1
|
Mirghani HO. Glucagon-like peptide-1 agonists: Role of the gut in hypoglycemia unawareness, and the rationale in type 1 diabetes. World J Diabetes 2024; 15:2167-2172. [DOI: 10.4239/wjd.v15.i11.2167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/30/2024] [Accepted: 09/05/2024] [Indexed: 10/16/2024] Open
Abstract
Type 1 diabetes is increasing and the majority of patients have poor glycemic control. Although advanced technology and nanoparticle use have greatly enhanced insulin delivery and glucose monitoring, weight gain and hypo-glycemia remain major challenges and a constant source of concern for patients with type 1 diabetes. Type 1 diabetes shares some pathophysiology with type 2 diabetes, and an overlap has been reported. The above observation created great interest in glucagon-like peptide-1 receptor agonists (GLP-1) as adjuvants for type 1 diabetes. Previous trials confirmed the positive influence of GLP-1 agonists on β cell function. However, hypoglycemia unawareness and dysregulated glucagon response have been previously reported in patients with recurrent hypoglycemia using GLP-1 agonists. Jin et al found that the source of glucagon dysregulation due to GLP-1 agonists resides in the gut. Plausible explanations could be gut nervous system dysregulation or gut microbiota disruption. This review evaluates the potential of GLP-1 agonists in managing type 1 diabetes, particularly focusing on their impact on glycemic control, weight management, and glucagon dysregulation. We provide a broader insight into the problem of type 1 diabetes mellitus management in the light of recent findings and provide future research directions.
Collapse
Affiliation(s)
- Hyder O Mirghani
- Internal Medicine, University of Tabuk, Tabuk 51941, Tabuk, Saudi Arabia
| |
Collapse
|
2
|
Kryska A, Depciuch J, Krysa M, Paja W, Wosiak A, Nicoś M, Budzynska B, Sroka-Bartnicka A. Lipids balance as a spectroscopy marker of diabetes. Analysis of FTIR spectra by 2D correlation and machine learning analyses. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 320:124653. [PMID: 38901232 DOI: 10.1016/j.saa.2024.124653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/28/2024] [Accepted: 06/11/2024] [Indexed: 06/22/2024]
Abstract
The number of people suffering from type 2 diabetes has rapidly increased. Taking into account, that elevated intracellular lipid concentrations, as well as their metabolism, are correlated with diminished insulin sensitivity, in this study we would like to show lipids spectroscopy markers of diabetes. For this purpose, serum collected from rats (animal model of diabetes) was analyzed using Fourier Transformed Infrared-Attenuated Total Reflection (FTIR-ATR) spectroscopy. Analyzed spectra showed that rats with diabetes presented higher concentration of phospholipids and cholesterol in comparison with non-diabetic rats. Moreover, the analysis of second (IInd) derivative spectra showed no structural changes in lipids. Machine learning methods showed higher accuracy for IInd derivative spectra (from 65 % to 89 %) than for absorbance FTIR spectra (53-65 %). Moreover, it was possible to identify significant wavelength intervals from IInd derivative spectra using random forest-based feature selection algorithm, which further increased the accuracy of the classification (up to 92 % for phospholipid region). Moreover decision tree based on the selected features showed, that peaks at 1016 cm-1 and 2936 cm-1 can be good candidates of lipids marker of diabetes.
Collapse
Affiliation(s)
- Adrianna Kryska
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Depciuch
- Institute of Nuclear Physics, Polish Academy of Sciences, Walerego Eljasza - Radzikowskiego 152, 31-342 Kraków, Poland; Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodźki 1, Lublin 20-093, Poland
| | - Mikolaj Krysa
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiesław Paja
- Institute of Computer Science, University of Rzeszow, Pigonia 1, 35-310 Rzeszów, Poland
| | - Agnieszka Wosiak
- Institute of Information Technology, Lodz University of Technology, Politechniki 8, 93-590 Łódź, Poland
| | - Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-090 Lublin, Poland
| | - Barbara Budzynska
- Independent Laboratory of Behavioral Studies, Faculty of Biomedical Sciences, Medical University of Lublin, Chodzki 4a, 20-093 Lublin, Poland
| | - Anna Sroka-Bartnicka
- Independent Unit of Spectroscopy and Chemical Imaging, Faculty of Biomedical Sciences, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland.
| |
Collapse
|
3
|
Eizirik DL, Zimath PL, Yi X, Roca Rivada A, Richardson SJ. Comment on the role of interferons in the pathology of beta cell destruction in type 1 diabetes. Reply to Lenzen S [letter]. Diabetologia 2024; 67:2600-2602. [PMID: 39231828 DOI: 10.1007/s00125-024-06269-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/06/2024]
Affiliation(s)
- Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium.
| | - Priscila L Zimath
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Xiaoyan Yi
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Arturo Roca Rivada
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Sarah J Richardson
- Islet Biology Exeter (IBEx), Exeter Centre of Excellence for Diabetes Research (EXCEED), Department of Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter, UK
| |
Collapse
|
4
|
Lee SH, Lin TA, Yan YH, Chien CC, Cheng TJ. Hepatic and metabolic outcomes induced by sub-chronic exposure to polystyrene microplastics in mice. Arch Toxicol 2024; 98:3811-3823. [PMID: 39183192 DOI: 10.1007/s00204-024-03847-7] [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: 04/26/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Microplastics (MPs) have attracted significant attention due to their global distribution in living environments. Although some studies have reported MP-induced hepatotoxicity in mouse models, a systematic approach to MP-mediated liver toxicity was still lacking. Therefore, we used a mouse model to study the sub-chronic effects of MP exposure on the liver. Female C57BL/6 mice, aged 6 weeks, received an oral administration of 0.3 mg of Nile Red-labeled polystyrene (PS) microplastics, with particle sizes of 0.5 µm (submicron) and 5 µm (micron), via gavage, while control mice received vehicle only. Each mouse was exposed to MPs twice a week for 12 weeks. After sacrifice, the levels of MP accumulation, oxidative stress, inflammation, and pathological changes were measured in the mouse liver, and blood samples were collected for serum biochemistry analysis. Our results demonstrated that 0.5 µm PS-MPs were accumulated in mouse livers post-MP exposure, but not in the 5 µm MP exposure group. Simultaneously, increased levels of glucose, triglyceride, alanine transaminase (ALT), aspartate transaminase (AST), superoxide dismutase, 4-hydroxy-2-nonenal-mercapturic acid (HNE-MA), interleukin-6, and lipid droplets were found in the 0.5 µm MP exposure group, while the fewer responses, including elevated liver weight index, glucose, high-density lipoprotein, AST, and decreased HNE-MA were observed in 5 µm MP exposure group. These results indicate that sub-chronic exposure to submicron MPs causes MP deposition in mouse livers, which further induces oxidative stress, increases inflammatory cytokines and perturbs glucose and lipid homeostasis, which might trigger more severe metabolic dysfunction or non-alcoholic steatohepatitis-like hepatotoxicity.
Collapse
Affiliation(s)
- Sheng-Han Lee
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ting-An Lin
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd, Taipei, 100, Taiwan
| | - Yuan-Horng Yan
- Department of Endocrinology and Metabolism, Kuang Tien General Hospital, Taichung, Taiwan
- Department of Nutrition and Institute of Biomedical Nutrition, Hung Kuang University, Taichung, Taiwan
| | - Chu-Chun Chien
- Department of Pathology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsun-Jen Cheng
- Institute of Environmental and Occupational Health Sciences, College of Public Health, National Taiwan University, No. 17, Xuzhou Rd, Taipei, 100, Taiwan.
| |
Collapse
|
5
|
Abdalla MMI. Advancing diabetes management: Exploring pancreatic beta-cell restoration's potential and challenges. World J Gastroenterol 2024; 30:4339-4353. [PMID: 39494103 PMCID: PMC11525866 DOI: 10.3748/wjg.v30.i40.4339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/05/2024] [Accepted: 09/24/2024] [Indexed: 10/16/2024] Open
Abstract
Diabetes mellitus, characterized by chronic hyperglycemia due to insulin deficiency or resistance, poses a significant global health burden. Central to its pathogenesis is the dysfunction or loss of pancreatic beta cells, which are res-ponsible for insulin production. Recent advances in beta-cell regeneration research offer promising strategies for diabetes treatment, aiming to restore endogenous insulin production and achieve glycemic control. This review explores the physiological basis of beta-cell function, recent scientific advan-cements, and the challenges in translating these findings into clinical applications. It highlights key developments in stem cell therapy, gene editing technologies, and the identification of novel regenerative molecules. Despite the potential, the field faces hurdles such as ensuring the safety and long-term efficacy of regen-erative therapies, ethical concerns around stem cell use, and the complexity of beta-cell differentiation and integration. The review highlights the importance of interdisciplinary collaboration, increased funding, the need for patient-centered approaches and the integration of new treatments into comprehensive care strategies to overcome these challenges. Through continued research and collaboration, beta-cell regeneration holds the potential to revolutionize diabetes care, turning a chronic condition into a manageable or even curable disease.
Collapse
Affiliation(s)
- Mona Mohamed Ibrahim Abdalla
- Department of Human Biology, School of Medicine, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| |
Collapse
|
6
|
Yao X, Gong Z, Yin W, Li H, Douroumis D, Huang L, Li H. Islet cell spheroids produced by a thermally sensitive scaffold: a new diabetes treatment. J Nanobiotechnology 2024; 22:657. [PMID: 39456025 PMCID: PMC11515210 DOI: 10.1186/s12951-024-02891-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
The primary issues in treating type 1 diabetes mellitus (T1DM) through the transplantation of healthy islets or islet β-cells are graft rejection and a lack of available donors. Currently, the majority of approaches use cell encapsulation technology and transplant replacement cells that can release insulin to address transplant rejection and donor shortages. However, existing encapsulation materials merely serve as carriers for islet cell growth. A new treatment approach for T1DM could be developed by creating a smart responsive material that encourages the formation of islet cell spheroids to replicate their 3D connections in vivo and controls the release of insulin aggregates. In this study, we used microfluidics to create thermally sensitive porous scaffolds made of poly(N-isopropyl acrylamide)/graphene oxide (PNIPAM/GO). The material was carefully shrunk under near-infrared light, enriched with mouse insulinoma pancreatic β cells (β-TC-6 cells), encapsulated, and cultivated to form 3D cell spheroids. The controlled contraction of the thermally responsive porous scaffold regulated insulin release from the spheroids, demonstrated using the glucose-stimulated insulin release assay (GSIS), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence assay. Eventually, implantation of the spheroids into C57BL/6 N diabetic mice enhanced the therapeutic effect, potentially offering a novel approach to the management of T1DM.
Collapse
Affiliation(s)
- Xueting Yao
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China
- Department of Laboratory Medicine, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, P. R. China
| | - Zehua Gong
- School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, P. R. China
| | - Wenyan Yin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Hanbing Li
- Department of Pharmaceutical Sciences, Institute of Pharmacology, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, P. R. China.
| | - Dennis Douroumis
- Centre for Research Innovation, CRI, University of Greenwich, Kent, ME4 4TB, UK
| | - Lijiang Huang
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
| | - Huaqiong Li
- Joint Research Centre on Medicine, The Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, Zhejiang, 315700, P. R. China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Joint Centre of Translational Medicine, Wenzhou Institute, University of Chinese Academy of Science, Wenzhou, Zhejiang, 325000, P. R. China.
| |
Collapse
|
7
|
Wang X, Guo Q, Liu Z, Wang Y, Cao C, Jin L, Li C, Xiao J, Zhao W. Alterations in the Gut Microbiota Composition in Obesity with and without Type 2 Diabetes: A Pilot Study. Diabetes Metab Syndr Obes 2024; 17:3965-3974. [PMID: 39469300 PMCID: PMC11514687 DOI: 10.2147/dmso.s477494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose Obesity has become a major public health concern worldwide, increasing the risk of T2DM. Growing evidence indicates gut microbiota dysbiosis is related to metabolic disorders. We aimed to firstly investigate the compositional and functional features of the gut microbiome between obesity with and without T2DM in the Chinese population. Methods A total of 32 obese individuals accompanied with T2DM and 18 age and gender-matched obesity with normal glucose tolerance (NGT) were enrolled. Fecal samples were collected, and the gut microbiota profile was determined using the Illumina MiSeq platform based on V3-V4 bacterial 16S rRNA gene. Results Compared with obesity- NGT, obesity-T2DM showed a significantly higher alpha diversity. Principal coordinates analysis based on both Bray-Curtis distance and weighted Unifrac revealed that the global microbial composition was significantly different between the two groups (P = 0.007 and P = 0.005, respectively). At the phylum level, Obesity-T2DM patients exhibited a significant decrease in Bacteroidetes, and a pronounced increase in Firmicutes. Regarding the genus level, Bacteroides and Escherichia-Shigella were found to increase considerably, while Prevotella_9 and Sutterella had an evident decrease in Obesity-T2DM. Furthermore, Spearman correlation analysis revealed that Prevotella_9 and Sutterella were negatively associated with HbA1c and fasting blood glucose. Conclusion We found clear differences in the gut microbiota composition in obesity-T2DM compared with obesity-NGT. Obesity accompanied with T2DM may aggravate the obesity-associated gut microbiota, and gut microbiota is expected to be a promising novel intervention target for obese management. However, larger sample size and more in-depth taxonomic identification studies are warranted.
