1
|
Dunbar EK, Greer PJ, Saloman JL, Albers KM, Yadav D, Whitcomb DC. Genetics of constant and severe pain in the NAPS2 cohort of recurrent acute and chronic pancreatitis patients. THE JOURNAL OF PAIN 2025; 27:104754. [PMID: 39674387 DOI: 10.1016/j.jpain.2024.104754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/08/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Recurrent acute and chronic pancreatitis (RAP, CP) are complex, progressive inflammatory diseases with variable pain experiences impacting patient function and quality of life. The genetic variants and pain pathways in patients contributing to most severe pain experiences are unknown. We used previously genotyped individuals with RAP/CP from the North American Pancreatitis Study II (NAPS2) of European Ancestry for nested genome-wide associated study (GWAS) for pain-severity, chronicity, or both. Lead variants from GWAS were determined using FUMA. Loci with p<1e-5 were identified for post-hoc candidate identification. Transcriptome-wide association studies (TWAS) identified loci in cis and trans to the lead variants. Serum from phenotyped individuals with CP from the PROspective Evaluation of Chronic Pancreatitis for EpidEmiologic and Translational StuDies (PROCEED) was assessed for BDNF levels using Meso Scale Discovery Immunoassay. We identified four pain systems defined by candidate genes: 1) Pancreas-associated injury/stress mitigation genes include: REG gene cluster, CTRC, NEURL3 and HSF22. 2) Neural development and axon guidance tracing genes include: SNPO, RGMA, MAML1 and DOK6 (part of the RET complex). 3) Genes linked to psychiatric stress disorders include TMEM65, RBFOX1, and ZNF385D. 4) Genes in the dorsal horn pain-modulating BDNF/neuropathic pathway included SYNPR, NTF3 and RBFOX1. In an independent cohort BDNF was significantly elevated in patients with constant-severe pain. Extension and expansion of this exploratory study may identify pathway- and mechanism-dependent targets for individualized pain treatments in CP patients. PERSPECTIVE: Pain is the most distressing and debilitating feature of chronic pancreatitis. Yet many patients with chronic pancreatitis have little or no pain. The North American Pancreatitis Study II (NAPS2) includes over 1250 pancreatitis patients of all progressive stages with all clinical and phenotypic characteristics carefully recorded. Pain did not correlate well with disease stage, inflammation, fibrosis or other features. Here we spit the patients into groups with the most severe pain and/or chronic pain syndromes and compared them genetically with patients reporting mild or minimal pain. Although some genetic variants associated with pain were expressed in cells (1) of the pancreas, most genetic variants were linked to genes expressed in the nervous system cells associated with (2) neural development and axon guidance (as needed for the descending inhibition pathway), (3) psychiatric stress disorders, and (4) cells regulating sensory nerves associated with BDNF and neuropathic pain. Similar and overlapping genetic variants in systems 2 -4 are also seen in pain syndromes form other organs. The implications for treating pancreatic pain are great in that we can no longer focus on just the pancreas. Furthermore, new treatments designed for pain disorders in other tissues may be effective in some patient with pain syndromes from the pancreas. Further research is needed to replicate and extend these observations so that new, genetics-guided rational treatments can be developed and delivered.
