota primarily resides and affect intestinal function, they also affect extra-intestinal organs by absorbing and transferring metabolites derived from microorganisms. As a result of this transfer, islets may be continuously exposed to gut-derived metabolites and components. Changes in the composition of gut microbiota can damage the intestinal barrier function to varying degrees, resulting in increased intestinal permeability to bacteria and their derivatives. All these changes contribute to the severe disturbance of critical metabolic pathways in peripheral tissues and organs. In this review, we have outlined the different gut-islet axis signalling mechanisms associated with metabolism and summarized the latest progress in the complex signalling molecules of the gut and gut microbiota. In addition, we will discuss the impact of the gut renin-angiotensin system (RAS) on the various components of the gut-islet axis that regulate energy and glucose homeostasis. This work also indicates that therapeutic approaches aiming to restore gut microbial homeostasis, such as probiotics and faecal microbiota transplantation (FMT), have shown great potential in improving treatment outcomes, enhancing patient prognosis and slowing down disease progression. Future research should further uncover the molecular links between the gut-islet axis and the gut microbiota and explore individualized microbial treatment strategies, which will provide an innovative perspective and approach for the diagnosis and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Qi Chen
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yuanyuan Gao
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Fangyu Li
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
2
|
Skjølberg C, Degani L, Sileikaite-Morvaközi I, Hawkins CL. Oxidative modification of extracellular histones by hypochlorous acid modulates their ability to induce β-cell dysfunction. Free Radic Biol Med 2025; 230:209-221. [PMID: 39956473 DOI: 10.1016/j.freeradbiomed.2025.02.018] [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: 01/09/2025] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 02/18/2025]
Abstract
Histones are nuclear proteins that play a key role in chromatin assembly and regulation of gene expression by their ability to bind to DNA. Histones can also be released from cells owing to necrosis or extracellular trap release from neutrophils (NETs) and other immune cells. The presence of histones in the extracellular environment has implications for many pathologies, including diabetes mellitus, owing to the cytotoxic nature of these proteins, and their ability to promote inflammation. NETs also contain myeloperoxidase, a defensive enzyme that produces hypochlorous acid (HOCl), to kill pathogens, but also readily damages host proteins. In this study, we examined the reactivity of histones with and without HOCl modification, with a pancreatic β-cell model. Exposure of β-cells to histones resulted in a loss of metabolic activity and cell death by a combination of apoptosis and necrosis. This toxicity was increased on pretreatment of the β-cells with tumour necrosis factor α and interleukin 1β. Histones upregulated endoplasmic reticulum (ER) stress genes, including the pro-apoptotic transcription factor CHOP. There was also evidence for alterations to the cellular redox environment and upregulation of antioxidant gene expression. However, downregulation of insulin-associated genes and insulin was observed. Interestingly, modification of the histones with HOCl reduced their toxicity and altered the patterns of gene expression observed, and a further decrease in the expression of insulin-associated genes was observed. These findings could be relevant to the development of Type 2 diabetes, where low-grade inflammation favours NET release, resulting in elevated histones in the circulation.
Collapse
Affiliation(s)
- Clara Skjølberg
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Laura Degani
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Inga Sileikaite-Morvaközi
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, University of Copenhagen, Panum, Blegdamsvej 3B, Copenhagen N, DK-2200, Denmark.
| |
Collapse
|
3
|
Kwak H, Lee E, Karki R. DNA sensors in metabolic and cardiovascular diseases: Molecular mechanisms and therapeutic prospects. Immunol Rev 2025; 329:e13382. [PMID: 39158380 PMCID: PMC11744256 DOI: 10.1111/imr.13382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
DNA sensors generally initiate innate immune responses through the production of type I interferons. While extensively studied for host defense against invading pathogens, emerging evidence highlights the involvement of DNA sensors in metabolic and cardiovascular diseases. Elevated levels of modified, damaged, or ectopically localized self-DNA and non-self-DNA have been observed in patients and animal models with obesity, diabetes, fatty liver disease, and cardiovascular disease. The accumulation of cytosolic DNA aberrantly activates DNA signaling pathways, driving the pathological progression of these disorders. This review highlights the roles of specific DNA sensors, such as cyclic AMP-GMP synthase and stimulator of interferon genes (cGAS-STING), absent in melanoma 2 (AIM2), toll-like receptor 9 (TLR9), interferon gamma-inducible protein 16 (IFI16), DNA-dependent protein kinase (DNA-PK), and DEAD-box helicase 41 (DDX41) in various metabolic disorders. We explore how DNA signaling pathways in both immune and non-immune cells contribute to the development of these diseases. Furthermore, we discuss the intricate interplay between metabolic stress and immune responses, offering insights into potential therapeutic targets for managing metabolic and cardiovascular disorders. Understanding the mechanisms of DNA sensor signaling in these contexts provides a foundation for developing novel interventions aimed at mitigating the impact of these pervasive health issues.
Collapse
Affiliation(s)
- Hyosang Kwak
- Department of Biological Sciences, College of Natural ScienceSeoul National UniversitySeoulSouth Korea
| | - Ein Lee
- Department of Biomedical Sciences, College of MedicineSeoul National UniversitySeoulSouth Korea
| | - Rajendra Karki
- Department of Biological Sciences, College of Natural ScienceSeoul National UniversitySeoulSouth Korea
- Nexus Institute of Research and Innovation (NIRI)KathmanduNepal
| |
Collapse
|
4
|
Kong C, Guo Z, Liu F, Tang N, Wang M, Yang D, Li C, Yang Z, Ma Y, Wang P, Tang Q. Triad3A-Mediated K48-Linked ubiquitination and degradation of TLR9 impairs mitochondrial bioenergetics and exacerbates diabetic cardiomyopathy. J Adv Res 2024; 61:65-81. [PMID: 37625569 PMCID: PMC11258663 DOI: 10.1016/j.jare.2023.08.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023] Open
Abstract
INTRODUCTION Targeted protein degradation represents a promising therapeutic approach, while diabetic cardiomyopathy (DCM) arises as a consequence of aberrant insulin secretion and impaired glucose and lipid metabolism in the heart.. OBJECTIVES Considering that the Toll-like receptor 9 (TLR9) signaling pathway plays a pivotal role in regulating energy metabolism, safeguarding cardiomyocytes, and influencing glucose uptake, the primary objective of this study was to investigate the impact of TLR9 on diabetic cardiomyopathy (DCM) and elucidate its underlying mechanism. METHODS Mouse model of DCM was established using intraperitoneal injection of STZ, and mice were transfected with adeno-associated virus serotype 9-TLR9 (AAV9-TLR9) to assess the role of TLR9 in DCM. To explore the mechanism of TLR9 in regulating DCM disease progression, we conducted interactome analysis and employed multiple molecular approaches. RESULTS Our study revealed a significant correlation between TLR9 expression and mouse DCM. TLR9 overexpression markedly mitigated cardiac dysfunction, myocardial fibrosis, oxidative stress, and apoptosis in DCM, while inflammation levels remained relatively unaffected. Mechanistically, TLR9 overexpression positively modulated mitochondrial