1
|
Hansen MS, Pokharel P, Piganelli J, Sussel L. The Chicken or the Egg Dilemma: Understanding the Interplay between the Immune System and the β Cell in Type 1 Diabetes. Cold Spring Harb Perspect Med 2025; 15:a041591. [PMID: 38951031 PMCID: PMC11960692 DOI: 10.1101/cshperspect.a041591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
In this review, we explore the complex interplay between the immune system and pancreatic β cells in the context of type 1 diabetes (T1D). While T1D is predominantly considered a T-cell-mediated autoimmune disease, the inability of human leukocyte antigen (HLA)-risk alleles alone to explain disease development suggests a role for β cells in initiating and/or propagating disease. This review delves into the vulnerability of β cells, emphasizing their susceptibility to endoplasmic reticulum (ER) stress and protein modifications, which may give rise to neoantigens. Additionally, we discuss the role of viral infections as contributors to T1D onset, and of genetic factors with dual impacts on the immune system and β cells. A greater understanding of the interplay between environmental triggers, autoimmunity, and the β cell will not only lead to insight as to why the islet β cells are specifically targeted by the immune system in T1D but may also reveal potential novel therapeutic interventions.
Collapse
Affiliation(s)
- Maria Skjøtt Hansen
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Pravil Pokharel
- Division of Endocrinology Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Jon Piganelli
- Division of Endocrinology Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
2
|
Yokoi A, Asahara SI, Inoue H, Goto A, Seike M, Kido N, Suzuki H, Kanno A, Kimura-Koyanagi M, Uto K, Saegusa J, Kido Y, Ogawa W. miR378a-3p in serum extracellular vesicles is associated with pancreatic beta-cell mass in diabetic states. Biochem Biophys Res Commun 2025; 750:151367. [PMID: 39879698 DOI: 10.1016/j.bbrc.2025.151367] [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: 01/14/2025] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 01/31/2025]
Abstract
The condition in which the insulin secretory ability of pancreatic β-cells decreases in diabetes is extremely important, but there are currently no biomarkers that reflect pancreatic β-cell failure. Therefore, we conducted a search for biomarkers, using pancreatic β-cell-specific 3-Phosphoinositide-dependent protein kinase 1 (PDK1) knockout mice, which develop severe hyperglycemia due to a decrease in pancreatic β-cell mass without insulin resistance. The analysis was performed in young mice when metabolic abnormalities were not yet apparent. Comprehensive analysis of microRNAs contained in extracellular vesicles in the blood of these mice revealed that miR378a-3p levels were significantly lower in PDK1 knockout mice than in control mice. Furthermore, in other mouse models of diabetes, namely, db/db mice and streptozotocin-induced diabetic mice, there was an increase and decrease in miR378a-3p expression, respectively, in line with the number of β-cells. These results suggest that miR378a-3p contained in serum extracellular vesicles is a biomarker that reflects pancreatic β-cell mass before the onset of diabetes. It is hoped that miR378a-3p can be utilized to realize earlier diagnosis and treatment of diabetes.
Collapse
Affiliation(s)
- Aisha Yokoi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Shun-Ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Hiroyuki Inoue
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Ayano Goto
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Masako Seike
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Nozomi Kido
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Hirotaka Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Ayumi Kanno
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Maki Kimura-Koyanagi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Kenichi Uto
- Department of Clinical Laboratory, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Jun Saegusa
- Department of Rheumatology and Clinical Immunology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Yoshiaki Kido
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| |
Collapse
|
3
|
Cheng M, Wei W, Chang Y. The Role and Research Progress of ACPA in the Diagnosis and Pathological Mechanism of Rheumatoid Arthritis. Hum Immunol 2025; 86:111219. [PMID: 39700967 DOI: 10.1016/j.humimm.2024.111219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
An autoimmune condition known as rheumatoid arthritis (RA) is characterized by persistent polyarticular inflammation. Within two years of the disease's onset, irreparable bone and joint deterioration can occur as a result of the inflammatory course of the illness, leading to joint deformity and loss of function. In most patients diagnosed with RA, a range of autoantibodies, particularly anti-citrullinated protein antibodies (ACPA), can be detected months to years before the onset of clinically recognizable disease. Additionally, an increasing number of studies suggest that ACPA is involved in the pathogenesis of RA and may play a direct pathogenic role in the disease. This paper focuses on the role of ACPA in the pathomechanism of RA and discusses its unique clinical applications for the early identification and prediction of RA, as well as the influencing factors. Moreover, this article outlines the association of ACPA-positive (ACPA+) RA with other autoimmune diseases.
Collapse
Affiliation(s)
- Meng Cheng
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China.