Collapse
Affiliation(s)
- Xiaojing Wang
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Qinli Guo
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Zhaoxiang Liu
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Yanlei Wang
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Chenxiang Cao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Lixia Jin
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Caihong Li
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Jianzhong Xiao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| | - Wenhui Zhao
- Department of Endocrinology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Zhang T, Wang N, Liao Z, Chen J, Meng H, Lin H, Xu T, Chen L, Zhu LQ, Liu H. A differentiation protocol for generating pancreatic delta cells from human pluripotent stem cells. Front Cell Dev Biol 2024; 12:1490040. [PMID: 39493348 PMCID: PMC11527672 DOI: 10.3389/fcell.2024.1490040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
In this protocol, we detail a seven-stage differentiation methodology for generating pancreatic delta cells (SC-delta cells) from human pluripotent stem cells (hPSCs). In the first step, definitive endoderm is generated by activin A and CHIR99021, followed by induction of primitive gut tube and posterior foregut by treatment with FGF7, SANT1, LDN193189, PdBU, and retinoic acid (RA). The subsequent endocrine generation and directed SC-delta cell induction is achieved by a combined treatment of the FGF7 with FGF2 during stage 4 and 5, together with RA, XXI, ALK5 inhibitor II, SANT1, Betacellulin and LDN193189. The planar cultivation is converted to a suspended system after stage 5, allowing cells to aggregate into delta cell-containing spheroids. The differentiation takes approximately 4-5 weeks for delta cell generation and an additional 1-2 weeks for cell expansion and evaluation. We believe that this amenable and simplified protocol can provide a stable source of SC-delta cells from efficient differentiation, facilitating further investigation of the physiological role of delta cells as well as refinement of islet cell therapeutic strategies.
Collapse
Affiliation(s)
- Tongran Zhang
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Nannan Wang
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhiying Liao
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jingyi Chen
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| | - Hao Meng
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haopeng Lin
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
| | - Tao Xu
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lihua Chen
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huisheng Liu
- Department of Testing and Diagnosis Technology Research, Guangzhou National Laboratory, Guangzhou, Guangdong, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong, China
| |
Collapse
|
9
|
Du X, Zhu Y, Lu W, Fu N, Wang Q, Shi B. Regulation of the Function of T Follicular Helper Cells and B Cells in Type 1 Diabetes Mellitus by the OX40/OX40L Axis. J Clin Endocrinol Metab 2024; 109:2823-2830. [PMID: 38625053 PMCID: PMC11479688 DOI: 10.1210/clinem/dgae248] [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: 10/17/2023] [Revised: 03/14/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
OBJECTIVE/MAIN OUTCOME To study the expression of OX40 on T follicular helper (Tfh) cells and the ligand OX40L on antigen-presenting cells (APCs) in peripheral blood of patients with type 1 diabetes mellitus (T1DM) and the role of OX40 signaling in promoting Tfh cells to assist B-cell differentiation. DESIGN Cross-sectional study. SETTING Endocrinology department of a university hospital. PARTICIPANTS Twenty-five patients with T1DM and 35 with newly diagnosed type 2 diabetes mellitus (T2DM) from January 2021 to December 2021 (39 males, 21 females; mean age: 31.0 ± 4.5, range: 19-46 years). INTERVENTIONS None. METHODS The peripheral blood proportion of CD4+CD25-CD127+CXCR5+PD1+ Tfh cells in patients with T1DM or T2DM and the OX40L expression in CD14+ monocytes and CD19+ B cells were analyzed by flow cytometry. The OX40 signal effect on Tfh-cell function was analyzed by coincubating B cells with Tfh cells under different conditions. Flow cytometry detected the ratio of CD19-CD138+ plasmacytes. RESULTS The Tfh cells ratio and intracellular IL-21 expression in peripheral blood was significantly higher in patients with T1DM than with T2DM, and the OX40 expression in peripheral Tfh cells and OX40L expression in APC were significantly higher in T1DM. After adding OX40L protein, the CD19-CD138+-plasmacytes percentage was significantly increased and higher in T1DM. Blocking of anti-OX40L monoclonal antibodies significantly reduced the plasmacytes ratio. CONCLUSION The peripheral Tfh cells proportion increased and the OX40 expression in peripheral Tfh cells was upregulated in patients with T1DM vs patients with T2DM. OX40/OX40L signaling enhanced the Tfh-cell function to assist B-cell differentiation, which may contribute to the pathogenesis of T1DM.
Collapse
Affiliation(s)
- Xuan Du
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215129, Jiangsu, China
| | - Yan Zhu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215129, Jiangsu, China
| | - Wen Lu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215129, Jiangsu, China
| | - Nannan Fu
- Department of Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Qin Wang
- Department of Immunology, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Bimin Shi
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Soochow University, Suzhou 215129, Jiangsu, China
| |
Collapse
|
10
|
Li Z, Li A, Liu P, Zhang B, Yan Y. Mapping the evolution and impact of ketogenic diet research on diabetes management: a comprehensive bibliometric analysis from 2005 to 2024. Front Nutr 2024; 11:1485642. [PMID: 39483785 PMCID: PMC11527367 DOI: 10.3389/fnut.2024.1485642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 09/30/2024] [Indexed: 11/03/2024] Open
Abstract
Objective The ketogenic diet (KD) has been explored for diabetes management; however, a quantitative synthesis of its specific effects on diabetes has not yet been conducted. This study aims to examine the current status and research hotspots of KD in diabetes management from 2005 to 2024, providing a reference for future research. Methods We retrieved articles published between 2005 and 2024 from the Web of Science database and analyzed them using R software, VOSviewer, and CiteSpace. Results This study includes 432 relevant publications. From 2005 to 2024, the volume of literature in this field has shown a steady upward trend, with a notable increase from 2017 to 2021, and a slight decline observed from 2021 to 2023. The United States is the leading country in terms of the number of publications, followed by China, Australia, and Canada. The United States not only leads in publication volume but also maintains a broader international collaboration network. Nutrients and the American Journal of Clinical Nutrition are the most frequently published and cited journals. Current research hotspots primarily focus on the impact of KD on blood glucose control, insulin resistance, and lipid metabolism in diabetic patients. Mechanistic studies on KD in diabetes management concentrate on aspects such as the "regulation of genes by β-hydroxybutyrate," "anti-inflammatory effects," and "oxidative stress." The role of the gut microbiome is also emerging as an important research area. Currently, exploring the application of KD in managing different age groups and types of diabetes has become a significant research trend. Conclusion As an emerging dietary intervention, KD is gradually attracting widespread attention from researchers around the world and is expected to become a major research focus in the future for diabetes management and control. This paper provides a systematic review and analysis of the current research status and hotspots of KD in diabetes management, offering important references and insights for future research in related fields.
Collapse
Affiliation(s)
- Zonghuai Li
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Anxia Li
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, Hainan, China
| | - Pingping Liu
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, Hainan, China
| | - Bo Zhang
- Scientific Research Center, Guilin Medical University, Guilin, China
| | - Yuanyuan Yan
- Department of Pharmacy, Sanya Central Hospital (The Third People's Hospital of Hainan Province), Sanya, Hainan, China
| |
Collapse
|
11
|
Zhao L, Hu M, Li L. Identifying the Genetic Associations Between Diabetes Mellitus and the Risk of Vitiligo. Clin Cosmet Investig Dermatol 2024; 17:2261-2271. [PMID: 39421797 PMCID: PMC11484772 DOI: 10.2147/ccid.s480199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/08/2024] [Indexed: 10/19/2024]
Abstract
Purpose While increasing observational studies have suggested an association between diabetes mellitus (DM) and vitiligo, the causal relationship and possible mechanism remain unclear. Methods Publicly accessible genome-wide association study (GWAS) was utilized to conduct a bidirectional two-sample Mendelian randomization (MR) analysis. GWAS data for diabetes and vitiligo were obtained from the UK Biobank Consortium (20203 cases and 388756 controls) and the current GWAS data with largest cases (GCST004785, 4680 cases and 39586 controls) for preliminary analysis, respectively. Inverse variance weighting (IVW) was used as the main analysis method. Several sensitivity analyses were utilized to test the pleiotropy or heterogeneity. To explore the possible mechanism of gene-generating effects represented by the final instrumental variables in the analysis, enrichment analysis was conducted using the DAVID and STRING database. Results IVW method showed a significant genetic causal association between DM and vitiligo (OR = 1.20, 95% CI: 1.08-1.33, PIVW = 0.0009). Diabetes subtype analysis showed that T2D (type 2 diabetes) were associated with an increased risk of vitiligo (OR = 1.13, 95% CI: 1.00-1.27, PIVW = 0.0432). Sensitivity analysis further confirmed the robustness of the results. The enrichment analysis revealed that the genetic inducing effects of diabetes mellitus on vitiligo were primarily about pancreatic secretion and protein digestion and absorption pathway. Conclusion Our findings provide genetic evidence that there is a notable association between T2D and an elevated risk of vitiligo in European populations. This result may explain why the co-presentation of T2D and vitiligo is often seen in observational studies, and emphasize the significance of vigilant monitoring and clinical evaluations for vitiligo in individuals diagnosed with T2D. The aberrant glucose and lipid metabolism and the primary nutrient absorption disorder of vitiligo brought on by diabetes may be the potential mechanisms behind this association.
Collapse
Affiliation(s)
- Lingyun Zhao
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Meng Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Li Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Cosmetics Safety and Efficacy Evaluation Center, Key Laboratory of Human Evaluation and Big Data of Cosmetics, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
12
|
Wang YX, Pi JC, Yao YF, Peng XP, Li WJ, Xie MY. Hypoglycemic effects of white hyacinth bean polysaccharide on type 2 diabetes mellitus rats involvement with entero-insular axis and GLP-1 via metabolomics study. Int J Biol Macromol 2024; 281:136489. [PMID: 39393741 DOI: 10.1016/j.ijbiomac.2024.136489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/02/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024]
Abstract
The present study aimed to investigate the potential effects of white hyacinth bean polysaccharide (WHBP) against type 2 diabetic mellitus (T2DM) which was established by high-glucose/high-fat for 8 weeks, combined with a low-dose streptozotocin (STZ) injection. Our results showed that WHBP behaved the hypoglycemic effect by attenuating fasting blood glucose in vivo. WHBP-mediated anti-diabetic effects associated with the attenuation of insulin resistance and pancreatic impairment, as evidenced by the mitigation of pathological changes, inflammatory response and oxidative stress in the pancreas of T2DM rats. Meanwhile, gut protection was also shown during WHBP-mediated anti-diabetic effects, and glucagon-like peptide-1 (GLP-1), a mediator of the entero-insular axis, was observed to be elevated in both gut and pancreas of WHBP groups when compared to DM group, suggesting that hypoglycemic effects of WHBP were implicated in gut-pancreas interaction. Subsequently, untargeted metabolomics analysis performed by UPLC-QTOF/MS and showed that WHBP administration significantly adjusted the levels of 40 metabolites when compared to DM group. Further data concerning pathway analysis showed that WHBP administration significantly regulated the phenylalanine metabolism, tryptophan metabolism, arginine and proline, isoleucine metabolism, and glycerophospholipid metabolism in T2DM rats. Together, our results suggested that WHBP performed hypoglycemic effects and pancreatic protection linked to entero-insular axis involvement with GLP-1 and reversed metabolic disturbances in T2DM rats.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| | - Jin-Chan Pi
- College of Future Technology, Nanchang University, Nanchang 330031, China
| | - Yu-Fei Yao
- Department of Critical Care Medicine, The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China
| | - Xiao-Ping Peng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Juan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China.