bioenergetics and activated the AMPK-PGC1a signaling pathway. Furthermore, we identified Triad3A as an interacting protein that facilitated TLR9's proteasomal degradation through K48-linked ubiquitination. Inhibiting Triad3A expression improved cardiac function and pathological changes in DCM by enhancing TLR9 activity. CONCLUSIONS The findings of this study highlight the critical role of TLR9 in maintaining cardiac function and mitigating pathological alterations in diabetic cardiomyopathy. Triad3A-mediated regulation of TLR9 expression and function has significant implications for understanding the pathogenesis of DCM. Targeting TLR9 and its interactions with Triad3A may hold promise for the development of novel therapeutic strategies for diabetic cardiomyopathy. Further research is warranted to fully explore the therapeutic potential of TLR9 modulation in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Fangyuan Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Nan Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Dan Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Chenfei Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zheng Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, PR China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
5
|
Yang X, Huang J, Peng J, Wang P, Wong FS, Wang R, Wang D, Wen L. Gut microbiota from B-cell-specific TLR9-deficient NOD mice promote IL-10 + Breg cells and protect against T1D. Front Immunol 2024; 15:1413177. [PMID: 38903498 PMCID: PMC11187306 DOI: 10.3389/fimmu.2024.1413177] [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: 04/06/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024] Open
Abstract
Introduction Type 1 diabetes (T1D) is an autoimmune disease characterized by the destruction of insulin-producing β cells. Toll-like receptor 9 (TLR9) plays a role in autoimmune diseases, and B cell-specific TLR9 deficiency delays T1D development. Gut microbiota are implicated in T1D, although the relationship is complex. However, the impact of B cell-specific deficiency of TLR9 on intestinal microbiota and the impact of altered intestinal microbiota on the development of T1D are unclear. Objectives This study investigated how gut microbiota and the intestinal barrier contribute to T1D development in B cell-specific TLR9-deficient NOD mice. Additionally, this study explored the role of microbiota in immune regulation and T1D onset. Methods The study assessed gut permeability, gene expression related to gut barrier integrity, and gut microbiota composition. Antibiotics depleted gut microbiota, and fecal samples were transferred to germ-free mice. The study also examined IL-10 production, Breg cell differentiation, and their impact on T1D development. Results B cell-specific TLR9-deficient NOD mice exhibited increased gut permeability and downregulated gut barrier-related gene expression. Antibiotics restored gut permeability, suggesting microbiota influence. Altered microbiota were enriched in Lachnospiraceae, known for mucin degradation. Transferring this microbiota to germ-free mice increased gut permeability and promoted IL-10-expressing Breg cells. Rag-/- mice transplanted with fecal samples from Tlr9 fl/fl Cd19-Cre+ mice showed delayed diabetes onset, indicating microbiota's impact. Conclusion B cell-specific TLR9 deficiency alters gut microbiota, increasing gut permeability and promoting IL-10-expressing Breg cells, which delay T1D. This study uncovers a link between TLR9, gut microbiota, and immune regulation in T1D, with implications for microbiota-targeted T1D therapies.
Collapse
Affiliation(s)
- Xin Yang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Juan Huang
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Jian Peng
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Pai Wang
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - F. Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Ruirui Wang
- Shanghai Innovation Center of Traditional Chinese Medicine (TCM) Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dapeng Wang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Li Wen
- Section of Endocrinology, Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
6
|
Wang P, Yang X, Zhang L, Sha S, Huang J, Peng J, Gu J, Pearson JA, Hu Y, Zhao H, Wong FS, Wang Q, Wen L. Tlr9 deficiency in B cells leads to obesity by promoting inflammation and gut dysbiosis. Nat Commun 2024; 15:4232. [PMID: 38762479 PMCID: PMC11102548 DOI: 10.1038/s41467-024-48611-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 05/02/2024] [Indexed: 05/20/2024] Open
Abstract
Toll-like receptor 9 (TLR9) recognizes bacterial, viral and self DNA and play an important role in immunity and inflammation. However, the role of TLR9 in obesity is less well-studied. Here, we generate B-cell-specific Tlr9-deficient (Tlr9fl/fl/Cd19Cre+/-, KO) B6 mice and model obesity using a high-fat diet. Compared with control mice, B-cell-specific-Tlr9-deficient mice exhibited increased fat tissue inflammation, weight gain, and impaired glucose and insulin tolerance. Furthermore, the frequencies of IL-10-producing-B cells and marginal zone B cells were reduced, and those of follicular and germinal center B cells were increased. This was associated with increased frequencies of IFNγ-producing-T cells and increased follicular helper cells. In addition, gut microbiota from the KO mice induced a pro-inflammatory state leading to immunological and metabolic dysregulation when transferred to germ-free mice. Using 16 S rRNA gene sequencing, we identify altered gut microbial communities including reduced Lachnospiraceae, which may play a role in altered metabolism in KO mice. We identify an important network involving Tlr9, Irf4 and Il-10 interconnecting metabolic homeostasis, with the function of B and T cells, and gut microbiota in obesity.
Collapse
Affiliation(s)
- Pai Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Xin Yang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Luyao Zhang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Sha Sha
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Juan Huang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Jian Peng
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Jianlei Gu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - James Alexander Pearson
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| | - Youjia Hu
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine and Systems Immunity University Research Institute, Cardiff University, Cardiff, UK
| | - Quan Wang
- Department of Gastrocolorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
7
|
Wang K, Huang H, Zhan Q, Ding H, Li Y. Toll-like receptors in health and disease. MedComm (Beijing) 2024; 5:e549. [PMID: 38685971 PMCID: PMC11057423 DOI: 10.1002/mco2.549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Toll-like receptors (TLRs) are inflammatory triggers and belong to a family of pattern recognition receptors (PRRs) that are central to the regulation of host protective adaptive immune responses. Activation of TLRs in innate immune myeloid cells directs lymphocytes to produce the most appropriate effector responses to eliminate infection and maintain homeostasis of the body's internal environment. Inappropriate TLR stimulation can lead to the development of general autoimmune diseases as well as chronic and acute inflammation, and even cancer. Therefore, TLRs are expected to be targets for therapeutic treatment of inflammation-related diseases, autoimmune diseases, microbial infections, and human cancers. This review summarizes the recent discoveries in the molecular and structural biology of TLRs. The role of different TLR signaling pathways in inflammatory diseases, autoimmune diseases such as diabetes, cardiovascular diseases, respiratory diseases, digestive diseases, and even cancers (oral, gastric, breast, colorectal) is highlighted and summarizes new drugs and related clinical treatments in clinical trials, providing an overview of the potential and prospects of TLRs for the treatment of TLR-related diseases.