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Center of Rheumatoid Arthritis of Anhui Medical University, Hefei 230032, China; Laboratory Animal Center, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
4
|
Srivastava N, Vomund AN, Peterson OJ, Abousaway O, Li T, Kain L, Stone P, Clement CC, Sharma S, Zhang B, Liu C, Joglekar AV, Campisi L, Hsieh CS, Santambrogio L, Teyton L, Arbelaez AM, Lichti CF, Wan X. A post-translational cysteine-to-serine conversion in human and mouse insulin generates a diabetogenic neoepitope. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622538. [PMID: 39605669 PMCID: PMC11601459 DOI: 10.1101/2024.11.07.622538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Type 1 diabetes (T1D) affects a genetically susceptible population that develops autoreactive T cells attacking insulin-producing pancreatic β cells. Increasingly, neoantigens are recognized as critical drivers of this autoimmune response. Here, we report a novel insulin neoepitope generated via post-translational cysteine-to-serine conversion (C>S) in human patients, which is also seen in the autoimmune-prone non-obese diabetic (NOD) mice. This modification is driven by oxidative stress within the microenvironment of pancreatic β cells and is further amplified by T1D-relevant inflammatory cytokines, enhancing neoantigen formation in both pancreatic β cells and dendritic cells. We discover that C>S-modified insulin is specifically recognized by CD4 + T cells in human T1D patients and NOD mice. In humans with established T1D, HLA-DQ8-restricted, C>S-specific CD4 + T cells exhibit an activated memory phenotype and lack regulatory signatures. In NOD mice, these neoepitope-specific T cells can orchestrate islet infiltration and promote diabetes progression. Collectively, these data advance a concept that microenvironment-driven and context-dependent post-translational modifications (PTMs) can generate neoantigens that contribute to organ-specific autoimmunity.
Collapse
|
5
|
Zhu JH, Ouyang SX, Zhang GY, Cao Q, Xin R, Yin H, Wu JW, Zhang Y, Zhang Z, Liu Y, Fu JT, Chen YT, Tong J, Zhang JB, Liu J, Shen FM, Li DJ, Wang P. GSDME promotes MASLD by regulating pyroptosis, Drp1 citrullination-dependent mitochondrial dynamic, and energy balance in intestine and liver. Cell Death Differ 2024; 31:1467-1486. [PMID: 39009654 PMCID: PMC11519926 DOI: 10.1038/s41418-024-01343-0] [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: 08/30/2023] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
Dysregulated metabolism, cell death, and inflammation contribute to the development of metabolic dysfunction-associated steatohepatitis (MASH). Pyroptosis, a recently identified form of programmed cell death, is closely linked to inflammation. However, the precise role of pyroptosis, particularly gasdermin-E (GSDME), in MASH development remains unknown. In this study, we observed GSDME cleavage and GSDME-associated interleukin-1β (IL-1β)/IL-18 induction in liver tissues of MASH patients and MASH mouse models induced by a choline-deficient high-fat diet (CDHFD) or a high-fat/high-cholesterol diet (HFHC). Compared with wild-type mice, global GSDME knockout mice exhibited reduced liver steatosis, steatohepatitis, fibrosis, endoplasmic reticulum stress, lipotoxicity and mitochondrial dysfunction in CDHFD- or HFHC-induced MASH models. Moreover, GSDME knockout resulted in increased energy expenditure, inhibited intestinal nutrient absorption, and reduced body weight. In the mice with GSDME deficiency, reintroduction of GSDME in myeloid cells-rather than hepatocytes-mimicked the MASH pathologies and metabolic dysfunctions, as well as the changes in the formation of neutrophil extracellular traps and hepatic macrophage/monocyte subclusters. These subclusters included shifts in Tim4+ or CD163+ resident Kupffer cells, Ly6Chi pro-inflammatory monocytes, and Ly6CloCCR2loCX3CR1hi patrolling monocytes. Integrated analyses of RNA sequencing and quantitative proteomics revealed a significant GSDME-dependent reduction in citrullination at the arginine-114 (R114) site of dynamin-related protein 1 (Drp1) during MASH. Mutation of Drp1 at R114 reduced its stability, impaired its ability to redistribute to mitochondria and regulate mitophagy, and ultimately promoted its degradation under MASH stress. GSDME deficiency reversed the de-citrullination of Drp1R114, preserved Drp1 stability, and enhanced mitochondrial function. Our study highlights the role of GSDME in promoting MASH through regulating pyroptosis, Drp1 citrullination-dependent mitochondrial function, and energy balance in the intestine and liver, and suggests that GSDME may be a potential therapeutic target for managing MASH.
Collapse
Affiliation(s)
- Jia-Hui Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shen-Xi Ouyang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guo-Yan Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi Cao
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University, Shanghai, China
| | - Rujuan Xin
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hang Yin
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing-Wen Wu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yan Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Liu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang-Tao Fu
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Yi-Ting Chen
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jia-Bao Zhang
- Department of Clinical Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China
| | - Jian Liu
- Department of Hepatic Surgery, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University, Shanghai, China.