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, China
| |
Collapse
|
13
|
Chen Y, Jiang Q, Xing X, Yuan T, Li P. Clinical research progress on β-cell dysfunction in T2DM development in the Chinese population. Rev Endocr Metab Disord 2024:10.1007/s11154-024-09914-9. [PMID: 39382753 DOI: 10.1007/s11154-024-09914-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/06/2024] [Indexed: 10/10/2024]
Abstract
The prevalence of type-2 diabetes mellitus (T2DM) has increased over 10-fold in the past 40 years in China, which now has the largest T2DM population in the world. Insulin resistance and β-cell dysfunction are the typical features of T2DM. Although both factors play a role, decreased β-cell function and β-cell mass are the predominant factors for progression to T2DM. Considering the differences between Chinese T2DM patients and those of other ethnicities, it is important to characterize β-cell dysfunction in Chinese patients during T2DM progression. Herein, we reviewed the studies on the relationships between β-cell function and T2DM progression in the Chinese population and discussed the differences among individuals of varying ethnicities. Meanwhile, we summarized the risk factors and current treatments of T2DM in Chinese individuals and discussed their impacts on β-cell function with the hope of identifying a better T2DM therapy.
Collapse
Affiliation(s)
- Yibing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Qian Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Xiaowei Xing
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China
| | - Tao Yuan
- Department of Endocrinology, Key Laboratory of Endocrinology of Ministry of Health, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Pingping Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Diabetes Research Center of Chinese Academy of Medical Sciences, Beijing, 100050, China.
- CAMS Key Laboratory of Molecular Mechanism and Target Discovery of Metabolic Disorder and Tumorigenesis, Beijing, 100050, China.
| |
Collapse
|
14
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024:S1550-4131(24)00370-X. [PMID: 39383866 DOI: 10.1016/j.cmet.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|
15
|
Rampazzo Morelli N, Préfontaine C, Pipella J, Thompson PJ. Secreted GDF15 maintains transcriptional responses during DNA damage-mediated senescence in human beta cells. Am J Physiol Endocrinol Metab 2024; 327:E552-E562. [PMID: 39196800 PMCID: PMC11482276 DOI: 10.1152/ajpendo.00257.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 08/30/2024]
Abstract
Type 1 diabetes (T1D) is a chronic metabolic disease resulting from an autoimmune destruction of pancreatic beta cells. Beta cells activate various stress responses during the development of T1D, including senescence, which involves cell cycle arrest, prosurvival signaling, and a proinflammatory secretome termed the senescence-associated secretory phenotype (SASP). We previously identified growth and differentiation factor 15 (GDF15) as a major SASP factor in human islets and human EndoC-βH5 beta cells in a model of DNA damage-mediated senescence that recapitulates features of senescent beta cells in T1D. Soluble GDF15 has been shown to exert protective effects on human and mouse beta cells during various forms of stress relevant to T1D; therefore, we hypothesized that secreted GDF15 may play a prosurvival role during DNA damage-mediated senescence in human beta cells. We found that elevated GDF15 secretion was associated with endogenous senescent beta cells in an islet preparation from a T1D donor, supporting the validity of our DNA damage model. Using antibody-based neutralization, we found that secreted endogenous GDF15 was not required for senescent human islet or EndoC cell viability. Rather, neutralization of GDF15 led to reduced expression of specific senescence-associated genes, including GDF15 itself and the prosurvival gene BCL2-like protein 1 (BCL2L1). Taken together, these data suggest that SASP factor GDF15 is not required to sustain senescent human islet viability, but it is required to maintain senescence-associated transcriptional responses.NEW & NOTEWORTHY Beta cell senescence is an emerging contributor to the pathogenesis of type 1 diabetes, but candidate therapeutic targets have not been identified in human beta cells. In this study, we examined the role of a secreted factor, GDF15, and found that although it is not required to maintain viability during senescence, it is required to fine-tune gene expression programs involved in the senescence response during DNA damage in human beta cells.
Collapse
Affiliation(s)
- Nayara Rampazzo Morelli
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Camille Préfontaine
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jasmine Pipella
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Peter J Thompson
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada
- Department of Physiology & Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Zhang F, Ning J, Chen C, Li B, Wei Y. Advances in the mechanisms of Gardenia jasminoides Ellis in improving diabetes and its complications. Fitoterapia 2024; 178:106140. [PMID: 39053745 DOI: 10.1016/j.fitote.2024.106140] [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: 05/13/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Gardenia jasminoides Ellis (Zhi-zi), which belongs to the Rubiaceae family, has been used mainly with its fry fruit for thousands of years, and it is an herb with the homology of medicine and food. In traditional Chinese medicine (TCM) theory, Zhi-zi can be used for "Quench Xiaoke", meaning for therapying diabetes in modern medicine. Based on numerous pharmacological studies, Gardenia jasminoides Ellis (Zhi-zi), and its ingredients, mainly including iridoid glycosides and carotenoids (crocins), possess potent antioxidant and anti-inflammatory properties, and can promote insulin secretion and sensitization, stimulate GLP-1 pathway activity, and protect islet β cells and the macro- and microvascular systems. These properties are the primary reasons why Zhi-zi and its ingredients are effective in reducing glucose levels, treating diabetes, and preventing its complications. This review aims to summarize the current situation and the advances of the studies on the mechanisms of Zhi-zi in improving diabetes and its complications, and it is expected to provide useful and systematic references for future research and clinical application of Zhi-zi and its active ingredients in the therapy of diabetes and complications.
Collapse
Affiliation(s)
- Fan Zhang
- The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Junhao Ning
- The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Chen Chen
- The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Boxia Li
- The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yuhui Wei
- The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
17
|
Elsayed AK, Aldous N, Alajez NM, Abdelalim EM. Identifying miRNA Signatures Associated with Pancreatic Islet Dysfunction in a FOXA2-Deficient iPSC Model. Stem Cell Rev Rep 2024; 20:1915-1931. [PMID: 38916841 PMCID: PMC11445299 DOI: 10.1007/s12015-024-10752-0] [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] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
The pathogenesis of diabetes involves complex changes in the expression profiles of mRNA and non-coding RNAs within pancreatic islet cells. Recent progress in induced pluripotent stem cell (iPSC) technology have allowed the modeling of diabetes-associated genes. Our recent study using FOXA2-deficient human iPSC models has highlighted an essential role for FOXA2 in the development of human pancreas. Here, we aimed to provide further insights on the role of microRNAs (miRNAs) by studying the miRNA-mRNA regulatory networks in iPSC-derived islets lacking the FOXA2 gene. Consistent with our previous findings, the absence of FOXA2 significantly downregulated the expression of islet hormones, INS, and GCG, alongside other key developmental genes in pancreatic islets. Concordantly, RNA-Seq analysis showed significant downregulation of genes related to pancreatic development and upregulation of genes associated with nervous system development and lipid metabolic pathways. Furthermore, the absence of FOXA2 in iPSC-derived pancreatic islets resulted in significant alterations in miRNA expression, with 61 miRNAs upregulated and 99 downregulated. The upregulated miRNAs targeted crucial genes involved in diabetes and pancreatic islet cell development. In contrary, the absence of FOXA2 in islets showed a network of downregulated miRNAs targeting genes related to nervous system development and lipid metabolism. These findings highlight the impact of FOXA2 absence on pancreatic islet development and suggesting intricate miRNA-mRNA regulatory networks affecting pancreatic islet cell development.
Collapse
Affiliation(s)
- Ahmed K Elsayed
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- Stem Cell Core, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Noura Aldous
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Nehad M Alajez
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
- Translational Cancer and Immunity Center (TCIC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar
| | - Essam M Abdelalim
- Laboratory of Pluripotent Stem Cell Disease Modeling, Translational Medicine Department, Research Branch, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar.
- Diabetes Research Center (DRC), Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), PO Box 34110, Doha, Qatar.
| |
Collapse
|
18
|
Ruiz-Esteves KN, Shank KR, Deutsch AJ, Gunturi A, Chamorro-Pareja N, Colling CA, Zubiri L, Perlman K, Ouyang T, Villani AC, Florez JC, Gusev A, Reynolds KL, Miller KK, Udler MS, Sise ME, Rengarajan M. Identification of Immune Checkpoint Inhibitor-Induced Diabetes. JAMA Oncol 2024; 10:1409-1416. [PMID: 39207773 PMCID: PMC11362970 DOI: 10.1001/jamaoncol.2024.3104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/15/2024] [Indexed: 09/04/2024]
Abstract
Importance Immune checkpoint inhibitors (ICIs) have revolutionized cancer care; however, accompanying immune-related adverse events (irAEs) confer substantial morbidity and occasional mortality. Life-threatening irAEs may require permanent cessation of ICI, even in patients with positive tumor response. Therefore, it is imperative to comprehensively define the spectrum of irAEs to aid individualized decision-making around the initiation of ICI therapy. Objective To define incidence, risk factors, and clinical spectrum of an irreversible and life-threatening irAE: ICI-induced diabetes. Design, Setting, and Participants This cohort study, conducted at an academic integrated health care system examined 14 328 adult patients treated with ICIs, including 64 patients who developed ICI-induced diabetes, from July 2010 to January 2022. The data were analyzed from 2022 to 2023. Cases of ICI-induced diabetes were manually confirmed; detailed clinical phenotyping was performed at diagnosis and 1-year follow-up. For 862 patients, genotyping data were available, and polygenic risk for type 1 diabetes was determined. Main Outcomes and Measures For ICI-induced diabetes cases and controls, demographic characteristics, comorbidities, tumor category, and ICI category were compared. Among ICI-induced diabetes cases, markers of glycemic physiology were examined at diagnosis and 1-year follow-up. For patients with available genotyping, a published type 1 diabetes polygenic score (T1D GRS2) was calculated. Results Of 14 328 participants, 6571 (45.9%) were women, and the median (range) age was 66 (8-106) years. The prevalence of ICI-induced diabetes among ICI-treated patients was 0.45% (64 of 14 328), with an incidence of 124.8 per 100 000 person-years. Preexisting type 2 diabetes (odds ratio [OR], 5.91; 95% CI, 3.34-10.45) and treatment with combination ICI (OR, 2.57; 95% CI, 1.44-4.59) were significant clinical risk factors of ICI-induced diabetes. T1D GRS2 was associated with ICI-induced diabetes risk, with an OR of 4.4 (95% CI, 1.8-10.5) for patients in the top decile of T1D GRS2, demonstrating a genetic association between spontaneous autoimmunity and irAEs. Patients with ICI-induced diabetes were in 3 distinct phenotypic categories based on autoantibodies and residual pancreatic function, with varying severity of initial presentation. Conclusions and Relevance The results of this analysis of 14 328 ICI-treated patients followed up from ICI initiation determined the incidence, risk factors and clinical spectrum of ICI-induced diabetes. Widespread implementation of this approach across organ-specific irAEs may enhance diagnosis and management of these conditions, and this becomes especially pertinent as ICI treatment rapidly expands to treat a wide spectrum of cancers and is used at earlier stages of treatment.