Collapse
Affiliation(s)
- Kunyu Wang
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Hanyao Huang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| | - Qi Zhan
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Haoran Ding
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yi Li
- Department of Head and Neck Oncology Surgery, State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
8
|
Alkudmani ZS, Alzailai AA, Aburisheh KH, Alshammary AF, Ali Khan I. Toll-like Receptor 9 Gene in the Development of Type 2 Diabetes Mellitus in the Saudi Arabian Population. BIOLOGY 2023; 12:1439. [PMID: 37998038 PMCID: PMC10669332 DOI: 10.3390/biology12111439] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/10/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Diabetes mellitus is a complex disease with a wide range of manifestations. Diabetes, notably type 2 diabetes mellitus (T2DM), is becoming more common in Saudi Arabia as a result of obesity and an aging population. T2DM is classified as a noncommunicable disease, and its incidence in the Saudi population continues to grow as a consequence of socioeconomic changes. Toll-like receptors (TLRs) are innate immune receptors that mediate the inflammatory response in diabetes mellitus. Previous studies have documented the relationship between different SNPs in the TLR9 gene in different forms of diabetes. As a result, the purpose of this study was to investigate the relationship between rs187084, rs352140, and rs5743836 SNPs in the TLR9 gene among T2DM patients in the Saudi population. This was a case-control study that included 100 T2DM cases and 100 control subjects. The three SNPs were identified in the study population (n = 200) using polymerase chain reaction (PCR), restriction enzymes for rs352140, and Sanger sequencing for rs187084 and rs5783836. Next, statistical analyses were performed using various software to determine the association between the SNPs and T2DM. rs187084 and rs5743836 were associated with an increased risk of T2DM development. rs187084 and rs5743836 allelic frequencies were associated with a 3.2 times increased risk of T2DM development (p < 0.05). DBP was associated with T2DM (p = 0.02). rs187084 was associated with TC and HDLc; rs352140 was associated with DBP, HbA1c, and HDLc; rs5743836 was associated with waist (p < 0.05). The CGT haplotype was strongly associated with T2DM (p < 0.003). Gene-gene interaction, graphical presentation, and dendrogram showed the strong association with T2DM patients (p < 0.05). This study concluded that rs187084 and rs5743836 were strongly associated with T2DM in Saudi Arabian patients. This study provides further evidence that SNPs in the TLR9 gene play a significant role in T2DM development in a Saudi community.
Collapse
Affiliation(s)
- Zeina S. Alkudmani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (Z.S.A.); (A.A.A.); (A.F.A.)
| | - Aminah Ahmad Alzailai
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (Z.S.A.); (A.A.A.); (A.F.A.)
| | - Khaled H. Aburisheh
- University Diabetes Center, King Saud University Medical City, King Saud University, Riyadh 11472, Saudi Arabia;
| | - Amal F. Alshammary
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (Z.S.A.); (A.A.A.); (A.F.A.)
| | - Imran Ali Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (Z.S.A.); (A.A.A.); (A.F.A.)
| |
Collapse
|
9
|
Wang Y, Xia Y, Chen Y, Xu L, Sun X, Li J, Huang G, Li X, Xie Z, Zhou Z. Association analysis between the TLR9 gene polymorphism rs352140 and type 1 diabetes. Front Endocrinol (Lausanne) 2023; 14:1030736. [PMID: 37139337 PMCID: PMC10150994 DOI: 10.3389/fendo.2023.1030736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/24/2023] [Indexed: 05/05/2023] Open
Abstract
Background To a great extent, genetic factors contribute to the susceptibility to type 1 diabetes (T1D) development, and by triggering immune imbalance, Toll-like receptor (TLR) 9 is involved in the development of T1D. However, there is a lack of evidence supporting a genetic association between polymorphisms in the TLR9 gene and T1D. Methods In total, 1513 individuals, including T1D patients (n=738) and healthy control individuals (n=775), from the Han Chinese population were recruited for an association analysis of the rs352140 polymorphism of the TLR9 gene and T1D. rs352140 was genotyped by MassARRAY. The allele and genotype distributions of rs352140 in the T1D and healthy groups and those in different T1D subgroups were analyzed by the chi-squared test and binary logistic regression model. The chi-square test and Kruskal-Wallis H test were performed to explore the association between genotype and phenotype in T1D patients. Results The allele and genotype distributions of rs352140 were significantly different in T1D patients and healthy control individuals (p=0.019, p=0.035). Specifically, the T allele and TT genotype of rs352140 conferred a higher risk of T1D (OR=1.194, 95% CI=1.029-1.385, p=0.019, OR=1.535, 95% CI=1.108-2.126, p=0.010). The allele and genotype distributions of rs352140 were not significantly different between childhood-onset and adult-onset T1D and between T1D with a single islet autoantibody and T1D with multiple islet autoantibodies (p=0.603, p=0.743). rs352140 was associated with T1D susceptibility according to the recessive and additive models (p=0.015, p=0.019) but was not associated with T1D susceptibility in the dominant and overdominant models (p=0.117, p=0.928). Moreover, genotype-phenotype association analysis showed that the TT genotype of rs352140 was associated with higher fasting C-peptide levels (p=0.017). Conclusion In the Han Chinese population, the TLR9 polymorphism rs352140 is associated with T1D and is a risk factor for susceptibility to T1D.
Collapse
|
10
|
Niemann B, Puleo A, Stout C, Markel J, Boone BA. Biologic Functions of Hydroxychloroquine in Disease: From COVID-19 to Cancer. Pharmaceutics 2022; 14:pharmaceutics14122551. [PMID: 36559044 PMCID: PMC9787624 DOI: 10.3390/pharmaceutics14122551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
Abstract
Chloroquine (CQ) and Hydroxychloroquine (HCQ), initially utilized in the treatment of malaria, have now developed a long list of applications. Despite their clinical relevance, their mechanisms of action are not clearly defined. Major pathways by which these agents are proposed to function include alkalinization of lysosomes and endosomes, downregulation of C-X-C chemokine receptor type 4 (CXCR4) expression, high-mobility group box 1 protein (HMGB1) inhibition, alteration of intracellular calcium, and prevention of thrombus formation. However, there is conflicting data present in the literature. This is likely the result of the complex overlapping pathways between these mechanisms of action that have not previously been highlighted. In fact, prior research has focused on very specific portions of particular pathways without describing these in the context of the extensive CQ/HCQ literature. This review summarizes the detailed data regarding CQ/HCQ's mechanisms of action while also providing insight into the overarching themes. Furthermore, this review provides clinical context to the application of these diverse drugs including their role in malaria, autoimmune disorders, cardiovascular disease, thrombus formation, malignancies, and viral infections.