Collapse
Affiliation(s)
- Ellyn K Dunbar
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Phil J Greer
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jami L Saloman
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn M Albers
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dhiraj Yadav
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David C Whitcomb
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology & Molecular Physiology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Austin MC, Muralidharan C, Roy S, Crowder JJ, Piganelli JD, Linnemann AK. Dysfunctional β-cell autophagy induces β-cell stress and enhances islet immunogenicity. Front Immunol 2025; 16:1504583. [PMID: 39944686 PMCID: PMC11814175 DOI: 10.3389/fimmu.2025.1504583] [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: 10/01/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Background Type 1 Diabetes (T1D) is caused by a combination of genetic and environmental factors that trigger autoimmune-mediated destruction of pancreatic β-cells. Defects in β-cell stress response pathways such as autophagy may play an important role in activating and/or exacerbating the immune response in disease development. Previously, we discovered that β-cell autophagy is impaired prior to the onset of T1D, implicating this pathway in T1D pathogenesis. Aims To assess the role of autophagy in β-cell health and survival, and whether defects in autophagy render islets more immunogenic. Methods We knocked out the critical autophagy enzyme, ATG7, in the β-cells of mice (ATG7Δβ-cell) then monitored blood glucose, performed glucose tolerance tests, and evaluated bulk islet mRNA and protein. We also assessed MHC-I expression and presence of CD45+ immune cells in ATG7Δβ-cell islets and evaluated how impaired autophagy affects EndoC-βH1 HLA-I expression under basal and IFNα stimulated conditions. Lastly, we co-cultured ATG7Δβ-cell islet cells with diabetogenic BDC2.5 helper T cells and evaluated T cell activation. Results We found that all ATG7Δβ-cell mice developed diabetes between 11-15 weeks of age. Gene ontology analysis revealed a significant upregulation of pathways involved in inflammatory processes, response to ER stress, and the ER-associated degradation pathway. Interestingly, we also observed upregulation of proteins involved in MHC-I presentation, suggesting that defective β-cell autophagy may alter the immunopeptidome, or antigen repertoire, and enhance β-cell immune visibility. In support of this hypothesis, we observed increased MHC-I expression and CD45+ immune cells in ATG7Δβ-cell islets. We also demonstrate that HLA-I is upregulated in EndoC β-cells when autophagic degradation is inhibited. This effect was observed under both basal and IFNα stimulated conditions. Conversely, a stimulator of lysosome acidification/function, C381, decreased HLA-I expression. Lastly, we showed that in the presence of islet cells with defective autophagy, there is enhanced BDC2.5 T cell activation. Conclusions Our findings demonstrate that β-cell autophagy is critical to cell survival/function. Defective β-cell autophagy induces ER stress, alters pathways of antigen production, and enhances MHC-I/HLA-I presentation to surveilling immune cells. Overall, our results suggest that defects in autophagy make β-cells more susceptible to immune attack and destruction.
Collapse
Affiliation(s)
- Matthew C. Austin
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Charanya Muralidharan
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Saptarshi Roy
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Justin J. Crowder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Jon D. Piganelli
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Amelia K. Linnemann
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, United States
- Indiana Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
3
|
Yu L, Wu Q, Jiang S, Liu J, Liu J, Chen G. Controversial Roles of Regenerating Family Proteins in Tissue Repair and Tumor Development. Biomedicines 2024; 13:24. [PMID: 39857608 PMCID: PMC11762848 DOI: 10.3390/biomedicines13010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Over the past 40 years since the discovery of regenerating family proteins (Reg proteins), numerous studies have highlighted their biological functions in promoting cell proliferation and resisting cell apoptosis, particularly in the regeneration and repair of pancreatic islets and exocrine glands. Successively, short peptides derived from Reg3δ and Reg3α have been employed in clinical trials, showing favorable therapeutic effects in patients with type I and type II diabetes. However, continued reports have been limited, presumably attributed to the potential side effects. Methods: This review summarizes extensive research on Reg proteins over the past decade, combined with our own related studies, proposing that Reg proteins exhibit dimorphic effects. Results: The activity of Reg proteins is not as simplistic as previously perceived but shows auto-immunogenicity depending on different pathophysiological microenvironments. The immunogenicity of Reg proteins could recruit immune cells leading to an anti-tumor effect. Such functional diversity is correlated with their structural characteristics: the N-terminal region contributes to autoantigenicity, while the C-type lectin fragment near the C-terminal determines the trophic action. It should be noted that B-cell masking antigens might also reside within the C-type lectin domain. Conclusions: Reg proteins have dual functional roles under various physiological and pathological conditions. These theoretical foundations facilitate the subsequent development of diagnostic reagents and therapeutic drugs targeting Reg proteins.