Collapse
Affiliation(s)
- Karina N. Ruiz-Esteves
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Kaitlyn R. Shank
- Department of Medicine, Massachusetts General Hospital and Department of Medicine, Brigham and Women’s Hospital, Boston
| | - Aaron J. Deutsch
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alekhya Gunturi
- Department of Medicine, Massachusetts General Hospital and Boston University School of Medicine, Boston
| | - Natalia Chamorro-Pareja
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Caitlin A. Colling
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Leyre Zubiri
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | | | - Tianqi Ouyang
- Department of Medicine, Massachusetts General Hospital, Boston
| | - Alexandra-Chloé Villani
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jose C. Florez
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Broad Institute, Cambridge, Massachusetts
- Division of Genetics, Brigham and Women’s Hospital and Harvard Medical School, Boston
| | - Kerry L. Reynolds
- Department of Medicine, Mass General Cancer Center, Massachusetts General Hospital, Boston
| | - Karen K. Miller
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Miriam S. Udler
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Michelle Rengarajan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Broad Institute of Massachusetts Institute of Technology and Harvard University, Boston
| |
Collapse
|
19
|
Huang P, Zhu Y, Qin J. Research advances in understanding crosstalk between organs and pancreatic β-cell dysfunction. Diabetes Obes Metab 2024; 26:4147-4164. [PMID: 39044309 DOI: 10.1111/dom.15787] [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: 06/18/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024]
Abstract
Obesity has increased dramatically worldwide. Being overweight or obese can lead to various conditions, including dyslipidaemia, hypertension, glucose intolerance and metabolic syndrome (MetS), which may further lead to type 2 diabetes mellitus (T2DM). Previous studies have identified a link between β-cell dysfunction and the severity of MetS, with multiple organs and tissues affected. Identifying the associations between pancreatic β-cell dysfunction and organs is critical. Research has focused on the interaction between the liver, gut and pancreatic β-cells. However, the mechanisms and related core targets are still not perfectly elucidated. The aims of this review were to summarize the mechanisms of β-cell dysfunction and to explore the potential pathogenic pathways and targets that connect the liver, gut, adipose tissue, muscle, and brain to pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunling Zhu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Jian Qin
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
20
|
Foran D, Antoniades C, Akoumianakis I. Emerging Roles for Sphingolipids in Cardiometabolic Disease: A Rational Therapeutic Target? Nutrients 2024; 16:3296. [PMID: 39408263 PMCID: PMC11478599 DOI: 10.3390/nu16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Cardiovascular disease is a leading cause of morbidity and mortality. New research elucidates increasingly complex relationships between cardiac and metabolic health, giving rise to new possible therapeutic targets. Sphingolipids are a heterogeneous class of bioactive lipids with critical roles in normal human physiology. They have also been shown to play both protective and deleterious roles in the pathogenesis of cardiovascular disease. Ceramides are implicated in dysregulating insulin signalling, vascular endothelial function, inflammation, oxidative stress, and lipoprotein aggregation, thereby promoting atherosclerosis and vascular disease. Ceramides also advance myocardial disease by enhancing pathological cardiac remodelling and cardiomyocyte death. Glucosylceramides similarly contribute to insulin resistance and vascular inflammation, thus playing a role in atherogenesis and cardiometabolic dysfunction. Sphingosing-1-phosphate, on the other hand, may ameliorate some of the pathological functions of ceramide by protecting endothelial barrier integrity and promoting cell survival. Sphingosine-1-phosphate is, however, implicated in the development of cardiac fibrosis. This review will explore the roles of sphingolipids in vascular, cardiac, and metabolic pathologies and will evaluate the therapeutic potential in targeting sphingolipids with the aim of prevention and reversal of cardiovascular disease in order to improve long-term cardiovascular outcomes.
Collapse
Affiliation(s)
| | | | - Ioannis Akoumianakis
- Cardiovascular Medicine Division, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; (D.F.); (C.A.)
| |
Collapse
|
21
|
Zhang H, Wei Y, Wang Y, Liang J, Hou Y, Nie X, Hou J. Emerging Diabetes Therapies: Regenerating Pancreatic β Cells. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39276101 DOI: 10.1089/ten.teb.2024.0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2024]
Abstract
The incidence of diabetes mellitus (DM) is steadily increasing annually, with 537 million diabetic patients as of 2021. Restoring diminished β cell mass or impaired islet function is crucial in treating DM, particularly type 1 DM. However, the regenerative capacity of islet β cells, which primarily produce insulin, is severely limited, and natural regeneration is only observed in young rodents or children. Hence, there is an urgent need to develop advanced therapeutic approaches that can regenerate endogenous β cells or replace them with stem cell (SC)-derived or engineered β-like cells. Current strategies for treating insulin-dependent DM mainly include promoting the self-replication of endogenous β cells, inducing SC differentiation, reprogramming non-β cells into β-like cells, and generating pancreatic-like organoids through cell-based intervention. In this Review, we discuss the current state of the art in these approaches, describe associated challenges, propose potential solutions, and highlight ongoing efforts to optimize β cell or islet transplantation and related clinical trials. These effective cell-based therapies will generate a sustainable source of functional β cells for transplantation and lay strong foundations for future curative treatments for DM.
Collapse
Affiliation(s)
- Haojie Zhang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yaxin Wei
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yubo Wang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Jialin Liang
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yifan Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Xiaobo Nie
- Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Urinary Surgery, Henan Provincial Research Center for the Prevention and Diagnosis of Prostate Diseases, Huaihe Hospital, Henan University, Kaifeng, China
| | - Junqing Hou
- Kaifeng 155 Hospital, China RongTong Medical Healthcare Group Co. Ltd., Kaifeng, China
| |
Collapse
|
22
|
Gao X, Qin X, Pei S, Wang W, Wu R, Mei J, Liu Y, Xie Y, Ni G. Multi-type maternal diabetes mellitus affects human placental villous geometric morphology: A three-dimensional imaging study. Placenta 2024; 155:70-77. [PMID: 39141963 DOI: 10.1016/j.placenta.2024.07.310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/18/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024]
Abstract
INTRODUCTION Diabetes mellitus leads to maldevelopment of the villous morphology in the human placenta, disrupting the exchange of materials between the maternal and fetal compartments, consequently compromising fetal development. This study aims to explore how different types of diabetes mellitus affect human placental villous geometric morphology including branching numbers and sizes (length, diameter). METHODS Here an optical coherence tomography (OCT)-based 3D imaging platform was utilized to capture 3D images of placental villi from different types of diabetes, including type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), and gestational diabetes mellitus (GDM). RESULTS Different types of diabetes mellitus exhibit different effects on human placental villous geometric morphological parameters: GDM had greater placenta villous parameters at intermediate villous diameter (IVD), terminal villous diameter (TVD), terminal villous length (TVL) compared to the healthy, T1DM, and T2DM, and these differences were statistically significant. The TVD of T1DM and T2DM had significantly greater sizes than the healthy. There was no statistically significant difference in the number of villous branches among the three types of diabetes, but T1DM and GDM had more villous branches than healthy individuals. DISCUSSION Diabetes mellitus affects the geometric morphology of human placental villi, with varying effects observed in pregnancies of different diabetes types. These findings offer a novel avenue for exploring underlying pathophysiological mechanisms and enhancing the management of women with diabetes from preconception through pregnancy.
Collapse
Affiliation(s)
- Xuemei Gao
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiaoyang Qin
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Shaoyu Pei
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Wenjing Wang
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Renxiong Wu
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Jie Mei
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yong Liu
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Yao Xie
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Guangming Ni
- School of Optoelectronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610054, China.
| |
Collapse
|
23
|
Hoffmann M, Poschenrieder J, Incudini M, Baier S, Fritz A, Maier A, Hartung M, Hoffmann C, Trummer N, Adamowicz K, Picciani M, Scheibling E, Harl M, Lesch I, Frey H, Kayser S, Wissenberg P, Schwartz L, Hafner L, Acharya A, Hackl L, Grabert G, Lee SG, Cho G, Cloward M, Jankowski J, Lee H, Tsoy O, Wenke N, Pedersen A, Bønnelykke K, Mandarino A, Melograna F, Schulz L, Climente-González H, Wilhelm M, Iapichino L, Wienbrandt L, Ellinghaus D, Van Steen K, Grossi M, Furth P, Hennighausen L, Di Pierro A, Baumbach J, Kacprowski T, List M, Blumenthal D. Network medicine-based epistasis detection in complex diseases: ready for quantum computing. Nucleic Acids Res 2024; 52:10144-10160. [PMID: 39175109 PMCID: PMC11417373 DOI: 10.1093/nar/gkae697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 07/12/2024] [Accepted: 08/01/2024] [Indexed: 08/24/2024] Open
Abstract
Most heritable diseases are polygenic. To comprehend the underlying genetic architecture, it is crucial to discover the clinically relevant epistatic interactions (EIs) between genomic single nucleotide polymorphisms (SNPs) (1-3). Existing statistical computational methods for EI detection are mostly limited to pairs of SNPs due to the combinatorial explosion of higher-order EIs. With NeEDL (network-based epistasis detection via local search), we leverage network medicine to inform the selection of EIs that are an order of magnitude more statistically significant compared to existing tools and consist, on average, of five SNPs. We further show that this computationally demanding task can be substantially accelerated once quantum computing hardware becomes available. We apply NeEDL to eight different diseases and discover genes (affected by EIs of SNPs) that are partly known to affect the disease, additionally, these results are reproducible across independent cohorts. EIs for these eight diseases can be interactively explored in the Epistasis Disease Atlas (https://epistasis-disease-atlas.com). In summary, NeEDL demonstrates the potential of seamlessly integrated quantum computing techniques to accelerate biomedical research. Our network medicine approach detects higher-order EIs with unprecedented statistical and biological evidence, yielding unique insights into polygenic diseases and providing a basis for the development of improved risk scores and combination therapies.
Collapse
Affiliation(s)
- Markus Hoffmann
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Julian M Poschenrieder
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Massimiliano Incudini
- Dipartimento di Informatica, Universit‘a di Verona, Strada le Grazie 15 - 34137 Verona, Italy
| | - Sylvie Baier
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Amelie Fritz
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, DTU, 2800 Kgs. Lyngby, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Maier
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Michael Hartung
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Christian Hoffmann
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Nico Trummer
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Klaudia Adamowicz
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Mario Picciani
- Computational Mass Spectrometry, Technical University of Munich, Freising, Germany
| | - Evelyn Scheibling
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Maximilian V Harl
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
| | - Ingmar Lesch
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Hunor Frey
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Simon Kayser
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Paul Wissenberg
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Leon Schwartz
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Leon Hafner
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
| | - Aakriti Acharya
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Lena Hackl
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Gordon Grabert
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), Technische Universität Braunschweig, Rebenring 56, 38106 Braunschweig, Germany
| | - Sung-Gwon Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Korea
| | - Gyuhyeok Cho
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, Korea
| | | | - Jakub Jankowski
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Hye Kyung Lee
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Olga Tsoy
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Nina Wenke
- Institute for Computational Systems Biology, University of Hamburg, Germany
| | - Anders Gorm Pedersen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, DTU, 2800 Kgs. Lyngby, Denmark
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Antonio Mandarino
- International Centre for Theory of Quantum Technologies, University of Gdańsk, 80-309 Gdańsk, Poland
| | - Federico Melograna
- BIO3 - Systems Genetics; GIGA-R Medical Genomics, University of Liège, Liège, Belgium
- BIO3 - Systems Medicine; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Laura Schulz
- Leibniz Supercomputing Centre of the Bavarian Academy of Sciences and Humanities (LRZ), Garching b. München, Germany
| | | | - Mathias Wilhelm
- Computational Mass Spectrometry, Technical University of Munich, Freising, Germany
- Munich Data Science Institute (MDSI), Technical University of Munich, Garching, Germany
| | - Luigi Iapichino
- Leibniz Supercomputing Centre of the Bavarian Academy of Sciences and Humanities (LRZ), Garching b. München, Germany
| | - Lars Wienbrandt
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian Albrechts University of Kiel, Kiel, Germany
| | - Kristel Van Steen
- BIO3 - Systems Genetics; GIGA-R Medical Genomics, University of Liège, Liège, Belgium
- BIO3 - Systems Medicine; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Michele Grossi
- European Organization for Nuclear Research (CERN), Geneva1211, Switzerland
| | - Priscilla A Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- Institute for Advanced Study (Lichtenbergstrasse 2 a) Technical University of Munich, D-85748 Garching, Germany
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Alessandra Di Pierro
- Dipartimento di Informatica, Universit‘a di Verona, Strada le Grazie 15 - 34137 Verona, Italy
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Germany
- Computational BioMedicine Lab, University of Southern Denmark, Denmark
| | - Tim Kacprowski
- Department of Health Sciences and Technology, Neuroscience Center Zürich (ZNZ), Swiss Federal Institute of Technology (ETH Zürich), Zürich 8092, Switzerland
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics, Technische Universität Braunschweig and Hannover Medical School, Rebenring 56, 38106 Braunschweig, Germany
| | - Markus List
- Data Science in Systems Biology, School of Life Sciences, Technical University of Munich, Freising, Germany
- Biomedical Network Science Lab, Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
24
|
Ma H, Xu J, Fang H, Su Y, Lu Y, Shu Y, Liu W, Li B, Cheng YY, Nie Y, Zhong Y, Song K. A capsule-based scaffold incorporating decellularized extracellular matrix and curcumin for islet beta cell therapy in type 1 diabetes mellitus. Biofabrication 2024; 16:045038. [PMID: 39255833 DOI: 10.1088/1758-5090/ad7907] [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: 08/04/2023] [Accepted: 09/10/2024] [Indexed: 09/12/2024]
Abstract
The transplantation of islet beta cells offers an alternative to heterotopic islet transplantation for treating type 1 diabetes mellitus (T1DM). However, the use of systemic immunosuppressive drugs in islet transplantation poses significant risks to the body. To address this issue, we constructed an encapsulated hybrid scaffold loaded with islet beta cells. This article focuses on the preparation of the encapsulated structure using 3D printing, which incorporates porcine pancreas decellularized extracellular matrix (dECM) to the core scaffold. The improved decellularization method successfully preserved a substantial proportion of protein (such as Collagen I and Laminins) architecture and glycosaminoglycans in the dECM hydrogel, while effectively removing most of the DNA. The inclusion of dECM enhanced the physical and chemical properties of the scaffold, resulting in a porosity of 83.62% ± 1.09% and a tensile stress of 1.85 ± 0.16 MPa. In teams of biological activity, dECM demonstrated enhanced proliferation, differentiation, and expression of transcription factors such as Ki67, PDX1, and NKX6.1, leading to improved insulin secretion function in MIN-6 pancreatic beta cells. In the glucose-stimulated insulin secretion experiment on day 21, the maximum insulin secretion from the encapsulated structure reached 1.96 ± 0.08 mIU ml-1, representing a 44% increase compared to the control group. Furthermore, conventional capsule scaffolds leaverage the compatibility of natural biomaterials with macrophages to mitigate immune rejection. Here, incorporating curcumin into the capsule scaffold significantly reduced the secretion of pro-inflammatory cytokine (IL-1β, IL-6, TNF-α, IFN-γ) secretion by RAW264.7 macrophages and T cells in T1DM mice. This approach protected pancreatic islet cells against immune cell infiltration mediated by inflammatory factors and prevented insulitis. Overall, the encapsulated scaffold developed in this study shows promise as a natural platform for clinical treatment of T1DM.