Collapse
Affiliation(s)
- Britney Niemann
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Correspondence: ; Tel.: +1-304-293-1254
| | - Amanda Puleo
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Conley Stout
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Justin Markel
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
| | - Brian A. Boone
- Department of Surgery, West Virginia University, Morgantown, WV 26506, USA
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV 26506, USA
| |
Collapse
|
11
|
White MG, Wargo JA. The Microbiome in Gastrointestinal Cancers. Gastroenterol Clin North Am 2022; 51:667-680. [PMID: 36153116 PMCID: PMC11833749 DOI: 10.1016/j.gtc.2022.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The human microbiome has been recognized as increasingly important to health and disease. This is especially prescient in the development of various cancers, their progression, and the microbiome's modulation of various anticancer therapeutics. Mechanisms behind these interactions have been increasingly well described through modulation of the host immune system as well as induction of genetic changes and local inactivation of cancer therapeutics. Here, we review these associations for a variety of gastrointestinal malignancies as well as contemporary strategies proposed to leverage these associations to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Michael G White
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 1484, Houston, TX 77030, USA.
| |
Collapse
|
12
|
Saber MM, Monir N, Awad AS, Elsherbiny ME, Zaki HF. TLR9: A friend or a foe. Life Sci 2022; 307:120874. [PMID: 35963302 DOI: 10.1016/j.lfs.2022.120874] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
The innate immune system is a primary protective line in our body. It confers its protection through different pattern recognition receptors (PRRs), especially toll like receptors (TLRs). Toll like receptor 9 (TLR9) is an intracellular TLR, expressed in different immunological and non-immunological cells. Release of cellular components, such as proteins, nucleotides, and DNA confers a beneficial inflammatory response and maintains homeostasis for removing cellular debris during normal physiological conditions. However, during pathological cellular damage and stress signals, engagement between mtDNA and TLR9 acts as an alarm for starting inflammatory and autoimmune disorders. The controversial role of TLR9 in different diseases baffled scientists if it has a protective or deleterious effect after activation during insults. Targeting the immune system, especially the TLR9 needs further investigation to provide a therapeutic strategy to control inflammation and autoimmune disorders.
Collapse
Affiliation(s)
- Mona M Saber
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt.
| | - Nada Monir
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Azza S Awad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Marwa E Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, Giza, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Giza, Egypt
| |
Collapse
|
13
|
The Yin and Yang of toll-like receptors in endothelial dysfunction. Int Immunopharmacol 2022; 108:108768. [DOI: 10.1016/j.intimp.2022.108768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
|
14
|
Guo K, Huang J, Zhou Z. Host gene effects on gut microbiota in type 1 diabetes. Biochem Soc Trans 2022; 50:1133-1142. [PMID: 35521897 PMCID: PMC9246325 DOI: 10.1042/bst20220004] [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: 01/11/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/03/2022]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by progressive pancreatic β-cell loss. Both a predisposing genetic background, that may encompass mutations in several genes, as well as exposure to environmental factors can affect the progression of autoimmune responses to multiple pancreatic islet autoantigens. Many genetic variants that increase the risk of T1D are found in immunity genes involved in sensing and responding to microorganisms. Although increasing evidence indicates that the gut microbiome composition may promote or prevent T1D development, little is known about the link between gut microbiota and T1D susceptibility genes in patients with T1D. Recent studies in the inbred non-obese diabetic (NOD) mouse, a widely used model of T1D, have suggested that many genetic loci can influence gut microbiome composition to modulate islet autoimmunity. This review summarizes evidence that examines the effect of host genes on gut microbiota diversity and function during T1D development. Knowledge of the host gene-gut microbiota interactions at play during T1D progression may help us identify new diagnostic and prognostic tools and help also design effective strategies for disease treatment.
Collapse
Affiliation(s)
- Keyu Guo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Juan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, U.S.A
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
15
|
Wujcicka WI, Kacerovsky M, Krygier A, Krekora M, Kaczmarek P, Grzesiak M. Association of Single Nucleotide Polymorphisms from Angiogenesis-Related Genes, ANGPT2, TLR2 and TLR9, with Spontaneous Preterm Labor. Curr Issues Mol Biol 2022; 44:2939-2955. [PMID: 35877427 PMCID: PMC9322696 DOI: 10.3390/cimb44070203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 12/07/2022] Open
Abstract
In this study, we hypothesized that the changes localized at angiopoietin-2 (ANGPT2), granulocyte-macrophage colony-stimulating factor (CSF2), fms-related tyrosine kinase 1 (FLT1) and toll-like receptor (TLR) 2, TLR6 and TLR9 genes were associated with spontaneous preterm labor (PTL), as well as with possible genetic alterations on PTL-related coagulation. This case-control genetic association study aimed to identify single nucleotide polymorphisms (SNPs) for the aforementioned genes, which are correlated with genetic risk or protection against PTL in Polish women. The study was conducted in 320 patients treated between 2016 and 2020, including 160 women with PTL and 160 term controls in labor. We found that ANGPT2 rs3020221 AA homozygotes were significantly less common in PTL cases than in controls, especially after adjusting for activated partial thromboplastin time (APTT) and platelet (PLT) parameters. TC heterozygotes for TLR2 rs3804099 were associated with PTL after correcting for anemia, vaginal bleeding, and history of threatened miscarriage or PTL. TC and CC genotypes in TLR9 rs187084 were significantly less common in women with PTL, compared to the controls, after adjusting for bleeding and gestational diabetes. For the first time, it was shown that three polymorphisms-ANGPT2 rs3020221, TLR2 rs3804099 and TLR9 rs187084 -were significantly associated with PTL, adjusted by pregnancy development influencing factors.
Collapse
Affiliation(s)
- Wioletta Izabela Wujcicka
- Scientific Laboratory of the Center of Medical Laboratory Diagnostics and Screening, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
- Correspondence: or ; Tel.: +48-42-271-15-20; Fax: +48-42-271-15-10
| | - Marian Kacerovsky
- Department of Obstetrics and Gynecology, University Hospital Hradec Kralove, Charles University, 500 03 Hradec Kralove, Czech Republic;
- Biomedical Research Center, University Hospital Hradec Kralove, 500 03 Hradec Kralove, Czech Republic
| | - Adrian Krygier
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Michał Krekora
- Department of Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland;
| | - Piotr Kaczmarek
- Department of Gynecology, Reproduction and Fetal Therapy, and Diagnostics and Treatment of Infertility, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland;
| | - Mariusz Grzesiak
- Department of Gynecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland;
- Department of Perinatology, Obstetrics and Gynecology, Polish Mother’s Memorial Hospital-Research Institute, 93-338 Lodz, Poland
| |
Collapse
|
16
|
Ding P, Tan Q, Wei Z, Chen Q, Wang C, Qi L, Wen L, Zhang C, Yao C. Toll-like receptor 9 deficiency induces osteoclastic bone loss via gut microbiota-associated systemic chronic inflammation. Bone Res 2022; 10:42. [PMID: 35624094 PMCID: PMC9142495 DOI: 10.1038/s41413-022-00210-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 02/06/2022] [Accepted: 03/13/2022] [Indexed: 02/08/2023] Open
Abstract
Toll-like receptors (TLRs) play pivotal roles in inflammation and provide important links between the immune and skeletal systems. Although the activation of TLRs may affect osteoclast differentiation and bone metabolism, whether and how TLRs are required for normal bone remodeling remains to be fully explored. In the current study, we show for the first time that TLR9-/- mice exhibit a low bone mass and low-grade systemic chronic inflammation, which is characterized by the expansion of CD4+ T cells and increased levels of inflammatory cytokines, including TNFα, RANKL, and IL1β. The increased levels of these cytokines significantly promote osteoclastogenesis and induce bone loss. Importantly, TLR9 deletion alters the gut microbiota, and this dysbiosis is the basis of the systemic inflammation and bone loss observed in TLR9-/- mice. Furthermore, through single-cell RNA sequencing, we identified myeloid-biased hematopoiesis in the bone marrow of TLR9-/- mice and determined that the increase in myelopoiesis, likely caused by the adaptation of hematopoietic stem cells to systemic inflammation, also contributes to inflammation-induced osteoclastogenesis and subsequent bone loss in TLR9-/- mice. Thus, our study provides novel evidence that TLR9 signaling connects the gut microbiota, immune system, and bone and is critical in maintaining the homeostasis of inflammation, hematopoiesis, and bone metabolism under normal conditions.