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University, Shanghai, China.
| |
Collapse
|
6
|
Herold KC, Delong T, Perdigoto AL, Biru N, Brusko TM, Walker LSK. The immunology of type 1 diabetes. Nat Rev Immunol 2024; 24:435-451. [PMID: 38308004 PMCID: PMC7616056 DOI: 10.1038/s41577-023-00985-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 02/04/2024]
Abstract
Following the seminal discovery of insulin a century ago, treatment of individuals with type 1 diabetes (T1D) has been largely restricted to efforts to monitor and treat metabolic glucose dysregulation. The recent regulatory approval of the first immunotherapy that targets T cells as a means to delay the autoimmune destruction of pancreatic β-cells highlights the critical role of the immune system in disease pathogenesis and tends to pave the way for other immune-targeted interventions for T1D. Improving the efficacy of such interventions across the natural history of the disease will probably require a more detailed understanding of the immunobiology of T1D, as well as technologies to monitor residual β-cell mass and function. Here we provide an overview of the immune mechanisms that underpin the pathogenesis of T1D, with a particular emphasis on T cells.
Collapse
Affiliation(s)
- Kevan C Herold
- Department of Immunobiology, Yale University, New Haven, CT, USA.
- Department of Internal Medicine, Yale University, New Haven, CT, USA.
| | - Thomas Delong
- Anschutz Medical Campus, University of Colorado, Denver, CO, USA
| | - Ana Luisa Perdigoto
- Department of Internal Medicine, Yale University, New Haven, CT, USA
- Internal Medicine, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Noah Biru
- Department of Immunobiology, Yale University, New Haven, CT, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, University College London, London, UK.
- Division of Infection & Immunity, University College London, London, UK.
| |
Collapse
|
7
|
Callebaut A, Guyer P, Derua R, Buitinga M, Manganaro A, Yi X, Sodré FMC, Vig S, Suleiman M, Marchetti P, Eizirik DL, Kent SC, Mathieu C, James EA, Overbergh L. CD4+ T Cells From Individuals With Type 1 Diabetes Respond to a Novel Class of Deamidated Peptides Formed in Pancreatic Islets. Diabetes 2024; 73:728-742. [PMID: 38387030 PMCID: PMC11043062 DOI: 10.2337/db23-0588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Abstract
The β-cell plays a crucial role in the pathogenesis of type 1 diabetes, in part through the posttranslational modification of self-proteins by biochemical processes such as deamidation. These neoantigens are potential triggers for breaking immune tolerance. We report the detection by LC-MS/MS of 16 novel Gln and 27 novel Asn deamidations in 14 disease-related proteins within inflammatory cytokine-stressed human islets of Langerhans. T-cell clones responsive against one Gln- and three Asn-deamidated peptides could be isolated from peripheral blood of individuals with type 1 diabetes. Ex vivo HLA class II tetramer staining detected higher T-cell frequencies in individuals with the disease compared with control individuals. Furthermore, there was a positive correlation between the frequencies of T cells specific for deamidated peptides, insulin antibody levels at diagnosis, and duration of disease. These results highlight that stressed human islets are prone to enzymatic and biochemical deamidation and suggest that both Gln- and Asn-deamidated peptides can promote the activation and expansion of autoreactive CD4+ T cells. These findings add to the growing evidence that posttranslational modifications undermine tolerance and may open the road for the development of new diagnostic and therapeutic applications for individuals living with type 1 diabetes. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Aïsha Callebaut
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA
| | - Perrin Guyer
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven, Belgium
| | - Mijke Buitinga
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Anthony Manganaro
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Saurabh Vig
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Decio L. Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Sally C. Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Eddie A. James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA
| | - Lut Overbergh
- Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Kang Q, Ren J, Cong J, Yu W. Diabetes mellitus and idiopathic pulmonary fibrosis: a Mendelian randomization study. BMC Pulm Med 2024; 24:142. [PMID: 38504175 PMCID: PMC10953180 DOI: 10.1186/s12890-024-02961-7] [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: 08/20/2023] [Accepted: 03/11/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND The question as to whether or not diabetes mellitus increases the risk of idiopathic pulmonary fibrosis (IPF) remains controversial. This study aimed to investigate the causal association between type 1 diabetes (T1D), type 2 diabetes (T2D), and IPF using Mendelian randomization (MR) analysis. METHODS We used two-sample univariate and multivariate MR (MVMR) analyses to investigate the causal relationship between T1D or T2D and IPF. We obtained genome-wide association study (GWAS) data for T1D and T2D from the European Bioinformatics Institute, comprising 29,652 T1D samples and 101,101 T1D single nucleotide polymorphisms (SNPs) and 655,666 T2D samples and 5,030,727 T2D SNPs. We also used IPF GWAS data from the FinnGen Biobank comprising 198,014 IPF samples and 16,380,413 IPF SNPs. All cases and controls in these datasets were derived exclusively from European populations. In the univariate MR analysis, we employed inverse variance-weighted (IVW), weighted median (WM), and MR-Egger regression methods. For the MVMR analysis, we used the multivariate IVW method primarily, and supplemented it with multivariate MR-Egger and multivariate MR- least absolute shrinkage and selection operator methods. Heterogeneity tests were conducted using the MR-IVW and MR-Egger regression methods, whereas pleiotropic effects were assessed using the MR-Egger intercept. The results of MR and sensitivity analyses were visualized using forest, scatter, leave-one-out, and funnel plots. RESULTS Univariate MR revealed a significant causal relationship between T1D and IPF (OR = 1.118, 95% CI = 1.021-1.225, P = 0.016); however, no significant causal relationship was found between T2D and IPF (OR = 0.911, 95% CI = 0.796-1.043, P = 0.178). MVMR analysis further confirmed a causal association between T1D and IPF (OR = 1.133, 95% CI = 1.011-1.270, P = 0.032), but no causal relationship between T2D and IPF (OR = 1.009, 95% CI = 0.790-1.288, P = 0.950). Sensitivity analysis results validated the stability and reliability of our findings. CONCLUSION Univariate and multivariate analyses demonstrated a causal relationship between T1D and IPF, whereas no evidence was found to support a causal relationship between T2D and IPF. Therefore, in clinical practice, patients with T1D should undergo lung imaging for early detection of IPF.