Collapse
Affiliation(s)
- Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Jie Xu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Huan Fang
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Ya Su
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Yueqi Lu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Yan Shu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Wang Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Bing Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology, Sydney, NSW 2007, Australia
| | - Yi Nie
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| | - Yiming Zhong
- Department of Hand and Foot Microsurgery, Dalian Municipal Central Hospital Affiliated of Dalian University of Technology, Dalian 116033, People's Republic of China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, People's Republic of China
- Zhengzhou Institute of Emerging Industrial Technology, Zhengzhou 450000, People's Republic of China
| |
Collapse
|
25
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
26
|
Hushmandi K, Einollahi B, Aow R, Suhairi SB, Klionsky DJ, Aref AR, Reiter RJ, Makvandi P, Rabiee N, Xu Y, Nabavi N, Saadat SH, Farahani N, Kumar AP. Investigating the interplay between mitophagy and diabetic neuropathy: Uncovering the hidden secrets of the disease pathology. Pharmacol Res 2024; 208:107394. [PMID: 39233055 DOI: 10.1016/j.phrs.2024.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitophagy, the cellular process of selectively eliminating damaged mitochondria, plays a crucial role in maintaining metabolic balance and preventing insulin resistance, both key factors in type 2 diabetes mellitus (T2DM) development. When mitophagy malfunctions in diabetic neuropathy, it triggers a cascade of metabolic disruptions, including reduced energy production, increased oxidative stress, and cell death, ultimately leading to various complications. Thus, targeting mitophagy to enhance the process may have emerged as a promising therapeutic strategy for T2DM and its complications. Notably, plant-derived compounds with β-cell protective and mitophagy-stimulating properties offer potential as novel therapeutic agents. This review highlights the intricate mechanisms linking mitophagy dysfunction to T2DM and its complications, particularly neuropathy, elucidating potential therapeutic interventions for this debilitating disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rachel Aow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suhana Binte Suhairi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Pooyan Makvandi
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India; University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
27
|
Fan J, Hu J. Retinol binding protein 4 and type 2 diabetes: from insulin resistance to pancreatic β-cell function. Endocrine 2024; 85:1020-1034. [PMID: 38520616 DOI: 10.1007/s12020-024-03777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/01/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND AND AIM Retinol binding protein 4 (RBP4) is an adipokine that has been explored as a key biomarker of type 2 diabetes mellitus (T2DM) in recent years. Researchers have conducted a series of experiments to understand the interplay between RBP4 and T2DM, including its role in insulin resistance and pancreatic β-cell function. The results of these studies indicate that RBP4 has a significant influence on T2DM and is considered a potential biomarker of T2DM. However, there have also been some controversies about the relationship between RBP4 levels and T2DM. In this review, we update and summarize recent studies focused on the relationship between RBP4 and T2DM and its role in insulin resistance and pancreatic β-cell function to clarify the existing controversy and provide evidence for future studies. We also assessed the potential therapeutic applications of RBP4 in treating T2DM. METHODS A narrative review. RESULTS Overall, there were significant associations between RBP4 levels, insulin resistance, pancreatic β-cell function, and T2DM. CONCLUSIONS More mechanistic studies are needed to determine the role of RBP4 in the onset of T2DM, especially in terms of pancreatic β-cell function. In addition, further studies are required to evaluate the effects of drug intervention, lifestyle intervention, and bariatric surgery on RBP4 levels to control T2DM and the role of reducing RBP4 levels in improving insulin sensitivity and pancreatic β-cell function.
Collapse
Affiliation(s)
- Jiahua Fan
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Clinical Nutrition, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, 510095, Guangdong, PR China.
| | - Jinxing Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Key Laboratory of Tuberculosis Research, Department of Tuberculosis, Guangzhou Chest Hospital, Institute of Tuberculosis, Guangzhou Medical University, Guangzhou, 510095, Guangdong, PR China
| |
Collapse
|
28
|
Kobyliak N, Khomenko M, Falalyeyeva T, Fedchenko A, Savchuk O, Tseyslyer Y, Ostapchenko L. Probiotics for pancreatic β-cell function: from possible mechanism of action to assessment of effectiveness. Crit Rev Microbiol 2024; 50:663-683. [PMID: 37705353 DOI: 10.1080/1040841x.2023.2257776] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 07/27/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Type 2 diabetes (T2D) is a metabolic disease characterized by chronic hyperglycemia because of insulin resistance (IR) and\or pancreatic β-cell dysfunction. Last century research showed that gut microbiota has a direct effect on metabolism and metabolic diseases. New studies into the human microbiome and its connection with the host is making it possible to develop new therapies for a wide variety of diseases. Inflammation is a well-known precursor to metabolic syndrome, which increases the risk of hypertension, visceral obesity, and dyslipidemia, which can lead to T2D through the damage of pancreatic β-cell and reduce insulin secretion. Current understanding for beneficial effects of probiotics in T2D strictly rely on both animal and clinical data, which mostly focused on their impact on IR, anthropometric parameters, glycemic control and markers of chronic systemic inflammation. From the other hand, there is a lack of evidence-based probiotic efficacy on pancreatic β-cell function in terms of T2D and related metabolic disorders. Therefore, current review will focus on the efficacy of probiotics for the protection of β-cells damage and it`s mechanism in patients with T2D.
Collapse
Affiliation(s)
- Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, Kyiv, Ukraine
- Medical Laboratory CSD, Kyiv, Ukraine
| | - Maria Khomenko
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Medical Laboratory CSD, Kyiv, Ukraine
- Taras Shevchenko National University of Kyiv, Kyiv, Ukraine
| | | | | | | | | |
Collapse
|
29
|
Umashankar B, Eliasson L, Ooi CY, Kim KW, Shaw JAM, Waters SA. Beyond insulin: Unraveling the complex interplay of ER stress, oxidative damage, and CFTR modulation in CFRD. J Cyst Fibros 2024; 23:842-852. [PMID: 38897882 DOI: 10.1016/j.jcf.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
CF-related diabetes (CFRD) is a prevalent comorbidity in people with Cystic Fibrosis (CF), significantly impacting morbidity and mortality rates. This review article critically evaluates the current understanding of CFRD molecular mechanisms, including the role of CFTR protein, oxidative stress, unfolded protein response (UPR) and intracellular communication. CFRD manifests from a complex interplay between exocrine pancreatic damage and intrinsic endocrine dysfunction, further complicated by the deleterious effects of misfolded CFTR protein on insulin secretion and action. Studies indicate that ER stress and subsequent UPR activation play critical roles in both exocrine and endocrine pancreatic cell dysfunction, contributing to β-cell loss and insulin insufficiency. Additionally, oxidative stress and altered calcium flux, exacerbated by CFTR dysfunction, impair β-cell survival and function, highlighting the significance of antioxidant pathways in CFRD pathogenesis. Emerging evidence underscores the importance of exosomal microRNAs (miRNAs) in mediating inflammatory and stress responses, offering novel insights into CFRD's molecular landscape. Despite insulin therapy remaining the cornerstone of CFRD management, the variability in response to CFTR modulators underscores the need for personalized treatment approaches. The review advocates for further research into non-CFTR therapeutic targets, emphasizing the need to address the multifaceted pathophysiology of CFRD. Understanding the intricate mechanisms underlying CFRD will pave the way for innovative treatments, moving beyond insulin therapy to target the disease's root causes and improve the quality of life for individuals with CF.
Collapse
Affiliation(s)
- Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Lena Eliasson
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Scania University Hospital, Malmö, Scania, Sweden
| | - Chee Y Ooi
- Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Department of Gastroenterology, Sydney Children's Hospital Randwick, NSW, Australia
| | - Ki Wook Kim
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Virology and Serology Division (SaViD), New South Wales Health Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - James A M Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
30
|
Strati M, Moustaki M, Psaltopoulou T, Vryonidou A, Paschou SA. Early onset type 2 diabetes mellitus: an update. Endocrine 2024; 85:965-978. [PMID: 38472622 DOI: 10.1007/s12020-024-03772-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/02/2024] [Indexed: 03/14/2024]
Abstract
The incidence and prevalence of type 2 diabetes mellitus (T2DM) in young individuals (aged <40 years) have significantly increased in recent years, approximating two to threefold increase in the respective rates. Numerous risk factors including severe obesity, family history, ethnicity, maternal diabetes or gestational diabetes, and female sex contribute to a younger age of onset. In terms of pathogenesis, impaired insulin secretion is the key operating mechanism, alongside with ectopic adiposity-related insulin resistance. T2DM diagnosis in a young adult requires the exclusion of type 1 diabetes mellitus (T1DM), latent autoimmune diabetes of adults (LADA) and maturity-onset diabetes of the young (MODY). The establishment of such diagnosis is critical for prognosis, because early-onset T2DM is associated with rapid deterioration in pancreatic β-cell secretory function leading to earlier initiation of insulin therapy. Furthermore, mortality and lifetime risk of developing complications, especially microvascular, is increased in these patients compared to both later-onset T2DM and T1DM patients; also, the latter are often developed earlier in the course of disease. The management of early-onset T2DM follows the same guidelines as in later-onset T2DM; yet patients aged 18-39 years are underrepresented in the big clinical trials on which the development of guidelines is based. Finally, young people with T2DM face significant challenges associated with social determinants, which compromise their adherence to therapy and induce diabetes distress. Future research focusing on the pathogenesis of β-cell decline and complications, as well as on specific treatment shall lead to better understanding and management of early-onset T2DM.