Collapse
Affiliation(s)
- Peng Ding
- Department of Orthopedic Surgery, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China
| | - Qiyuan Tan
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China
| | - Zhanying Wei
- Department of Osteoporosis and Skeletal Disorders, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China
| | - Qiyu Chen
- Department of Orthopedic Surgery, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China
| | - Chun Wang
- Department of Osteoporosis and Skeletal Disorders, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China
| | - Luyue Qi
- Department of Endocrinology and Metabolism, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA
| | - Changqing Zhang
- Department of Orthopedic Surgery, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China.
| | - Chen Yao
- Department of Orthopedic Surgery, Shanghai Jiaotong University affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
17
|
Han J, Zheng Q, Cheng Y, Liu Y, Bai Y, Yan B, Guo S, Yu J, Li X, Wang C. Toll-like receptor 9 (TLR9) gene deletion-mediated fracture healing in type II diabetic osteoporosis associates with inhibition of the nuclear factor-kappa B (NF-κB) signaling pathway. Bioengineered 2022; 13:13689-13702. [PMID: 35707851 PMCID: PMC9275877 DOI: 10.1080/21655979.2022.2063663] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Diabetes is characterized by increased fracture risk. Evidence from in vivo studies is lacking for anti-fracture strategies in diabetes. Our microarray analyses predicted association of Toll-like receptor 9 (TLR9) with both diabetes and osteoporosis, which was the focus of this work in a murine model of type II diabetic osteoporosis (T2DOP). A T2DOP model with fracture was established in TLR9 knockout (TLR9−/−) mice, which were then treated with the NF-κB signaling pathway inhibitor (PDTC) and activator (TNF-α). The obtained data suggested that TLR9 knockout augmented regeneration of bone tissues and cartilage area in the callus, and diminished fibrous tissues in T2DOP mice. Moreover, TLR9 depletion significantly affected bone mineral density (BMD), bone volume/tissue volume (BV/TV), connectivity density, trabecular number, trabecular separation and trabecular thickness, thus promoting fracture recovery. Bone morphology and structure were also improved in response to TLR9 depletion in T2DOP mice. TLR9 depletion inactivated NF-κB signaling in T2DOP mice. PDTC was found to enhance fracture healing in T2DOP mice, while TNF-α negated this effect. Collectively, these data indicate that TLR9 depletion may hold anti-fracture properties, making it a potential therapeutic target for T2DOP. Abbreviations: Diabetic osteoporosis (DOP); bone mineral density (BMD); Toll-like receptors (TLRs); type 2 diabetes (T2D); Toll-like receptor 9 (TLR9); nuclear factor-kappaB (NF-κB); streptozotocin (STZ); type 2 diabetic osteoporosis (T2DOP); Gene Expression Omnibus (GEO); Kyoto encyclopedia of genes and genomes (KEGG); pyrrolidine dithiocarbamate (PDTC); computed tomography (CT); Hematoxylin–eosin (HE); bone morphogenetic protein 7 (BMP7); analysis of variance (ANOVA);
Collapse
Affiliation(s)
- Jiakai Han
- Endocrinology Department, Huaihe Hospital of Henan University, Kaifeng, PR, China
| | - Qian Zheng
- Endocrinology Department, Yan'an Hospital of Kunming Medical University, Kunming, PR, China
| | - Yongxia Cheng
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Yong Liu
- Platform Management Division, Scientific Research Division of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Yuxin Bai
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Bin Yan
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Sufen Guo
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Jianbo Yu
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Xinxin Li
- Ultrasound Department, Second Hospital of Mudanjiang Medical College, Mudanjiang, PR, China
| | - Chong Wang
- Pathology Diagnosis Center, The HongQi Hospital, The First Clinical Medical School of Mudanjiang Medical College, Mudanjiang, PR, China
| |
Collapse
|
18
|
Iskender H, Dokumacioglu E, Terim Kapakin KA, Yenice G, Mohtare B, Bolat I, Hayirli A. Effects of oleanolic acid on inflammation and metabolism in diabetic rats. Biotech Histochem 2022; 97:269-276. [PMID: 34261397 DOI: 10.1080/10520295.2021.1954691] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease that threatens the health of the world population. We investigated the effects of oleanolic acid (OA) administration on inflammation status and metabolic profile in streptozotocin (STZ) induced diabetic rats. Four experimental groups were established: healthy rats not administered OA, healthy rats administered OA, diabetic rats not administered OA, diabetic rats administered OA. OA, 5 mg/kg, was administered by oral gavage for 21 days. Serum samples collected at the end of the experiment and analyzed for toll-like receptor-9, interleukin-18, nuclear factor kappa B, malondialdehyde MDA, glucose, total cholesterol, triglycerides, high-density lipoprotein, low-density lipoprotein, calcium, phosphorus, magnesium and potassium. Pancreas tissue was examined for pathology. Induction of DM caused increased serum concentrations of inflammation and oxidative damage markers. DM also caused hyperglycemia-hyperlipidemia and decreased serum concentration of minerals. The islets of Langerhans were degenerated and necrotic. Administration of OA reversed the adverse effects of DM. OA treatment can ameliorate inflammation and oxidative damage due to DM by normalizing hyperglycemia and decreasing TLR-9, IL-18, NF-κB and MDA levels.