Collapse
Affiliation(s)
- Luting Yu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Qingyun Wu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Shenglong Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Jia Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| | - Junli Liu
- MeDiC Program, The Research Institute of McGill University Health Centre, Montreal, QC H4A 3J1, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211816, China; (L.Y.)
| |
Collapse
|
4
|
Fan H, Shi Y, Liu H, Zuo X, Yang Y, Yin H, Li Y, Wang X, Liu L, Wang F, Han H, Wu Q, Yang N, Tang Y, Lu G. Inhalation of H 2/O 2 (66.7 %/33.3 %) mitigates depression-like behaviors in diabetes mellitus complicated with depression mice via suppressing inflammation and preventing hippocampal damage. Biomed Pharmacother 2024; 180:117559. [PMID: 39405908 DOI: 10.1016/j.biopha.2024.117559] [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: 06/15/2024] [Revised: 09/16/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
Diabetes mellitus complicated with depression (DD) is a prevalent psychosomatic disorder. It is characterized by severe cognitive impairment, and associated with high rates of disability and mortality. Although conventional treatment options are available, the efficacy of these regimens in managing DD remains limited. Molecular hydrogen (H2), a selective hydroxyl radical scavenger, has shown therapeutic potential in the treatment of various systemic diseases. This study aims to investigate the therapeutic effects of H2 on DD. A DD mouse model was established through intraperitoneal injection of streptozotocin (STZ, 150 mg/kg) and lipopolysaccharide (LPS, 0.5 mg/kg). Following the induction of DD, the mice were treated with H2/O2 (66.7 %/33.3 %)inhalation for 7 days. Behavioral assessments were conducted by standard behavioral tests, and the levels of inflammatory cytokines in peripheral blood serum and hippocampal tissue were measured using enzyme-linked immunosorbent assay (ELISA). Furthermore, magnetic resonance imaging (MRI) scans and immunofluorescence staining of the hippocampus were performed to evaluate hippocampal structural integrity. The results demonstrated that inhalation of H2/O2 (66.7 %/33.3 %) significantly ameliorated depressive behaviors and symptoms in DD mice, reversed hippocampal volume reduction, decreased inflammatory cytokine levels in peripheral blood serum and hippocampal tissue, and inhibited the activation of A1 astrocytes in the hippocampus. Our study suggests that H2/O2 (66.7 %/33.3 %) inhalation therapy may offer a promising treatment strategy for DD and its associated symptoms.
Collapse
Affiliation(s)
- Huaju Fan
- Medical Laboratory Animal Center, School of Psychology, Shandong Second Medical University, Weifang, Shandong 261053, China; Sichuan Second Veterans Hospital, Chengdu, Sichuan 611230, China
| | - Yanhua Shi
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Haiqiang Liu
- Weifang People's Hospital Weifang, Shandong 261000, China
| | - Xiaofei Zuo
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Yanmei Yang
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Hao Yin
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Yanyan Li
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Xianghui Wang
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Li Liu
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Fengjiao Wang
- Medical Laboratory Animal Center, School of Public Health, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Huifang Han
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Qianying Wu
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China
| | - Nana Yang
- Medical Laboratory Animal Center, School of Bioscience and Technology, Shandong Second Medical University, Weifang, Shandong 261053, China.
| | - Yaohui Tang
- School of Biomedical Engineering and Affiliated Sixth People's Hospital, Shanghai JiaoTong University, 1954 Hua Shan Rd., Shanghai 200030, China.
| | - Guohua Lu
- Medical Laboratory Animal Center, School of Psychology, Shandong Second Medical University, Weifang, Shandong 261053, China.
| |
Collapse
|
5
|
Tegehall A, Ingvast S, Krogvold L, Dahl-Jørgensen K, Korsgren O. Reduced expression of central innate defense molecules in pancreatic biopsies from subjects with Type 1 diabetes. Acta Diabetol 2024; 61:1117-1127. [PMID: 38717484 PMCID: PMC11379773 DOI: 10.1007/s00592-024-02286-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 04/06/2024] [Indexed: 09/07/2024]
Abstract
AIMS/HYPOTHESIS Defensins play a crucial role in the innate immune system's first defense against microbial threats. However, little is known about the defensin system in the pancreas, especially in relation to Type 1 diabetes. We explore the expression of defensins in different disease stages of Type 1 diabetes and correlated obtained findings to the degree of inflammation, providing new insights into the disease and the innate immune system. MATERIAL AND METHODS Pancreases from non-diabetic human organ donors of different age groups and donors with Type 1 diabetes with different disease duration were examined. Sections from head, body and tail of the pancreas were stained for eight different defensins and for immune cells; CD3+, CD45+, CD68+ and NES+ (granulocytes). RESULTS In non-diabetic adult controls the level of expression for defensins Beta-1,Alpha-1, Cathelicidin and REG3A correlated with the level of inflammation. In contrast, individuals with Type 1 diabetes exhibit a reduction or absence of several central defensins regardless of the level of inflammation in their pancreas. The expression of Cathelicidin is present in neutrophils and macrophages but not in T-cells in subjects with Type 1 diabetes. CONCLUSIONS Obtained findings suggest a pancreatic dysfunction in the innate immune system and the bridging to the adaptive system in Type 1 diabetes. Further studies on the role of the local innate immune system in Type 1 diabetes is needed.