Collapse
Affiliation(s)
- Myrsini Strati
- School of Medicine, University of Patras, Patras, Greece
| | - Melpomeni Moustaki
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Theodora Psaltopoulou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes Center, Hellenic Red Cross Hospital, Athens, Greece
| | - Stavroula A Paschou
- Endocrine Unit and Diabetes Center, Department of Clinical Therapeutics, Alexandra Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
31
|
Li M, Wang Y, Wu X, Chen Q, Huang J, Zhu H, Yang S, Wang J, Li LT, Liu X, Fu K, Song F, Wang C. KIAA0753 enhances osteoblast differentiation suppressed by diabetes. J Cell Mol Med 2024; 28:e70035. [PMID: 39245790 PMCID: PMC11381189 DOI: 10.1111/jcmm.70035] [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: 03/22/2024] [Revised: 08/26/2024] [Accepted: 07/26/2024] [Indexed: 09/10/2024] Open
Abstract
Diabetes-related bone loss represents a significant complication that persistently jeopardizes the bone health of individuals with diabetes. Primary cilia proteins have been reported to play a vital role in regulating osteoblast differentiation in diabetes-related bone loss. However, the specific contribution of KIAA0753, a primary cilia protein, in bone loss induced by diabetes remains unclear. In this investigation, we elucidated the pivotal role of KIAA0753 as a promoter of osteoblast differentiation in diabetes. RNA sequencing demonstrated a marked downregulation of KIAA0753 expression in pro-bone MC3T3 cells exposed to a high glucose environment. Diabetes mouse models further validated the downregulation of KIAA0753 protein in the femur. Diabetes was observed to inhibit osteoblast differentiation in vitro, evidenced by downregulating the protein expression of OCN, OPN and ALP, decreasing primary cilia biosynthesis, and suppressing the Hedgehog signalling pathway. Knocking down KIAA0753 using shRNA methods was found to shorten primary cilia. Conversely, overexpression KIAA0753 rescued these changes. Additional insights indicated that KIAA0753 effectively restored osteoblast differentiation by directly interacting with SHH, OCN and Gli2, thereby activating the Hedgehog signalling pathway and mitigating the ubiquitination of Gli2 in diabetes. In summary, we report a negative regulatory relationship between KIAA0753 and diabetes-related bone loss. The clarification of KIAA0753's role offers valuable insights into the intricate mechanisms underlying diabetic bone complications.
Collapse
Affiliation(s)
- Mengxue Li
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yongqin Wang
- Department of Gastrointestinal Surgery, Traditional Chinese Medicine Hospital of Shizhu, Chongqing, China
| | - Xiangmei Wu
- Department of Physiology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Quanmei Chen
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Jianguo Huang
- A Division of Providence Cancer Institute, Earle A. Chiles Research Institute, Portland, Oregon, USA
| | - Huifang Zhu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Shengyong Yang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Jichun Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Le Tai Li
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Xianjun Liu
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Kang Fu
- Sangon Biotech (Shanghai) Co., Ltd., Shanghai, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Changdong Wang
- Department of Biochemistry and Molecular Biology, Molecular Medicine and Cancer Research Center, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| |
Collapse
|
32
|
Leungsuwan DS, Chandran M. Bone Fragility in Diabetes and its Management: A Narrative Review. Drugs 2024; 84:1111-1134. [PMID: 39103693 DOI: 10.1007/s40265-024-02078-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Bone fragility is a serious yet under-recognised complication of diabetes mellitus (DM) that is associated with significant morbidity and mortality. Multiple complex pathophysiological mechanisms mediating bone fragility amongst DM patients have been proposed and identified. Fracture risk in both type 1 diabetes (T1D) and type 2 diabetes (T2D) continues to be understated and underestimated by conventional risk assessment tools, posing an additional challenge to the identification of at-risk patients who may benefit from earlier intervention or preventive strategies. Over the years, an increasing body of evidence has demonstrated the efficacy of osteo-pharmacological agents in managing skeletal fragility in DM. This review seeks to elaborate on the risk of bone fragility in DM, the underlying pathogenesis and skeletal alterations, the approach to fracture risk assessment in DM, management strategies and therapeutic options.
Collapse
Affiliation(s)
| | - Manju Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, 20 College Road, ACADEMIA, Singapore, 169856, Singapore.
- DUKE NUS Medical School, Singapore, Singapore.
| |
Collapse
|
33
|
Li Y, Miao Y, Feng Q, Zhu W, Chen Y, Kang Q, Wang Z, Lu F, Zhang Q. Mitochondrial dysfunction and onset of type 2 diabetes along with its complications: a multi-omics Mendelian randomization and colocalization study. Front Endocrinol (Lausanne) 2024; 15:1401531. [PMID: 39280009 PMCID: PMC11392782 DOI: 10.3389/fendo.2024.1401531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024] Open
Abstract
Background Mitochondrial dysfunction plays a crucial role in Type 2 Diabetes Mellitus (T2DM) and its complications. However, the genetic pathophysiology remains under investigation. Through multi-omics Mendelian Randomization (MR) and colocalization analyses, we identified mitochondrial-related genes causally linked with T2DM and its complications. Methods Summary-level quantitative trait loci data at methylation, RNA, and protein levels were retrieved from European cohort studies. GWAS summary statistics for T2DM and its complications were collected from the DIAGRAM and FinnGen consortiums, respectively. Summary-data-based MR was utilized to estimate the causal effects. The heterogeneity in dependent instrument test assessed horizontal pleiotropy, while colocalization analysis determined whether genes and diseases share the same causal variant. Enrichment analysis, drug target analysis, and phenome-wide MR were conducted to further explore the biological functions, potential drugs, and causal associations with other diseases. Results Integrating evidence from multi-omics, we identified 18 causal mitochondrial-related genes. Enrichment analysis revealed they were not only related to nutrient metabolisms but also to the processes like mitophagy, autophagy, and apoptosis. Among these genes, Tu translation elongation factor mitochondrial (TUFM), 3-hydroxyisobutyryl-CoA hydrolase (HIBCH), and iron-sulfur cluster assembly 2 (ISCA2) were identified as Tier 1 genes, showing causal links with T2DM and strong colocalization evidence. TUFM and ISCA2 were causally associated with an increased risk of T2DM, while HIBCH showed an inverse causal relationship. The causal associations and colocalization effects for TUFM and HIBCH were validated in specific tissues. TUFM was also found to be a risk factor for microvascular complications in T2DM patients including retinopathy, nephropathy, and neuropathy. Furthermore, drug target analysis and phenome-wide MR underscored their significance as potential therapeutic targets. Conclusions This study identified 18 mitochondrial-related genes causally associated with T2DM at multi-omics levels, enhancing the understanding of mitochondrial dysfunction in T2DM and its complications. TUFM, HIBCH, and ISCA2 emerge as potential therapeutic targets for T2DM and its complications.
Collapse
Affiliation(s)
- Yang Li
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yahu Miao
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qing Feng
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Weixi Zhu
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yijing Chen
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qingqing Kang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhen Wang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangting Lu
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiu Zhang
- Department of Endocrinology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
34
|
Elisha C, Bhagwat P, Pillai S. Emerging production techniques and potential health promoting properties of plant and animal protein-derived bioactive peptides. Crit Rev Food Sci Nutr 2024:1-30. [PMID: 39206881 DOI: 10.1080/10408398.2024.2396067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Bioactive peptides (BPs) are short amino acid sequences that that are known to exhibit physiological characteristics such as antioxidant, antimicrobial, antihypertensive and antidiabetic properties, suggesting that they could be exploited as functional foods in the nutraceutical industry. These BPs can be derived from a variety of food sources, including milk, meat, marine, and plant proteins. In the past decade, various methods including in silico, in vitro, and in vivo techniques have been explored to unravel underlying mechanisms of BPs. To forecast interactions between peptides and their targets, in silico methods such as BIOPEP, molecular docking and Quantitative Structure-Activity Relationship modeling have been employed. Additionally, in vitro research has examined how BPs affect enzyme activities, protein expressions, and cell cultures. In vivo studies on the contrary have appraised the impact of BPs on animal models and human subjects. Hence, in the light of recent literature, this review examines the multifaceted aspects of BPs production from milk, meat, marine, and plant proteins and their potential bioactivities. We envisage that the various concepts discussed will contribute to a better understanding of the food derived BP production, which could pave a way for their potential applications in the nutraceutical industry.
Collapse
Affiliation(s)
- Cherise Elisha
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Prashant Bhagwat
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - Santhosh Pillai
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| |
Collapse
|
35
|
Jin J, Xia X, Ruan C, Luo Z, Yang Y, Wang D, Qin Y, Li D, Zhang Y, Hu Y, Lei P. GAPDH-Silence Microsphere via Reprogramming Macrophage Metabolism and eradicating Bacteria for Diabetic infection bone regeneration. J Nanobiotechnology 2024; 22:517. [PMID: 39210435 PMCID: PMC11361104 DOI: 10.1186/s12951-024-02787-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024] Open
Abstract
Macrophage metabolism dysregulation, which is exacerbated by persistent stimulation in infectious and inflammatory diseases, such as diabetic infectious bone defects (DIBD), eventually leads to the failure of bone repair. Here, we have developed an injectable, macrophage-modulated GAPDH-Silence drug delivery system. This microsphere comprises chondroitin sulfate methacrylate (CM) and methacrylated gelatin (GM), while the dimethyl fumarate (DMF)-loaded liposome (D-lip) is encapsulated within the microsphere (CM@GM), named D-lip/CM@GM. Triggered by the over-expressed collagenase in DIBD, the microspheres degrade and release the encapsulated D-lip. D-lip could modulate metabolism by inhibiting GAPDH, which suppresses the over-activation of glycolysis, thus preventing the inflammatory response of macrophages in vitro. While beneficial for macrophages, D-lip/CM@GM is harmful to bacteria. GAPDH, while crucial for glycolysis of staphylococcal species (S. aureus), can be effectively countered by D-lip/CM@GM. We are utilizing existing drugs in innovative ways to target central metabolism for effective eradication of bacteria. In the DIBD model, our results confirmed that the D-lip/CM@GM enhanced bacteria clearance and reprogrammed dysregulated metabolism, thereby significantly improving bone regeneration. In conclusion, this GAPDH-Silence microsphere system may provide a viable strategy to promote diabetic infection bone regeneration.
Collapse
Affiliation(s)
- Jiale Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xiaowei Xia
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China
| | - Chengxin Ruan
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhiyuan Luo
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yiqi Yang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Dongyu Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yifang Qin
- Department of Endocrinology, The Children's Hospital, School of Medicine, Zhejiang University, Hangzhou, 310052, China
| | - Dongdong Li
- Department of Orthopedic Surgery, Ningxia Medical University, Yinchuan, 200233, China
| | - Yong Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215006, China.
| | - Yihe Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Pengfei Lei
- Department of Orthopedic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
36
|
Damarov IS, Korbolina EE, Rykova EY, Merkulova TI. Multi-Omics Analysis Revealed the rSNPs Potentially Involved in T2DM Pathogenic Mechanism and Metformin Response. Int J Mol Sci 2024; 25:9297. [PMID: 39273245 PMCID: PMC11394919 DOI: 10.3390/ijms25179297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
The goal of our study was to identify and assess the functionally significant SNPs with potentially important roles in the development of type 2 diabetes mellitus (T2DM) and/or their effect on individual response to antihyperglycemic medication with metformin. We applied a bioinformatics approach to identify the regulatory SNPs (rSNPs) associated with allele-asymmetric binding and expression events in our paired ChIP-seq and RNA-seq data for peripheral blood mononuclear cells (PBMCs) of nine healthy individuals. The rSNP outcomes were analyzed using public data from the GWAS (Genome-Wide Association Studies) and Genotype-Tissue Expression (GTEx). The differentially expressed genes (DEGs) between healthy and T2DM individuals (GSE221521), including metformin responders and non-responders (GSE153315), were searched for in GEO RNA-seq data. The DEGs harboring rSNPs were analyzed using the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). We identified 14,796 rSNPs in the promoters of 5132 genes of human PBMCs. We found 4280 rSNPs to associate with both phenotypic traits (GWAS) and expression quantitative trait loci (eQTLs) from GTEx. Between T2DM patients and controls, 3810 rSNPs were detected in the promoters of 1284 DEGs. Based on the protein-protein interaction (PPI) network, we identified 31 upregulated hub genes, including the genes involved in inflammation, obesity, and insulin resistance. The top-ranked 10 enriched KEGG pathways for these hubs included insulin, AMPK, and FoxO signaling pathways. Between metformin responders and non-responders, 367 rSNPs were found in the promoters of 131 DEGs. Genes encoding transcription factors and transcription regulators were the most widely represented group and many were shown to be involved in the T2DM pathogenesis. We have formed a list of human rSNPs that add functional interpretation to the T2DM-association signals identified in GWAS. The results suggest candidate causal regulatory variants for T2DM, with strong enrichment in the pathways related to glucose metabolism, inflammation, and the effects of metformin.