Collapse
Affiliation(s)
- Hatice Iskender
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Coruh University, Artvin, Turkey
| | - Eda Dokumacioglu
- Department of Nutrition and Dietetics, Faculty of Healthy Sciences, Artvin Coruh University, Artvin, Turkey
| | | | - Guler Yenice
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Behzat Mohtare
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Ismail Bolat
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Armagan Hayirli
- Department of Animal Nutrition and Nutritional Disorders, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| |
Collapse
|
19
|
Interleukin-35 Prevents Development of Autoimmune Diabetes Possibly by Maintaining the Phenotype of Regulatory B Cells. Int J Mol Sci 2021; 22:ijms222312988. [PMID: 34884797 PMCID: PMC8657454 DOI: 10.3390/ijms222312988] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/12/2023] Open
Abstract
The anti-inflammatory role of regulatory B cells (Breg cells) has been associated with IL-35 based on studies of experimental autoimmune uveitis and encephalitis. The role of Breg cells and IL-35+ Breg cells for type 1 diabetes (T1D) remains to be investigated. We studied PBMCs from T1D subjects and healthy controls (HC) and found lowered proportions of Breg cells and IL-35+ Breg cells in T1D. To elucidate the role of Breg cells, the lymphoid organs of two mouse models of T1D were examined. Lower proportions of Breg cells and IL-35+ Breg cells were found in the animal models of T1D compared with control mice. In addition, the systemic administration of recombinant mouse IL-35 prevented hyperglycemia after multiple low dose streptozotocin (MLDSTZ) injections and increased the proportions of Breg cells and IL-35+ Breg cells. A higher proportion of IFN-γ+ cells among Breg cells were found in the PBMCs of the T1D subjects. In the MLDSTZ mice, IL-35 administration decreased the proportions of IFN-γ+ cells among the Breg cells. Our data illustrate that Breg cells may play an important role in the development of T1D and that IL-35 treatment prevents the development of hyperglycemia by maintaining the phenotype of the Breg cells under an experimental T1D condition.
Collapse
|
20
|
Badal D, Sachdeva N, Maheshwari D, Basak P. Role of nucleic acid sensing in the pathogenesis of type 1 diabetes. World J Diabetes 2021; 12:1655-1673. [PMID: 34754369 PMCID: PMC8554372 DOI: 10.4239/wjd.v12.i10.1655] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/22/2021] [Accepted: 07/05/2021] [Indexed: 02/06/2023] Open
Abstract
During infections, nucleic acids of pathogens are also engaged in recognition via several exogenous and cytosolic pattern recognition receptors, such as the toll-like receptors, retinoic acid inducible gene-I-like receptors, and nucleotide-binding and oligomerization domain-like receptors. The binding of the pathogen-derived nucleic acids to their corresponding sensors initiates certain downstream signaling cascades culminating in the release of type-I interferons (IFNs), especially IFN-α and other cytokines to induce proinflammatory responses towards invading pathogens leading to their clearance from the host. Although these sensors are hardwired to recognize pathogen associated molecular patterns, like viral and bacterial nucleic acids, under unusual physiological conditions, such as excessive cellular stress and increased apoptosis, endogenous self-nucleic acids like DNA, RNA, and mitochondrial DNA are also released. The presence of these self-nucleic acids in extranuclear compartments or extracellular spaces or their association with certain proteins sometimes leads to the failure of discriminating mechanisms of nucleic acid sensors leading to proinflammatory responses as seen in autoimmune disorders, like systemic lupus erythematosus, psoriasis and to some extent in type 1 diabetes (T1D). This review discusses the involvement of various nucleic acid sensors in autoimmunity and discusses how aberrant recognition of self-nucleic acids by their sensors activates the innate immune responses during the pathogenesis of T1D.
Collapse
Affiliation(s)
- Darshan Badal
- Department of Pediatrics, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Deep Maheshwari
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Preetam Basak
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
21
|
Ding F, Luo X, Tu Y, Duan X, Liu J, Jia L, Zheng P. Alpk1 Sensitizes Pancreatic Beta Cells to Cytokine-Induced Apoptosis via Upregulating TNF-α Signaling Pathway. Front Immunol 2021; 12:705751. [PMID: 34621265 PMCID: PMC8490819 DOI: 10.3389/fimmu.2021.705751] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 09/06/2021] [Indexed: 11/13/2022] Open
Abstract
Pancreatic beta cell failure is the hallmark of type 1 diabetes (T1D). Recent studies have suggested that pathogen recognizing receptors (PRRs) are involved in the survival, proliferation and function of pancreatic beta cells. So far, little is known about the role of alpha-protein kinase 1 (ALPK1), a newly identified cytosolic PRR specific for ADP-β-D-manno-heptose (ADP-heptose), in beta cell survival. In current study we aimed to fill the knowledge gap by investigating the role of Alpk1 in the apoptosis of MIN6 cells, a murine pancreatic beta cell line. We found that the expression of Alpk1 was significantly elevated in MIN6 cells exposed to pro-inflammatory cytokines, but not to streptozotocin, low-dose or high-dose glucose. Activation of Alpk1 by ADP heptose alone was insufficient to induce beta cell apoptosis. However, it significantly exacerbated cytokine-induced apoptosis in MIN6 cells. Mechanistic investigations showed that Alpk1 activation was potent to further induce the expression of tumor necrosis factor (TNF)-α and Fas after cytokine stimulation, possibly due to enhanced activation of the TIFA/TAK1/NF-κB signaling axis. Treatment of GLP-1 receptor agonist decreased the expression of TNF-α and Fas and improved the survival of beta cells exposed to pro-inflammatory cytokines and ADP heptose. In summary, our data suggest that Alpk1 sensitizes beta cells to cytokine-induced apoptosis by potentiating TNF-α signaling pathway, which may provide novel insight into beta cell failure and T1D development.
Collapse
Affiliation(s)
- Fei Ding
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Xi Luo
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yiting Tu
- Department of Neurology, Shenzhen Samii International Medical Center (The Fourth People's Hospital of Shenzhen), Shenzhen, China
| | - Xianlan Duan
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jia Liu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijing Jia
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Peilin Zheng
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
22
|
Antidiabetic effects of hydroxychloroquine in two Japanese patients with systemic lupus erythematosus. Diabetol Int 2021; 13:447-451. [DOI: 10.1007/s13340-021-00544-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 09/12/2021] [Indexed: 10/20/2022]
|
23
|
Li D, Wu M. Pattern recognition receptors in health and diseases. Signal Transduct Target Ther 2021; 6:291. [PMID: 34344870 PMCID: PMC8333067 DOI: 10.1038/s41392-021-00687-0] [Citation(s) in RCA: 842] [Impact Index Per Article: 210.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/23/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
Pattern recognition receptors (PRRs) are a class of receptors that can directly recognize the specific molecular structures on the surface of pathogens, apoptotic host cells, and damaged senescent cells. PRRs bridge nonspecific immunity and specific immunity. Through the recognition and binding of ligands, PRRs can produce nonspecific anti-infection, antitumor, and other immunoprotective effects. Most PRRs in the innate immune system of vertebrates can be classified into the following five types based on protein domain homology: Toll-like receptors (TLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), C-type lectin receptors (CLRs), and absent in melanoma-2 (AIM2)-like receptors (ALRs). PRRs are basically composed of ligand recognition domains, intermediate domains, and effector domains. PRRs recognize and bind their respective ligands and recruit adaptor molecules with the same structure through their effector domains, initiating downstream signaling pathways to exert effects. In recent years, the increased researches on the recognition and binding of PRRs and their ligands have greatly promoted the understanding of different PRRs signaling pathways and provided ideas for the treatment of immune-related diseases and even tumors. This review describes in detail the history, the structural characteristics, ligand recognition mechanism, the signaling pathway, the related disease, new drugs in clinical trials and clinical therapy of different types of PRRs, and discusses the significance of the research on pattern recognition mechanism for the treatment of PRR-related diseases.