Collapse
Affiliation(s)
- Quou Kang
- Department of Pulmonary and Critical Care Medicine, The affiliated hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Jing Ren
- Department of Pulmonary and Critical Care Medicine, The affiliated hospital of Qingdao University, Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Jinpeng Cong
- Department of Pulmonary and Critical Care Medicine, The affiliated hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Wencheng Yu
- Department of Pulmonary and Critical Care Medicine, The affiliated hospital of Qingdao University, Qingdao University, Qingdao, China.
| |
Collapse
|
9
|
Sharma C, Hamza A, Boyle E, Donu D, Cen Y. Post-Translational Modifications and Diabetes. Biomolecules 2024; 14:310. [PMID: 38540730 PMCID: PMC10968569 DOI: 10.3390/biom14030310] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 09/22/2024] Open
Abstract
Diabetes and its associated complications have increasingly become major challenges for global healthcare. The current therapeutic strategies involve insulin replacement therapy for type 1 diabetes (T1D) and small-molecule drugs for type 2 diabetes (T2D). Despite these advances, the complex nature of diabetes necessitates innovative clinical interventions for effective treatment and complication prevention. Accumulative evidence suggests that protein post-translational modifications (PTMs), including glycosylation, phosphorylation, acetylation, and SUMOylation, play important roles in diabetes and its pathological consequences. Therefore, the investigation of these PTMs not only sheds important light on the mechanistic regulation of diabetes but also opens new avenues for targeted therapies. Here, we offer a comprehensive overview of the role of several PTMs in diabetes, focusing on the most recent advances in understanding their functions and regulatory mechanisms. Additionally, we summarize the pharmacological interventions targeting PTMs that have advanced into clinical trials for the treatment of diabetes. Current challenges and future perspectives are also provided.
Collapse
Affiliation(s)
- Chiranjeev Sharma
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Abu Hamza
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Emily Boyle
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Dickson Donu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
| | - Yana Cen
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23219, USA; (C.S.); (A.H.); (E.B.); (D.D.)
- Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
10
|
Greeny A, Nair A, Sadanandan P, Satarker S, Famurewa AC, Nampoothiri M. Epigenetic Alterations in Alzheimer's Disease: Impact on Insulin Signaling and Advanced Drug Delivery Systems. BIOLOGY 2024; 13:157. [PMID: 38534427 DOI: 10.3390/biology13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that predominantly affects the hippocampus and the entorhinal complex, leading to memory lapse and cognitive impairment. This can have a negative impact on an individual's behavior, speech, and ability to navigate their surroundings. AD is one of the principal causes of dementia. One of the most accepted theories in AD, the amyloid β (Aβ) hypothesis, assumes that the buildup of the peptide Aβ is the root cause of AD. Impaired insulin signaling in the periphery and central nervous system has been considered to have an effect on the pathophysiology of AD. Further, researchers have shifted their focus to epigenetic mechanisms that are responsible for dysregulating major biochemical pathways and intracellular signaling processes responsible for directly or indirectly causing AD. The prime epigenetic mechanisms encompass DNA methylation, histone modifications, and non-coding RNA, and are majorly responsible for impairing insulin signaling both centrally and peripherally, thus leading to AD. In this review, we provide insights into the major epigenetic mechanisms involved in causing AD, such as DNA methylation and histone deacetylation. We decipher how the mechanisms alter peripheral insulin signaling and brain insulin signaling, leading to AD pathophysiology. In addition, this review also discusses the need for newer drug delivery systems for the targeted delivery of epigenetic drugs and explores targeted drug delivery systems such as nanoparticles, vesicular systems, networks, and other nano formulations in AD. Further, this review also sheds light on the future approaches used for epigenetic drug delivery.