Collapse
Affiliation(s)
- Igor S Damarov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena E Korbolina
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Elena Y Rykova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Department of Engineering Problems of Ecology, Novosibirsk State Technical University, 630087 Novosibirsk, Russia
| | - Tatiana I Merkulova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
37
|
Ramos-Rodríguez M, Subirana-Granés M, Norris R, Sordi V, Fernández Á, Fuentes-Páez G, Pérez-González B, Berenguer Balaguer C, Raurell-Vila H, Chowdhury M, Corripio R, Partelli S, López-Bigas N, Pellegrini S, Montanya E, Nacher M, Falconi M, Layer R, Rovira M, González-Pérez A, Piemonti L, Pasquali L. Implications of noncoding regulatory functions in the development of insulinomas. CELL GENOMICS 2024; 4:100604. [PMID: 38959898 PMCID: PMC11406191 DOI: 10.1016/j.xgen.2024.100604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/22/2024] [Accepted: 06/11/2024] [Indexed: 07/05/2024]
Abstract
Insulinomas are rare neuroendocrine tumors arising from pancreatic β cells, characterized by aberrant proliferation and altered insulin secretion, leading to glucose homeostasis failure. With the aim of uncovering the role of noncoding regulatory regions and their aberrations in the development of these tumors, we coupled epigenetic and transcriptome profiling with whole-genome sequencing. As a result, we unraveled somatic mutations associated with changes in regulatory functions. Critically, these regions impact insulin secretion, tumor development, and epigenetic modifying genes, including polycomb complex components. Chromatin remodeling is apparent in insulinoma-selective domains shared across patients, containing a specific set of regulatory sequences dominated by the SOX17 binding motif. Moreover, many of these regions are H3K27me3 repressed in β cells, suggesting that tumoral transition involves derepression of polycomb-targeted domains. Our work provides a compendium of aberrant cis-regulatory elements affecting the function and fate of β cells in their progression to insulinomas and a framework to identify coding and noncoding driver mutations.
Collapse
Affiliation(s)
- Mireia Ramos-Rodríguez
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Marc Subirana-Granés
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Richard Norris
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Valeria Sordi
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ángel Fernández
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain; Department of Physiological Science, School of Medicine, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain; Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Barcelona, Spain
| | - Georgina Fuentes-Páez
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Beatriz Pérez-González
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Clara Berenguer Balaguer
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Helena Raurell-Vila
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Murad Chowdhury
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Raquel Corripio
- Paediatric Endocrinology Department, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí I3PT, Universitat Autònoma de Barcelona, Sabadell, Spain
| | - Stefano Partelli
- Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital and University Vita-Salute, Milan, Italy
| | - Núria López-Bigas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Research Program on Biomedical Informatics, Universitat Pompeu Fabra, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Silvia Pellegrini
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eduard Montanya
- Bellvitge Hospital-IDIBELL, Barcelona, Spain; Department of Clinical Sciences, University of Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Montserrat Nacher
- Bellvitge Hospital-IDIBELL, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
| | - Massimo Falconi
- Pancreas Translational & Research Institute, Scientific Institute San Raffaele Hospital and University Vita-Salute, Milan, Italy
| | - Ryan Layer
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA; Department of Computer Science, University of Colorado Boulder, Boulder, CO, USA
| | - Meritxell Rovira
- Department of Physiological Science, School of Medicine, Universitat de Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain; Pancreas Regeneration: Pancreatic Progenitors and Their Niche Group, Regenerative Medicine Program, Institut d'Investigació Biomèdica de Bellvitge - IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], L'Hospitalet de Llobregat, Barcelona, Spain
| | - Abel González-Pérez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Research Program on Biomedical Informatics, Universitat Pompeu Fabra, Barcelona, Spain
| | - Lorenzo Piemonti
- Diabetes Research Institute (DRI) - IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Pasquali
- Endocrine Regulatory Genomics, Department of Medicine and Life Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain.
| |
Collapse
|
38
|
Wu J, Chen M, Xiao Y, Yang H, Wang G, Zhang X, Dai L, Yuan Z. The Bioactive Interface of Titanium Implant with Both Anti-Oxidative Stress and Immunomodulatory Properties for Enhancing Osseointegration under Diabetic Condition. Adv Healthc Mater 2024:e2401974. [PMID: 39132780 DOI: 10.1002/adhm.202401974] [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: 05/29/2024] [Revised: 07/29/2024] [Indexed: 08/13/2024]
Abstract
The poor implant-osseointegration under diabetic condition remains a challenge to be addressed urgently. Studies have confirmed that the diabetic pathological microenvironment is accompanied by excessive oxidative stress, imbalanced immune homeostasis, and persistent chronic inflammation, which seriously impairs the osteogenic process. Herein, a multifunctional bioactive interface with both anti-oxidative stress and immunomodulatory properties is constructed on titanium implants. Briefly, manganese dioxide nanosheets are coated onto mesoporous polydopamine nanoparticles loaded with carbon monoxide gas precursor, namely MnO2-CO@MPDA NPs, and then they are integrated on the titanium implant to obtain MCM-Ti. In the simulated diabetic microenvironment, under the action of MnO2 nanoenzymes, MCM-Ti can effectively eliminate intracellular reactive oxygen species while alleviating hypoxic state. Interestingly, the microenvironment mediates the responsive release of CO gas, which effectively drives macrophages toward M2 polarization, thereby ameliorating inflammatory response. The potential mechanism is that CO gas up-regulates the expression of heme oxygenase-1, further activating the Notch/Hes1/Stat3 signaling pathway. Furthermore, the conditioned medium derived from macrophages on MCM-Ti surface significantly enhances the osteogenic differentiation of BMSCs. In a type 2 diabetic rat model, MCM-Ti implant effectively alleviates the accompanying inflammation and enhances the osseointegration through the synergistic effects of resisting oxidative stress and remodeling immune homeostasis.
Collapse
Affiliation(s)
- Jianshuang Wu
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Sanhang Science & Technology Building No. 45th, Gaoxin South 9th Road, Nanshan District, Shenzhen, Guangdong, 518063, P. R. China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Maowen Chen
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, 81 Meishan Road, Hefei, Anhui, 230012, P. R. China
| | - Yao Xiao
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Gaoyang Wang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Xiaohong Zhang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Liangliang Dai
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
| | - Zhang Yuan
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Sanhang Science & Technology Building No. 45th, Gaoxin South 9th Road, Nanshan District, Shenzhen, Guangdong, 518063, P. R. China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi, 710072, P. R. China
- Northwestern Polytechnical University Chongqing Technology Innovation Center, 36 Pufu Road, Chongqing, 400000, P. R. China
| |
Collapse
|
39
|
Mummey HM, Elison W, Korgaonkar K, Elgamal RM, Kudtarkar P, Griffin E, Benaglio P, Miller M, Jha A, Fox JEM, McCarthy MI, Preissl S, Gloyn AL, MacDonald PE, Gaulton KJ. Single cell multiome profiling of pancreatic islets reveals physiological changes in cell type-specific regulation associated with diabetes risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.03.606460. [PMID: 39149326 PMCID: PMC11326183 DOI: 10.1101/2024.08.03.606460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Physiological variability in pancreatic cell type gene regulation and the impact on diabetes risk is poorly understood. In this study we mapped gene regulation in pancreatic cell types using single cell multiomic (joint RNA-seq and ATAC-seq) profiling in 28 non-diabetic donors in combination with single cell data from 35 non-diabetic donors in the Human Pancreas Analysis Program. We identified widespread associations with age, sex, BMI, and HbA1c, where gene regulatory responses were highly cell type- and phenotype-specific. In beta cells, donor age associated with hypoxia, apoptosis, unfolded protein response, and external signal-dependent transcriptional regulators, while HbA1c associated with inflammatory responses and gender with chromatin organization. We identified 10.8K loci where genetic variants were QTLs for cis regulatory element (cRE) accessibility, including 20% with lineage- or cell type-specific effects which disrupted distinct transcription factor motifs. Type 2 diabetes and glycemic trait associated variants were enriched in both phenotype- and QTL-associated beta cell cREs, whereas type 1 diabetes showed limited enrichment. Variants at 226 diabetes and glycemic trait loci were QTLs in beta and other cell types, including 40 that were statistically colocalized, and annotating target genes of colocalized QTLs revealed genes with putatively novel roles in disease. Our findings reveal diverse responses of pancreatic cell types to phenotype and genotype in physiology, and identify pathways, networks, and genes through which physiology impacts diabetes risk.
Collapse
Affiliation(s)
- Hannah M Mummey
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla CA
| | - Weston Elison
- Biomedical Sciences Program, University of California San Diego, La Jolla CA, USA
| | - Katha Korgaonkar
- Department of Pediatrics, University of California San Diego, La Jolla CA, USA
| | - Ruth M Elgamal
- Biomedical Sciences Program, University of California San Diego, La Jolla CA, USA
| | - Parul Kudtarkar
- Department of Pediatrics, University of California San Diego, La Jolla CA, USA
| | - Emily Griffin
- Department of Pediatrics, University of California San Diego, La Jolla CA, USA
| | - Paola Benaglio
- Department of Pediatrics, University of California San Diego, La Jolla CA, USA
| | - Michael Miller
- Center for Epigenomics, University of California San Diego, La Jolla CA, USA
| | - Alokkumar Jha
- Department of Pediatrics, Stanford School of Medicine, Stanford University, Stanford CA, USA
| | - Jocelyn E Manning Fox
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Mark I McCarthy
- Wellcome Trust Center for Human Genetics, University of Oxford, Oxford, UK*
| | - Sebastian Preissl
- Center for Epigenomics, University of California San Diego, La Jolla CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford CA, USA
| | - Anna L Gloyn
- Department of Pediatrics, Stanford School of Medicine, Stanford University, Stanford CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford University, Stanford CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
| | - Patrick E MacDonald
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kyle J Gaulton
- Department of Pediatrics, University of California San Diego, La Jolla CA, USA
| |
Collapse
|
40
|
Jin Y, Wan K, Liu C, Cheng W, Wang R. Mechanisms of exercise intervention in type 2 diabetes: a bibliometric and visualization analysis based on CiteSpace. Front Endocrinol (Lausanne) 2024; 15:1401342. [PMID: 39149117 PMCID: PMC11324446 DOI: 10.3389/fendo.2024.1401342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/16/2024] [Indexed: 08/17/2024] Open
Abstract
Objective Type 2 diabetes (T2D) is a common chronic metabolic disease, and its prevalence is increasing globally. Exercise is crucial for T2D management, yet many aspects of its mechanisms remain unclear. This study employs CiteSpace to reveal research hotspots and frontier issues in exercise intervention for T2D. Method A literature review spanning from January 1, 2013 to December 31, 2022, was conducted using the Web of Science Core Collection (WoSCC), with keywords including "exercise," "type 2 diabetes," and "mechanisms." We analyzed network diagrams generated by CiteSpace, which depicted relationships among countries, authors, and keywords. Results This study includes 1,210 English papers from 555 journals, affiliated with 348 institutions across 80 countries/regions. Notably, the United States, China, and the United Kingdom account for nearly half of all publications. The University of Copenhagen leads in publication volume, followed by Harvard Medical School and the University of Colorado. Key authors include Kirwan, John P (Case Western Reserve University), Malin, Steven K (Rutgers University), and Pedersen, Bente Klarlund (University of Copenhagen). Based on co-occurrence analysis of keywords, it is evident that terms such as "disease," "glucagon-like peptide 1," and "cardiovascular risk factor" exhibit high intermediary centrality. Conclusion The analysis highlights ongoing investigations into molecular mechanisms, such as β-cell function enhancement, exerkines, and epigenetic mechanisms. Emerging areas include exercise response heterogeneity, circadian rhythm regulation, transcription factors, neurotrophic factors, and mitochondrial function. Future studies should prioritize understanding interactions between different exercise mechanisms and optimizing exercise prescriptions for T2D. Exercise prescriptions are crucial for effective interventions. Collaboration between countries and institutions is essential to understand the influences of different genetic backgrounds and environmental factors. Currently, a combination of aerobic and resistance training is considered the optimal form of exercise. However, considering time efficiency, high-intensity interval training (HIIT) has gained widespread attention and research due to its ability to achieve similar exercise effects in a shorter duration. Additionally, circadian rhythm regulation may affect the exercise outcomes of diabetic individuals at different times of the day, particularly concerning the specific types, doses, and intensities used for precision intervention in T2D.