Collapse
Affiliation(s)
- Danyang Li
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Provincial Tumor Hospital and the Affiliated Tumor Hospital of Xiangya Medical School, Central South University, Changsha, Hunan, China.
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China.
| |
Collapse
|
24
|
Innate immune receptors in type 1 diabetes: the relationship to cell death-associated inflammation. Biochem Soc Trans 2021; 48:1213-1225. [PMID: 32510139 DOI: 10.1042/bst20200131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/17/2022]
Abstract
The importance of innate immunity in host defense and inflammatory responses has been clearly demonstrated after the discovery of innate immune receptors such as Toll-like receptors (TLRs) or Nucleotide-binding oligomerization domain-containing protein (Nod)-like receptors (NLRs). Innate immunity also plays a critical role in diverse pathological conditions including autoimmune diseases such as type 1 diabetes (T1D). In particular, the role of a variety of innate immune receptors in T1D has been demonstrated using mice with targeted disruption of such innate immune receptors. Here, we discuss recent findings showing the role of innate immunity in T1D that were obtained mostly from studies of genetic mouse models of innate immune receptors. In addition, the role of innate immune receptors involved in the pathogenesis of T1D in sensing death-associated molecular patterns (DAMPs) released from dead cells or pathogen-associated molecular patterns (PAMPs) will also be covered. Elucidation of the role of innate immune receptors in T1D and the nature of DAMPs sensed by such receptors may lead to the development of new therapeutic modalities against T1D.
Collapse
|
25
|
Sha S, Pearson JA, Peng J, Hu Y, Huang J, Xing Y, Zhang L, Zhu Y, Zhao H, Wong FS, Chen L, Wen L. TLR9 Deficiency in B Cells Promotes Immune Tolerance via Interleukin-10 in a Type 1 Diabetes Mouse Model. Diabetes 2021; 70:504-515. [PMID: 33154070 PMCID: PMC7881860 DOI: 10.2337/db20-0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 11/01/2020] [Indexed: 12/22/2022]
Abstract
Toll-like receptor 9 (TLR9) is highly expressed in B cells, and B cells are important in the pathogenesis of type 1 diabetes (T1D) development. However, the intrinsic effect of TLR9 in B cells on β-cell autoimmunity is not known. To fill this knowledge gap, we generated NOD mice with a B-cell-specific deficiency of TLR9 (TLR9fl/fl/CD19-Cre+ NOD). The B-cell-specific deletion of TLR9 resulted in near-complete protection from T1D development. Diabetes protection was accompanied by an increased proportion of interleukin-10 (IL-10)-producing B cells. We also found that TLR9-deficient B cells were hyporesponsive to both innate and adaptive immune stimuli. This suggested that TLR9 in B cells modulates T1D susceptibility in NOD mice by changing the frequency and function of IL-10-producing B cells. Molecular analysis revealed a network of TLR9 with matrix metalloproteinases, tissue inhibitor of metalloproteinase-1, and CD40, all of which are interconnected with IL-10. Our study has highlighted an important connection of an innate immune molecule in B cells to the immunopathogenesis of T1D. Thus, targeting the TLR9 pathway, specifically in B cells, may provide a novel therapeutic strategy for T1D treatment.
Collapse
Affiliation(s)
- Sha Sha
- Department of Nephrology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Jian Peng
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Youjia Hu
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Juan Huang
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| | - Yanpeng Xing
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Luyao Zhang
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, Jilin, China
| | - Ying Zhu
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT
| | - Hongyu Zhao
- Department of Biostatistics, School of Public Health, Yale University, New Haven, CT
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, U.K
| | - Li Chen
- Institute of Endocrine and Metabolic Diseases of Shandong University, Jinan, Shandong, China
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT
| |
Collapse
|
26
|
Huang J, Peng J, Pearson JA, Efthimiou G, Hu Y, Tai N, Xing Y, Zhang L, Gu J, Jiang J, Zhao H, Zhou Z, Wong FS, Wen L. Toll-like receptor 7 deficiency suppresses type 1 diabetes development by modulating B-cell differentiation and function. Cell Mol Immunol 2021; 18:328-338. [PMID: 33432061 PMCID: PMC8027372 DOI: 10.1038/s41423-020-00590-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/31/2020] [Indexed: 01/29/2023] Open
Abstract
Innate immunity mediated by Toll-like receptors (TLRs), which can recognize pathogen molecular patterns, plays a critical role in type 1 diabetes development. TLR7 is a pattern recognition receptor that senses single-stranded RNAs from viruses and host tissue cells; however, its role in type 1 diabetes development remains unclear. In our study, we discovered that Tlr7-deficient (Tlr7-/-) nonobese diabetic (NOD) mice, a model of human type 1 diabetes, exhibited a significantly delayed onset and reduced incidence of type 1 diabetes compared with Tlr7-sufficient (Tlr7+/+) NOD mice. Mechanistic investigations showed that Tlr7 deficiency significantly altered B-cell differentiation and immunoglobulin production. Moreover, Tlr7-/- NOD B cells were found to suppress diabetogenic CD4+ T-cell responses and protect immunodeficient NOD mice from developing diabetes induced by diabetogenic T cells. In addition, we found that Tlr7 deficiency suppressed the antigen-presenting functions of B cells and inhibited cytotoxic CD8+ T-cell activation by downregulating the expression of both nonclassical and classical MHC class I (MHC-I) molecules on B cells. Our data suggest that TLR7 contributes to type 1 diabetes development by regulating B-cell functions and subsequent interactions with T cells. Therefore, therapeutically targeting TLR7 may prove beneficial for disease protection.