Collapse
Affiliation(s)
- Alosh Greeny
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo 482123, Nigeria
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
11
|
Stern LJ, Clement C, Galluzzi L, Santambrogio L. Non-mutational neoantigens in disease. Nat Immunol 2024; 25:29-40. [PMID: 38168954 PMCID: PMC11075006 DOI: 10.1038/s41590-023-01664-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/29/2023] [Indexed: 01/05/2024]
Abstract
The ability of mammals to mount adaptive immune responses culminating with the establishment of immunological memory is predicated on the ability of the mature T cell repertoire to recognize antigenic peptides presented by syngeneic MHC class I and II molecules. Although it is widely believed that mature T cells are highly skewed towards the recognition of antigenic peptides originating from genetically diverse (for example, foreign or mutated) protein-coding regions, preclinical and clinical data rather demonstrate that novel antigenic determinants efficiently recognized by mature T cells can emerge from a variety of non-mutational mechanisms. In this Review, we describe various mechanisms that underlie the formation of bona fide non-mutational neoantigens, such as epitope mimicry, upregulation of cryptic epitopes, usage of non-canonical initiation codons, alternative RNA splicing, and defective ribosomal RNA processing, as well as both enzymatic and non-enzymatic post-translational protein modifications. Moreover, we discuss the implications of the immune recognition of non-mutational neoantigens for human disease.
Collapse
Affiliation(s)
- Lawrence J Stern
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbiology Program, UMass Chan Medical School, Worcester, MA, USA
| | - Cristina Clement
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
12
|
Ettinger RA, Buitinga M, Vandamme C, Afonso G, Gomez R, Arribas-Layton D, Bissenova S, Speake C, Reijonen H, Kinnunen T, Overbergh L, Mallone R, Kwok WW, James EA. Technical Validation and Utility of an HLA Class II Tetramer Assay for Type 1 Diabetes: A Multicenter Study. J Clin Endocrinol Metab 2023; 109:183-196. [PMID: 37474341 DOI: 10.1210/clinem/dgad434] [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: 03/17/2023] [Revised: 06/28/2023] [Accepted: 06/17/2023] [Indexed: 07/22/2023]
Abstract
CONTEXT Validated assays to measure autoantigen-specific T-cell frequency and phenotypes are needed for assessing the risk of developing diabetes, monitoring disease progression, evaluating responses to treatment, and personalizing antigen-based therapies. OBJECTIVE Toward this end, we performed a technical validation of a tetramer assay for HLA-DRA-DRB1*04:01, a class II allele that is strongly associated with susceptibility to type 1 diabetes (T1D). METHODS HLA-DRA-DRB1*04:01-restricted T cells specific for immunodominant epitopes from islet cell antigens GAD65, IGRP, preproinsulin, and ZnT8, and a reference influenza epitope, were enumerated and phenotyped in a single staining tube with a tetramer assay. Single and multicenter testing was performed, using a clone-spiked specimen and replicate samples from T1D patients, with a target coefficient of variation (CV) less than 30%. The same assay was applied to an exploratory cross-sectional sample set with 24 T1D patients to evaluate the utility of the assay. RESULTS Influenza-specific T-cell measurements had mean CVs of 6% for the clone-spiked specimen and 11% for T1D samples in single-center testing, and 20% and 31%, respectively, for multicenter testing. Islet-specific T-cell measurements in these same samples had mean CVs of 14% and 23% for single-center and 23% and 41% for multicenter testing. The cross-sectional study identified relationships between T-cell frequencies and phenotype and disease duration, sex, and autoantibodies. A large fraction of the islet-specific T cells exhibited a naive phenotype. CONCLUSION Our results demonstrate that the assay is reproducible and useful to characterize islet-specific T cells and identify correlations between T-cell measures and clinical traits.
Collapse
Affiliation(s)
- Ruth A Ettinger
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Mijke Buitinga
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Céline Vandamme
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
| | - Georgia Afonso
- Diabetes and Autoimmunity Research Laboratory, Université Paris Cité, Institut Cochin, CNRS, INSERM, 75014 Paris, France
| | - Rebecca Gomez
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - David Arribas-Layton
- Department of Immunology and Theranostics, City of Hope Medical Center, Beckman Research Institute, Duarte, CA 91010, USA
| | - Samal Bissenova
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Helena Reijonen
- Department of Immunology and Theranostics, City of Hope Medical Center, Beckman Research Institute, Duarte, CA 91010, USA
| | - Tuure Kinnunen
- Department of Clinical Microbiology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland
- Eastern Finland Laboratory Centre (ISLAB), 70210 Kuopio, Finland
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Roberto Mallone
- Diabetes and Autoimmunity Research Laboratory, Université Paris Cité, Institut Cochin, CNRS, INSERM, 75014 Paris, France