Collapse
Affiliation(s)
- Angie Tegehall
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden.
| | - Sofie Ingvast
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| | - Lars Krogvold
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Olle Korsgren
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85, Uppsala, Sweden
| |
Collapse
|
6
|
Zhou YH, Yu LT, Wang XN, Li YJ, Xu KY, Li X, Pu CC, Xie FL, Xie BB, Gao Y, Luo C. Reg2 treatment is protective but the induced Reg2 autoantibody is destructive to the islets in NOD mice. Biochem Pharmacol 2024; 227:116444. [PMID: 39038551 DOI: 10.1016/j.bcp.2024.116444] [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: 01/04/2024] [Revised: 07/14/2024] [Accepted: 07/18/2024] [Indexed: 07/24/2024]
Abstract
Regenerating family protein 2 (Reg2) is a trophic factor which stimulates β-cell replication and resists islet destruction. However, Reg2 also serves as an islet autoantigen, which makes it complicated to judge the effectiveness in treating diabetes. How Reg2 treatment behaves in non-obese diabetic (NOD) mice is to be investigated. NOD mice were treated with recombinant Reg2 protein, Complete Freund's adjuvant (CFA) + PBS and CFA+Reg2 vaccinations, CFA+PBS- and CFA+Reg2-immunized antisera, and single chain variable fragment (scFv)-Reg2 and mIgG2a-Reg2 antibodies. Glycemic level, bodyweight, serum Reg2 antibody titer, glucose tolerance, and insulin secretion were determined. Islet morphological characteristics, insulitis, cell apoptosis, islet cell components, and T cell infiltration were analyzed by histological examinations. The autoantigenicity of constructed Reg2C and Reg2X fragments was determined in healthy BALB/c mice, and the bioactivity in stimulating cell proliferation and survival was assessed in insulinoma MIN6 cells. Reg2 administration alleviated diabetes in NOD mice with improved glucose tolerance and insulin secretion but elevated serum Reg2 autoantibodies. Histomorphometry showed reduced inflammatory area, TUNEL signal and CD8 + T cell infiltration, and increased β-cell proportion in support of the islet-protective effect of Reg2 treatment. CFA+PBS and CFA+Reg2 immunizations prevented diabetic onset and alleviated insulitis while injections of the antisera offered mild protections. Antibody treatments accelerated diabetic onset without increasing the overall incidence. Reg2C fragment depletes antigenicity, but reserves protective activity in streptozotocin (STZ)-treated MIN6 cells. In conclusion, Reg2 treatment alleviates type 1 diabetes (T1D) by preserving islet β-cells, but induces Reg2 autoantibody production which poses a potential risk of accelerating diabetic progression.
Collapse
Affiliation(s)
- Yi-Han Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lu-Ting Yu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China; School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Xiao-Nan Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - You-Jie Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ke-Yi Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chun-Cheng Pu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Fei-Lu Xie
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bing-Bing Xie
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yan Gao
- Institute of Suzhou Biobank, Suzhou Center for Disease Prevention and Control, Suzhou, China; Suzhou Institute of Advanced Study in Public Health, Gusu School, Nanjing Medical University, Suzhou, China.