Collapse
Affiliation(s)
- Juan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Jian Peng
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - James Alexander Pearson
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Georgios Efthimiou
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Microbiology, University of Hull, Hull, UK
| | - Youjia Hu
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Yanpeng Xing
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Gastrointestinal Surgery of the First Hospital of Jilin University, Changchun, Jilin, China
| | - Luyao Zhang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
- Department of Gastrointestinal Surgery of the First Hospital of Jilin University, Changchun, Jilin, China
| | - Jianlei Gu
- Department of Biostatistics, Data Science & Genetics, Yale School of Public Health, New Haven, CT, USA
| | - Jianping Jiang
- Department of Biostatistics, Data Science & Genetics, Yale School of Public Health, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Data Science & Genetics, Yale School of Public Health, New Haven, CT, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - F Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
27
|
Aneman I, Pienaar D, Suvakov S, Simic TP, Garovic VD, McClements L. Mechanisms of Key Innate Immune Cells in Early- and Late-Onset Preeclampsia. Front Immunol 2020; 11:1864. [PMID: 33013837 PMCID: PMC7462000 DOI: 10.3389/fimmu.2020.01864] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/10/2020] [Indexed: 12/23/2022] Open
Abstract
Preeclampsia is a complex cardiovascular disorder of pregnancy with underlying multifactorial pathogeneses; however, its etiology is not fully understood. It is characterized by the new onset of maternal hypertension after 20 weeks of gestation, accompanied by proteinuria, maternal organ damage, and/or uteroplacental dysfunction. Preeclampsia can be subdivided into early- and late-onset phenotypes (EOPE and LOPE), diagnosed before 34 weeks or from 34 weeks of gestation, respectively. Impaired placental development in early pregnancy and subsequent growth restriction is often associated with EOPE, while LOPE is associated with maternal endothelial dysfunction. The innate immune system plays an essential role in normal progression of physiological pregnancy and fetal development. However, inappropriate or excessive activation of this system can lead to placental dysfunction or poor maternal vascular adaptation and contribute to the development of preeclampsia. This review aims to comprehensively outline the mechanisms of key innate immune cells including macrophages, neutrophils, natural killer (NK) cells, and innate B1 cells, in normal physiological pregnancy, EOPE and LOPE. The roles of the complement system, syncytiotrophoblast extracellular vesicles and mesenchymal stem cells (MSCs) are also discussed in the context of innate immune system regulation and preeclampsia. The outlined molecular mechanisms, which represent potential therapeutic targets, and associated emerging treatments, are evaluated as treatments for preeclampsia. Therefore, by addressing the current understanding of innate immunity in the pathogenesis of EOPE and LOPE, this review will contribute to the body of research that could lead to the development of better diagnosis, prevention, and treatment strategies. Importantly, it will delineate the differences in the mechanisms of the innate immune system in two different types of preeclampsia, which is necessary for a more personalized approach to the monitoring and treatment of affected women.
Collapse
Affiliation(s)
- Ingrid Aneman
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Dillan Pienaar
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| | - Sonja Suvakov
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Tatjana P. Simic
- Faculty of Medicine, Institute of Medical and Clinical Biochemistry, University of Belgrade, Belgrade, Serbia
- Department of Medical Sciences, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | - Vesna D. Garovic
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, United States
| | - Lana McClements
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Pinheiro-Machado E, Gurgul-Convey E, Marzec MT. Immunometabolism in type 2 diabetes mellitus: tissue-specific interactions. Arch Med Sci 2020; 19:895-911. [PMID: 37560741 PMCID: PMC10408029 DOI: 10.5114/aoms.2020.92674] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/23/2019] [Indexed: 08/11/2023] Open
Abstract
The immune system is frequently described in the context of its protective function against infections and its role in the development of autoimmunity. For more than a decade, the interactions between the immune system and metabolic processes have been reported, in effect creating a new research field, termed immunometabolism. Accumulating evidence supports the hypothesis that the development of metabolic diseases may be linked to inflammation, and reflects, in some cases, the activation of immune responses. As such, immunometabolism is defined by 1) inflammation as a driver of disease development and/or 2) metabolic processes stimulating cellular differentiation of the immune components. In this review, the main factors capable of altering the immuno-metabolic communication leading to the development and establishment of obesity and diabetes are comprehensively presented. Tissue-specific immune responses suggested to impair metabolic processes are described, with an emphasis on the adipose tissue, gut, muscle, liver, and pancreas.
Collapse
Affiliation(s)
- Erika Pinheiro-Machado
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, Netherlands
| | - Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Michal T. Marzec
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
Tanaka R, Umemura M, Narikawa M, Hikichi M, Osaw K, Fujita T, Yokoyama U, Ishigami T, Tamura K, Ishikawa Y. Reactive fibrosis precedes doxorubicin-induced heart failure through sterile inflammation. ESC Heart Fail 2020; 7:588-603. [PMID: 31984667 PMCID: PMC7160475 DOI: 10.1002/ehf2.12616] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/06/2019] [Accepted: 12/22/2019] [Indexed: 12/14/2022] Open
Abstract
Aims Doxorubicin (DOX)‐induced heart failure has a poor prognosis, and effective treatments have not been established. Because DOX shows cumulative cardiotoxicity, we hypothesized that minimal cardiac remodelling occurred at the initial stage in activating cardiac fibroblasts. Our aim was to investigate the initial pathophysiology of DOX‐exposed cardiac fibroblasts and propose prophylaxis. Methods and results An animal study was performed using a lower dose of DOX (4 mg/kg/week for 3 weeks, i.p.) than a toxic cumulative dose. Histological analysis was performed with terminal deoxynucleotidyl transferase‐mediated dUTP nick‐end labelling assay, picrosirius red staining, and immunohistochemical staining. The mechanism was analysed in vitro with a low dose of DOX, which did not induce cell apoptosis. Microarray analysis was performed. Differentially expressed genes were confirmed by enrichment analysis. Mitochondrial damage was assessed by mitochondrial membrane potential. The production of inflammatory cytokines and fibrosis markers was assessed by western blot, quantitative polymerase chain reaction, and ELISA. A phosphokinase antibody array was performed to detect related signalling pathways. Low‐dose DOX did not induced cell death, and fibrosis was localized to the perivascular area in mice. Microarray analysis suggested that DOX induced genes associated with the innate immune system and inflammatory reactions, resulting in cardiac remodelling. DOX induced mitochondrial damage and increased the expression of interleukin‐1. DOX also promoted the expression of fibrotic markers, such as alpha smooth muscle actin and galectin‐3. These responses were induced through stress‐activated protein kinase/c‐Jun NH2‐terminal kinase signalling. A peroxisome proliferator‐activated receptor (PPARγ) agonist attenuated the expression of fibrotic markers through suppressing stress‐activated protein kinase/c‐Jun NH2‐terminal kinase. Furthermore, this molecule also suppressed DOX‐induced early fibrotic responses in vivo. Conclusions Low‐dose DOX provoked reactive fibrosis through sterile inflammation evoked by the damaged mitochondria.
Collapse
Affiliation(s)
- Ryo Tanaka
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan.,Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Masanari Umemura
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan.,Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Masatoshi Narikawa
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan.,Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Mayu Hikichi
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kohei Osaw
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan
| | - Takayuki Fujita
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan.,Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Utako Yokoyama
- Department of Physiology, Tokyo Medical University, Tokyo, Japan
| | - Tomoaki Ishigami
- Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Kouichi Tamura
- Medical Science and Cardiorenal Medicine, Yokohama City University School of Medicine, Yokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Yokohama City University School of Medicine, Yokohama, Japan
| |
Collapse
|
30
|
Zentsova I, Parackova Z, Kayserova J, Palova-Jelinkova L, Vrabcova P, Volfova N, Sumnik Z, Pruhova S, Petruzelkova L, Sediva A. Monocytes contribute to DNA sensing through the TBK1 signaling pathway in type 1 diabetes patients. J Autoimmun 2019; 105:102294. [DOI: 10.1016/j.jaut.2019.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/13/2019] [Accepted: 06/14/2019] [Indexed: 12/22/2022]
|