- Department of Internal Medicine, Assistance Publique Hôpitaux de Paris, Service de Diabétologie et Immunologie Clinique, Cochin Hospital, 75014 Paris, France
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| | - Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA 98101, USA
| |
Collapse
|
13
|
Yang ML, Lam TT, Kanyo J, Kang I, Zhou ZS, Clarke SG, Mamula MJ. Natural isoaspartyl protein modification of ZAP70 alters T cell responses in lupus. Autoimmunity 2023; 56:2282945. [PMID: 37994408 PMCID: PMC10897934 DOI: 10.1080/08916934.2023.2282945] [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: 06/08/2023] [Accepted: 11/08/2023] [Indexed: 11/24/2023]
Abstract
Protein posttranslational modifications (PTMs) arise in a number of normal cellular biological pathways and in response to pathology caused by inflammation and/or infection. Indeed, a number of PTMs have been identified and linked to specific autoimmune responses and metabolic pathways. One particular PTM, termed isoaspartyl (isoAsp or isoD) modification, is among the most common spontaneous PTM occurring at physiological pH and temperature. Herein, we demonstrate that isoAsp modifications arise within the ZAP70 protein tyrosine kinase upon T-cell antigen receptor (TCR) engagement. The enzyme protein L-isoaspartate O-methyltransferase (PCMT1, or PIMT, EC 2.1.1.77) evolved to repair isoaspartyl modifications in cells. In this regard, we observe that increased levels of isoAsp modification that arise under oxidative stress are correlated with reduced PIMT activity in patients with systemic lupus erythematosus (SLE). PIMT deficiency leads to T cell hyper-proliferation and hyper-phosphorylation through ZAP70 signaling. We demonstrate that inducing the overexpression of PIMT can correct the hyper-responsive phenotype in lupus T cells. Our studies reveal a phenotypic role of isoAsp modification and phosphorylation of ZAP70 in lupus T cell autoimmunity and provide a potential therapeutic target through the repair of isoAsp modification.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - TuKiet T. Lam
- Keck MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, New Haven, CT, USA
- Department of Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Jean Kanyo
- Keck MS & Proteomics Resource, WM Keck Foundation Biotechnology Resource Laboratory, New Haven, CT, USA
| | - Insoo Kang
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, USA
| | - Zhaohui Sunny Zhou
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, USA
| | - Steven G. Clarke
- Department of Chemistry and Biochemistry, Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Mark J. Mamula
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
14
|
Farahmandpour M, Kordrostami Z, Rajabzadeh M, Khalifeh R. Flexible Bio-Electronic Hybrid Metal-Oxide Channel FET as a Glucose Sensor. IEEE Trans Nanobioscience 2023; 22:855-862. [PMID: 37018717 DOI: 10.1109/tnb.2023.3236460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A high-sensitivity flexible field-effect transistor (FET) based glucose sensor is fabricated that can surpass the conventional electrochemical glucometers in terms of sensitivity, limit of detection, and other performance parameters. The proposed biosensor is based on the FET operation with the advantage of amplification which provides a high sensitivity and a very low limit of detection. Hybrid metal oxide (ZnO and CuO) nanostructures have been synthesized in the form of hollow spheres (ZnO/CuO-NHS). The FET was fabricated by depositing ZnO/CuO-NHS on the interdigitated electrodes. Glucose oxidase (GOx) was immobilized successfully on the ZnO/CuO-NHS. Three different outputs of the sensor are examined, the FET current, the relative current change, and the drain voltage. The sensitivity of the sensor for each output type has been calculated. The readout circuit can convert the current change to the voltage change that has been used for wireless transmission. The sensor has a very low limit of detection of 30 nM with satisfactory reproducibility, good stability, and high selectivity. The electrical response of the FET biosensor towards the real human blood serum samples demonstrated that it can be offered as a potential device for glucose detection in any medical application.
Collapse
|
15
|
Yang C, Wei M, Zhao Y, Yang Z, Song M, Mi J, Yang X, Tian G. Regulation of insulin secretion by the post-translational modifications. Front Cell Dev Biol 2023; 11:1217189. [PMID: 37601108 PMCID: PMC10436566 DOI: 10.3389/fcell.2023.1217189] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Post-translational modification (PTM) has a significant impact on cellular signaling and function regulation. In pancreatic β cells, PTMs are involved in insulin secretion, cell development, and viability. The dysregulation of PTM in β cells is clinically associated with the development of diabetes mellitus. Here, we summarized current findings on major PTMs occurring in β cells and their roles in insulin secretion. Our work provides comprehensive insight into understanding the mechanisms of insulin secretion and potential therapeutic targets for diabetes from the perspective of protein PTMs.