| | - Chen Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China; Antibody Engineering Laboratory, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
7
|
Xu KY, Li M, Yu WH, Li X, Zeng Y, Xie FL, Zhou YH, Xu PS, Pu CC, Xie BB, Yu LT, Luo C. Reg3A Overexpression Facilitates Hepatic Metastasis by Altering Cell Adhesion in LoVo Colon Cancer Cells. DNA Cell Biol 2024; 43:298-310. [PMID: 38771249 DOI: 10.1089/dna.2024.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Affiliation(s)
- Ke-Yi Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mao Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wei-Hong Yu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yuan Zeng
- Department of Clinical Pharmacology and Bioanalytics, Pfizer (China) Research and Development Co., Ltd., Shanghai, China
| | - Fei-Lu Xie
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yi-Han Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Pin-Shen Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chun-Cheng Pu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bing-Bing Xie
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Lu-Ting Yu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China
| | - Chen Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
- Antibody Engineering Laboratory, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
8
|
Wang J, Yan D, Cui H, Zhang R, Ma X, Chen L, Hu C, Wu J. Identification of eight genomic protective alleles for mitochondrial diabetes by Kinship-graph convolutional network. J Diabetes Investig 2024; 15:52-62. [PMID: 38157301 PMCID: PMC10759726 DOI: 10.1111/jdi.14125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
AIMS Nearly 85% of maternally inherited diabetes and deafness (MIDD) are caused by the m.3243A>G mutation in the mitochondrial DNA. However, the clinical phenotypes of MIDD may also be influenced by the nuclear genome, this study aimed to investigate nuclear genome variants that influence clinical phenotypes associated with m.3243A>G mutation in MIDD based on whole-genome sequencing of the patients belonging to pedigrees. MATERIALS AND METHODS We analyzed a whole-genome sequencing (WGS) dataset from blood samples of 38 MIDD patients with the m.3243A > G mutation belonging to 10 pedigrees, by developing a Kinship-graph convolutional network approach, called Ki-GCN, integrated with the conventional genome-wide association study (GWAS) methods. RESULTS We identified eight protective alleles in the nuclear genome that have protective effects against the onset of MIDD, related deafness, and also type 2 diabetes. Based on these eight protective alleles, we constructed an effective logistic regression model to predict the early or late onset of MIDD patients. CONCLUSIONS There are protective alleles in the nuclear genome that are associated with the onset-age of MIDD patients and might also provide protective effects on the deafness derived from MIDD patients.
Collapse
Affiliation(s)
- Jiahao Wang
- CAS Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
| | - Dandan Yan
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Diabetes InstituteShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Haoyue Cui
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Rong Zhang
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Diabetes InstituteShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Xiaojing Ma
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Diabetes InstituteShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
| | - Luonan Chen
- CAS Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouZhejiangChina
| | - Cheng Hu
- Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Diabetes InstituteShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghaiChina
- Institute for Metabolic DiseaseFengxian Central Hospital Affiliated to Southern Medical UniversityShanghaiChina
| | - Jiarui Wu
- CAS Key Laboratory of Systems Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesHangzhouZhejiangChina
| |
Collapse
|
9
|
Yu L, Zhou Y, Sun S, Wang R, Yu W, Xiao H, Yu Z, Luo C. Tumor-suppressive effect of Reg3A in COAD is mediated by T cell activation in nude mice. Biomed Pharmacother 2023; 169:115922. [PMID: 38011786 DOI: 10.1016/j.biopha.2023.115922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/14/2023] [Accepted: 11/20/2023] [Indexed: 11/29/2023] Open
Abstract
Regenerating family protein 3 A (Reg3A) is highly expressed in a variety of organs and inflammatory tissues, and is closely related to tumorigenesis and cancer progression. However, clinical statistics show that high expression of Reg3A is associated with better prognosis in colorectal cancer (CRC) patients, suggesting a tumor-suppressive effect. The precise action and underlying mechanism of Reg3A in CRC remain controversial. The present study sought to investigate the relationship among Reg3A expression, CRC development, and immune cell alteration in patients using the TCGA, GEPIA, PrognoScan, TIMER and TISIDB databases. Reg3A-overexpressing LoVo cell line (LoVo-Reg3A), a representative of colon adenocarcinoma (COAD), was constructed and the action of Reg3A was assessed in a xenograft nude mouse model. Our bioinformatical analyses revealed that Reg3A upregulation is highly associated with CRC, along with increased frequency of immune cell infiltration. In the xenograft nude mice, Reg3A overexpression offered a tumor-suppressive effect by inhibiting cell proliferation and promoting apoptosis. The result of RNA-seq suggested a positive regulation of leukocytes and an upregulation of T cells in LoVo-Reg3A tumor tissue. CD4+ and CD8+ T cells in tumors, splenic Reg3A-reactive IFN-γ+/CD4+ T cells, and serum TNF-α, IFN-γ and IL-17 were significantly increased by Reg3A overexpression. In the ex vivo co-culture experiment, elevated cytotoxic effect, increased proportion of CD3ε+ T cells, and upregulated expressions of TNF-α, IFN-γ and IL-17 were detected in the PBMCs isolated from LoVo-Reg3A cell-xenografted nude mice. In conclusion, high expression of Reg3A could activate and recruit T cells in COAD leading to the cytotoxic tumor-suppressive effect.