Collapse
Affiliation(s)
- Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Mengna Wei
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Yanpu Zhao
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Zhanyi Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Mengyao Song
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoyong Yang
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
16
|
Wu X, Xu M, Geng M, Chen S, Little PJ, Xu S, Weng J. Targeting protein modifications in metabolic diseases: molecular mechanisms and targeted therapies. Signal Transduct Target Ther 2023; 8:220. [PMID: 37244925 PMCID: PMC10224996 DOI: 10.1038/s41392-023-01439-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 05/29/2023] Open
Abstract
The ever-increasing prevalence of noncommunicable diseases (NCDs) represents a major public health burden worldwide. The most common form of NCD is metabolic diseases, which affect people of all ages and usually manifest their pathobiology through life-threatening cardiovascular complications. A comprehensive understanding of the pathobiology of metabolic diseases will generate novel targets for improved therapies across the common metabolic spectrum. Protein posttranslational modification (PTM) is an important term that refers to biochemical modification of specific amino acid residues in target proteins, which immensely increases the functional diversity of the proteome. The range of PTMs includes phosphorylation, acetylation, methylation, ubiquitination, SUMOylation, neddylation, glycosylation, palmitoylation, myristoylation, prenylation, cholesterylation, glutathionylation, S-nitrosylation, sulfhydration, citrullination, ADP ribosylation, and several novel PTMs. Here, we offer a comprehensive review of PTMs and their roles in common metabolic diseases and pathological consequences, including diabetes, obesity, fatty liver diseases, hyperlipidemia, and atherosclerosis. Building upon this framework, we afford a through description of proteins and pathways involved in metabolic diseases by focusing on PTM-based protein modifications, showcase the pharmaceutical intervention of PTMs in preclinical studies and clinical trials, and offer future perspectives. Fundamental research defining the mechanisms whereby PTMs of proteins regulate metabolic diseases will open new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiumei Wu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China
| | - Mengyun Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Mengya Geng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Shuo Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, 4102, Australia
- Sunshine Coast Health Institute and School of Health and Behavioural Sciences, University of the Sunshine Coast, Birtinya, QLD, 4575, Australia
| | - Suowen Xu
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Jianping Weng
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Department of Endocrinology and Metabolism, Guangdong Provincial Key Laboratory of Diabetology, The Third Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, China.
- Bengbu Medical College, Bengbu, 233000, China.
| |
Collapse
|
17
|
Moustakas AK, Nguyen H, James EA, Papadopoulos GK. Autoimmune susceptible HLA class II motifs facilitate the presentation of modified neoepitopes to potentially autoreactive T cells. Cell Immunol 2023; 390:104729. [PMID: 37301094 DOI: 10.1016/j.cellimm.2023.104729] [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: 01/30/2023] [Revised: 03/27/2023] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Rheumatoid arthritis (RA), multiple sclerosis (MS), type 1 diabetes (T1D), and celiac disease (CD), are strongly associated with susceptible HLA class II haplotypes. The peptide-binding pockets of these molecules are polymorphic, thus each HLA class II protein presents a distinct set of peptides to CD4+ T cells. Peptide diversity is increased through post-translational modifications, generating non-templated sequences that enhance HLA binding and/or T cell recognition. The high-risk HLA-DR alleles that confer susceptibility to RA are notable for their ability to accommodate citrulline, promoting responses to citrullinated self-antigens. Likewise, HLA-DQ alleles associated with T1D and CD favor the binding of deamidated peptides. In this review, we discuss structural features that promote modified self-epitope presentation, provide evidence supporting the relevance of T cell recognition of such antigens in disease processes, and make a case that interrupting the pathways that generate such epitopes and reprogramming neoepitope-specific T cells are key strategies for effective therapeutic intervention.
Collapse
Affiliation(s)
- Antonis K Moustakas
- Department of Food Science and Technology, Faculty of Environmental Sciences, Ionian University, GR26100 Argostoli, Cephalonia, Greece
| | - Hai Nguyen
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Eddie A James
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - George K Papadopoulos
- Laboratory of Biophysics, Biochemistry, Bioprocessing and Bioproducts, Faculty of Agricultural Technology, Technological Educational Institute of Epirus, GR47100 Arta, Greece
| |
Collapse
|
18
|
Ishina IA, Zakharova MY, Kurbatskaia IN, Mamedov AE, Belogurov AA, Gabibov AG. MHC Class II Presentation in Autoimmunity. Cells 2023; 12:314. [PMID: 36672249 PMCID: PMC9856717 DOI: 10.3390/cells12020314] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Antigen presentation by major histocompatibility complex class II (MHC-II) molecules is crucial for eliciting an efficient immune response by CD4+ T cells and maintaining self-antigen tolerance. Some MHC-II alleles are known to be positively or negatively associated with the risk of the development of different autoimmune diseases (ADs), including those characterized by the emergence of autoreactive T cells. Apparently, the MHC-II presentation of self-antigens contributes to the autoimmune T cell response, initiated through a breakdown of central tolerance to self-antigens in the thymus. The appearance of autoreactive T cell might be the result of (i) the unusual interaction between T cell receptors (TCRs) and self-antigens presented on MHC-II; (ii) the posttranslational modifications (PTMs) of self-antigens; (iii) direct loading of the self-antigen to classical MHC-II without additional nonclassical MHC assistance; (iv) the proinflammatory environment effect on MHC-II expression and antigen presentation; and (v) molecular mimicry between foreign and self-antigens. The peculiarities of the processes involved in the MHC-II-mediated presentation may have crucial importance in the elucidation of the mechanisms of triggering and developing ADs as well as for clarification on the protective effect of MHC-II alleles that are negatively associated with ADs.