Collapse
Affiliation(s)
- Luting Yu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China; School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Yihan Zhou
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Shaozheng Sun
- College of Science, Northeastern University, Boston, United States
| | - Runlin Wang
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Weihong Yu
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Hanyu Xiao
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China
| | - Zhuxi Yu
- Department of critical care medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Chen Luo
- School of Life Science & Technology, China Pharmaceutical University, Nanjing, China; State Key Laboratory of Nature Medicines, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
10
|
Hou X, Chen Y, Zhou B, Tang W, Ding Z, Chen L, Wu Y, Yang H, Du C, Yang D, Ma G, Cao H. Talin-1 inhibits Smurf1-mediated Stat3 degradation to modulate β-cell proliferation and mass in mice. Cell Death Dis 2023; 14:709. [PMID: 37903776 PMCID: PMC10616178 DOI: 10.1038/s41419-023-06235-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2023]
Abstract
Insufficient pancreatic β-cell mass and reduced insulin expression are key events in the pathogenesis of diabetes mellitus (DM). Here we demonstrate the high expression of Talin-1 in β-cells and that deficiency of Talin-1 reduces β-cell proliferation, which leads to reduced β-cell mass and insulin expression, thus causing glucose intolerance without affecting peripheral insulin sensitivity in mice. High-fat diet fed exerbates these phenotypes. Mechanistically, Talin-1 interacts with the E3 ligase smad ubiquitination regulatory factor 1 (Smurf1), which prohibits ubiquitination of the signal transducer and activator of transcription 3 (Stat3) mediated by Smurf1, and ablation of Talin-1 enhances Smurf1-mediated ubiquitination of Stat3, leading to decreased β-cell proliferation and mass. Furthermore, haploinsufficiency of Talin-1 and Stat3 genes, but not that of either gene, in β-cell in mice significantly impairs glucose tolerance and insulin expression, indicating that both factors indeed function in the same genetic pathway. Finally, inducible deletion Talin-1 in β-cell causes glucose intolerance in adult mice. Collectively, our findings reveal that Talin-1 functions as a crucial regulator of β-cell mass, and highlight its potential as a therapeutic target for DM patients.
Collapse
Affiliation(s)
- Xiaoting Hou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhen Ding
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yun Wu
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital; Guangdong Provincial High-level Clinical Key Specialty; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University; The Hong Kong University of Science and Technology Medical Center, Guangdong, China
| | - Hongyu Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital; Guangdong Provincial High-level Clinical Key Specialty; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University; The Hong Kong University of Science and Technology Medical Center, Guangdong, China
| | - Changzheng Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dazhi Yang
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
11
|
Shin JH, Bozadjieva-Kramer N, Seeley RJ. Reg3γ: current understanding and future therapeutic opportunities in metabolic disease. Exp Mol Med 2023; 55:1672-1677. [PMID: 37524871 PMCID: PMC10474034 DOI: 10.1038/s12276-023-01054-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 05/01/2023] [Indexed: 08/02/2023] Open
Abstract
Regenerating family member gamma, Reg3γ (the mouse homolog of human REG3A), belonging to the antimicrobial peptides (AMPs), functions as a part of the host immune system to maintain spatial segregation between the gut bacteria and the host in the intestine via bactericidal activity. There is emerging evidence that gut manipulations such as bariatric surgery, dietary supplementation or drug treatment to produce metabolic benefits alter the gut microbiome. In addition to changes in a wide range of gut hormones, these gut manipulations also induce the expression of Reg3γ in the intestine. Studies over the past decades have revealed that Reg3γ not only plays a role in the gut lumen but can also contribute to host physiology through interaction with the gut microbiota. Herein, we discuss the current knowledge regarding the biology of Reg3γ, its role in various metabolic functions, and new opportunities for therapeutic strategies to treat metabolic disorders.
Collapse
Affiliation(s)
- Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|