Collapse
Affiliation(s)
- Irina A. Ishina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Maria Y. Zakharova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Inna N. Kurbatskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Azad E. Mamedov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
| | - Alexey A. Belogurov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Department of Biological Chemistry, Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| | - Alexander G. Gabibov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry RAS, 117997 Moscow, Russia
- Department of Life Sciences, Higher School of Economics, 101000 Moscow, Russia
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
19
|
Yang ML, Kibbey RG, Mamula MJ. Biomarkers of autoimmunity and beta cell metabolism in type 1 diabetes. Front Immunol 2022; 13:1028130. [PMID: 36389721 PMCID: PMC9647083 DOI: 10.3389/fimmu.2022.1028130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/13/2022] [Indexed: 09/10/2023] Open
Abstract
Posttranslational protein modifications (PTMs) are an inherent response to physiological changes causing altered protein structure and potentially modulating important biological functions of the modified protein. Besides cellular metabolic pathways that may be dictated by PTMs, the subtle change of proteins also may provoke immune attack in numerous autoimmune diseases. Type 1 diabetes (T1D) is a chronic autoimmune disease destroying insulin-producing beta cells within the pancreatic islets, a result of tissue inflammation to specific autoantigens. This review summarizes how PTMs arise and the potential pathological consequence of PTMs, with particular focus on specific autoimmunity to pancreatic beta cells and cellular metabolic dysfunction in T1D. Moreover, we review PTM-associated biomarkers in the prediction, diagnosis and in monitoring disease activity in T1D. Finally, we will discuss potential preventive and therapeutic approaches of targeting PTMs in repairing or restoring normal metabolic pathways in pancreatic islets.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Richard G. Kibbey
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Mark J. Mamula
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
20
|
Yang ML, Connolly SE, Gee RJ, Lam TT, Kanyo J, Peng J, Guyer P, Syed F, Tse HM, Clarke SG, Clarke CF, James EA, Speake C, Evans-Molina C, Arvan P, Herold KC, Wen L, Mamula MJ. Carbonyl Posttranslational Modification Associated With Early-Onset Type 1 Diabetes Autoimmunity. Diabetes 2022; 71:1979-1993. [PMID: 35730902 PMCID: PMC9450849 DOI: 10.2337/db21-0989] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/15/2022] [Indexed: 11/13/2022]
Abstract
Inflammation and oxidative stress in pancreatic islets amplify the appearance of various posttranslational modifications to self-proteins. In this study, we identified a select group of carbonylated islet proteins arising before the onset of hyperglycemia in NOD mice. Of interest, we identified carbonyl modification of the prolyl-4-hydroxylase β subunit (P4Hb) that is responsible for proinsulin folding and trafficking as an autoantigen in both human and murine type 1 diabetes. We found that carbonylated P4Hb is amplified in stressed islets coincident with decreased glucose-stimulated insulin secretion and altered proinsulin-to-insulin ratios. Autoantibodies against P4Hb were detected in prediabetic NOD mice and in early human type 1 diabetes prior to the onset of anti-insulin autoimmunity. Moreover, we identify autoreactive CD4+ T-cell responses toward carbonyl-P4Hb epitopes in the circulation of patients with type 1 diabetes. Our studies provide mechanistic insight into the pathways of proinsulin metabolism and in creating autoantigenic forms of insulin in type 1 diabetes.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT
| | - Sean E. Connolly
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT
| | - Renelle J. Gee
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT
| | - TuKiet T. Lam
- Mass Spectrometry & Proteomics Resource, W.M. Keck Foundation Biotechnology Resource Laboratory, New Haven
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT
| | - Jean Kanyo
- Mass Spectrometry & Proteomics Resource, W.M. Keck Foundation Biotechnology Resource Laboratory, New Haven
| | - Jian Peng
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT
| | - Perrin Guyer
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Farooq Syed
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Hubert M. Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL
| | - Steven G. Clarke
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA
| | - Catherine F. Clarke
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA
| | - Eddie A. James
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Cate Speake
- Center for Interventional Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Peter Arvan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Kevan C. Herold
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT
- Department of Immunobiology, Yale University, New Haven, CT
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT
| | - Mark J. Mamula
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT
| |
Collapse
|
21
|
Christophorou MA. The virtues and vices of protein citrullination. ROYAL SOCIETY OPEN SCIENCE 2022; 9:220125. [PMID: 35706669 PMCID: PMC9174705 DOI: 10.1098/rsos.220125] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/16/2022] [Indexed: 05/03/2023]
Abstract
The post-translational modification of proteins expands the regulatory scope of the proteome far beyond what is achievable through genome regulation. The field of protein citrullination has seen significant progress in the last two decades. The small family of peptidylarginine deiminase (PADI or PAD) enzymes, which catalyse citrullination, have been implicated in virtually all facets of molecular and cell biology, from gene transcription and epigenetics to cell signalling and metabolism. We have learned about their association with a remarkable array of disease states and we are beginning to understand how they mediate normal physiological functions. However, while the biochemistry of PADI activation has been worked out in exquisite detail in vitro, we still lack a clear mechanistic understanding of the processes that regulate PADIs within cells, under physiological and pathophysiological conditions. This review summarizes and discusses the current knowledge, highlights some of the unanswered questions of immediate importance and gives a perspective on the outlook of the citrullination field.
Collapse
|