Collapse
Affiliation(s)
- Yue Jin
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Kang Wan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Physical Education College, Henan Sport University, Zhengzhou, China
| | - Cheng Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Wei Cheng
- Department of Endocrinology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
41
|
Bresson SE, Ruzzin J. Persistent organic pollutants disrupt the oxidant/antioxidant balance of INS-1E pancreatic β-cells causing their physiological dysfunctions. ENVIRONMENT INTERNATIONAL 2024; 190:108821. [PMID: 38885551 DOI: 10.1016/j.envint.2024.108821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Persistent organic pollutants (POPs) have emerged as potent diabetogenic agents, but their mechanisms of action remain poorly identified. OBJECTIVES In this study, we aim to determine the mechanisms regulating the damaging effects of POPs in pancreatic β-cells, which have a central role in the development of diabetes. METHODS We treated INS-1E pancreatic β-cells with PCB-153, p,p'-DDE, PCB-126, or TCDD at doses ranging from 1 × 10-15to 5 × 10-6M. We measured insulin content and secretion, cell viability and assessed the mRNA expression of the xenobiotic nuclear receptors Nr1i2 and Nr1i3, and the aryl hydrocarbon receptor (Ahr). In addition, we evaluated the antioxidant defense and production of reactive oxygen species (ROS). Finally, we studied the ability of the antioxidant N-acetyl-L-cysteine (NAC) to counteract the effects of POPs in INS-1E cells. RESULTS When exposed to environmental POP levels, INS-1E cells had impaired production and secretion of insulin. These defects were observed for all tested POPs and were paralleled by reduced Ins1 and Ins2 mRNA expression. While POP treatment for 3 days did not affect INS-1E cell viability, longer treatment progressively killed the cells. Furthermore, we found that the xenobiotic detoxification machinery is poorly expressed in the INS-1E cells, as characterized by the absence of Nr1i2 and Nr1i3 and their respective downstream targets Cyp3a1/Cyp3a2 and Cyp2b1/Cyp2b3, and the weak functionality of the Ahr/Cyp1a1 signaling. Interestingly, POPs dysregulated key antioxidant enzymes such as glutathione peroxidases, peroxiredoxins, thioredoxins, and catalases. In parallel, the production of intracellular ROS, including superoxide anion (O2•-) and hydrogen peroxide (H2O2), was increased by POP exposure. Improving the oxidant scavenging capacity of INS-1E cells by NAC treatment restored the production and secretion of insulin. CONCLUSION By promoting oxidative stress and impairing the ability of INS-1E cells to produce and secrete insulin, this study reveals how POPs can mechanistically act as diabetogenic agents, and provides new scientific evidence supporting the concept that POPs are fueling the diabetes epidemics.
Collapse
Affiliation(s)
- Sophie Emilie Bresson
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jérôme Ruzzin
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
42
|
Mathisen AF, Legøy TA, Larsen U, Unger L, Abadpour S, Paulo JA, Scholz H, Ghila L, Chera S. The age-dependent regulation of pancreatic islet landscape is fueled by a HNF1a-immune signaling loop. Mech Ageing Dev 2024; 220:111951. [PMID: 38825059 DOI: 10.1016/j.mad.2024.111951] [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: 01/09/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/04/2024]
Abstract
Animal longevity is a function of global vital organ functionality and, consequently, a complex polygenic trait. Yet, monogenic regulators controlling overall or organ-specific ageing exist, owing their conservation to their function in growth and development. Here, by using pathway analysis combined with wet-biology methods on several dynamic timelines, we identified Hnf1a as a novel master regulator of the maturation and ageing in the adult pancreatic islet during the first year of life. Conditional transgenic mice bearing suboptimal levels of this transcription factor in the pancreatic islets displayed age-dependent changes, with a profile echoing precocious maturation. Additionally, the comparative pathway analysis revealed a link between Hnf1a age-dependent regulation and immune signaling, which was confirmed in the ageing timeline of an overly immunodeficient mouse model. Last, the global proteome analysis of human islets spanning three decades of life largely backed the age-specific regulation observed in mice. Collectively, our results suggest a novel role of Hnf1a as a monogenic regulator of the maturation and ageing process in the pancreatic islet via a direct or indirect regulatory loop with immune signaling.
Collapse
Affiliation(s)
- Andreas Frøslev Mathisen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Thomas Aga Legøy
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ulrik Larsen
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Lucas Unger
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Shadab Abadpour
- Hybrid Technology Hub-Centre of Excellence, Faculty of Medicine, University of Oslo, Norway; Institute for Surgical Research, Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Hanne Scholz
- Hybrid Technology Hub-Centre of Excellence, Faculty of Medicine, University of Oslo, Norway; Institute for Surgical Research, Department of Transplant Medicine, Oslo University Hospital, Oslo, Norway
| | - Luiza Ghila
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simona Chera
- Mohn Research Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
43
|
Niu F, Liu W, Ren Y, Tian Y, Shi W, Li M, Li Y, Xiong Y, Qian L. β-cell neogenesis: A rising star to rescue diabetes mellitus. J Adv Res 2024; 62:71-89. [PMID: 37839502 PMCID: PMC11331176 DOI: 10.1016/j.jare.2023.10.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/08/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023] Open
Abstract
BACKGROUND Diabetes Mellitus (DM), a chronic metabolic disease characterized by elevated blood glucose, is caused by various degrees of insulin resistance and dysfunctional insulin secretion, resulting in hyperglycemia. The loss and failure of functional β-cells are key mechanisms resulting in type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). AIM OF REVIEW Elucidating the underlying mechanisms of β-cell failure, and exploring approaches for β-cell neogenesis to reverse β-cell dysfunction may provide novel strategies for DM therapy. KEY SCIENTIFIC CONCEPTS OF REVIEW Emerging studies reveal that genetic susceptibility, endoplasmic reticulum (ER) stress, oxidative stress, islet inflammation, and protein modification linked to multiple signaling pathways contribute to DM pathogenesis. Over the past few years, replenishing functional β-cell by β-cell neogenesis to restore the number and function of pancreatic β-cells has remarkably exhibited a promising therapeutic approach for DM therapy. In this review, we provide a comprehensive overview of the underlying mechanisms of β-cell failure in DM, highlight the effective approaches for β-cell neogenesis, as well as discuss the current clinical and preclinical agents research advances of β-cell neogenesis. Insights into the challenges of translating β-cell neogenesis into clinical application for DM treatment are also offered.
Collapse
Affiliation(s)
- Fanglin Niu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Wenxuan Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Ye Tian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Neurology, Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Wenzhen Shi
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Medical Research Center, the affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Man Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yujia Li
- Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, PR China; Department of Endocrinology, the Affiliated Hospital of Northwest University, Xi'an No.3 Hospital, Xi'an, Shaanxi, China
| |
Collapse
|
44
|
Lu F, Li E, Yang X. Proprotein convertase subtilisin/kexin type 9 deficiency in extrahepatic tissues: emerging considerations. Front Pharmacol 2024; 15:1413123. [PMID: 39139638 PMCID: PMC11319175 DOI: 10.3389/fphar.2024.1413123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 08/15/2024] Open
Abstract
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is primarily secreted by hepatocytes. PCSK9 is critical in liver low-density lipoprotein receptors (LDLRs) metabolism. In addition to its hepatocellular presence, PCSK9 has also been detected in cardiac, cerebral, islet, renal, adipose, and other tissues. Once perceived primarily as a "harmful factor," PCSK9 has been a focal point for the targeted inhibition of both systemic circulation and localized tissues to treat diseases. However, PCSK9 also contributes to the maintenance of normal physiological functions in numerous extrahepatic tissues, encompassing both LDLR-dependent and -independent pathways. Consequently, PCSK9 deficiency may harm extrahepatic tissues in close association with several pathophysiological processes, such as lipid accumulation, mitochondrial impairment, insulin resistance, and abnormal neural differentiation. This review encapsulates the beneficial effects of PCSK9 on the physiological processes and potential disorders arising from PCSK9 deficiency in extrahepatic tissues. This review also provides a comprehensive analysis of the disparities between experimental and clinical research findings regarding the potential harm associated with PCSK9 deficiency. The aim is to improve the current understanding of the diverse effects of PCSK9 inhibition.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
45
|
Toledo MP, Xie G, Wang YJ. Comprehensive Characterization of Islet Remodeling in Development and in Diabetes Using Mass Cytometry. Endocrinology 2024; 165:bqae094. [PMID: 39058908 DOI: 10.1210/endocr/bqae094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/11/2024] [Accepted: 07/25/2024] [Indexed: 07/28/2024]
Abstract
The pancreatic islet is the functional and structural unit of the pancreatic endocrine portion. Islet remodeling occurs in both normal development and pathogenesis of type 1 (T1D) and type 2 diabetes (T2D). However, accurately quantifying changes in islet cellular makeup and hormone expressions poses significant challenges due to large intra- and inter-donor heterogeneity and the limited scalability of traditional methods such as immunostaining. The cytometry by time-of-flight (CyTOF) technology enables simultaneous quantification of more than 30 protein markers at single-cell resolution in a high-throughput fashion. Moreover, with distinct DNA and viability markers, single live cells can be explicitly selected in CyTOF. Here, leveraging the CyTOF data generated by the Human Pancreas Analysis Program, we characterized more than 12 million islet cells from 71 donors. Our data revealed continued age-related changes in islet endocrine cell compositions, but the maturity of endocrine cells is reached by 3 years of age. We also observed significant changes in beta cell numbers and key protein expressions, along with a significant increase in bihormonal cells in T1D donors. In contrast, T2D donors exhibited minimal islet remodeling events. Our data shine a light on the islet dynamics during development and diabetes pathogenesis and suggest divergent pathogenesis processes of T1D and T2D. Our comprehensive approach not only elucidates islet plasticity but also establishes a foundation for integrated CyTOF analysis in islet biology and beyond.
Collapse
Affiliation(s)
- Maria Pilar Toledo
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Gengqiang Xie
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Yue J Wang
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
46
|
Yang Q, Wang X, Liu Y, Liu J, Zhu D. Metabolic factors are not the direct mediators of the association between type 2 diabetes and osteoporosis. Front Endocrinol (Lausanne) 2024; 15:1404747. [PMID: 39119008 PMCID: PMC11306037 DOI: 10.3389/fendo.2024.1404747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Objective The causal relationship between type 2 diabetes mellitus (T2DM) and osteoporosis (OS) remains unclear. This study aims to investigate the causal relationship and explore the potential metabolic mechanism and its mediating role. Methods We conducted a comprehensive study, gathering data on 490,089 T2DM patients from the genome-wide association study (GWAS) database and selecting OS data from FinnGen and MRC-IEU sources, including 212,778 and 463,010 patients, respectively, for causal analysis. Simultaneously, we explored the potential roles of three obesity traits and 30 metabolic and inflammation-related mediating variables in the causal relationship. Results There is a strong causal relationship between T2DM and OS. The data from our two different database sources appeared in the same direction, but after correcting for body mass index (BMI), waist circumference (WC), and waist-to-hip ratio (WHR), the direction became the same. T2DM may increase the risk of OS [odds ratio (OR) > 1.5, p < 0.001]. Steiger's test results show that there is no reverse causality. No risk factors related to glycolipid metabolism, amino acid metabolism, and inflammation were found to mediate the causal relationship. Conclusion This study's findings indicate a robust causal relationship between T2DM and OS, influenced by relevant factors such as BMI. Our results shed light on the pathogenesis of OS and underscore the importance for clinicians to treat metabolic disorders to prevent osteoporosis.
Collapse
Affiliation(s)
- Qifan Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Xinyu Wang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Yanwei Liu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jing Liu
- Department of Gynecology and Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Dong Zhu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
47
|
Pliszka M, Szablewski L. Associations between Diabetes Mellitus and Selected Cancers. Int J Mol Sci 2024; 25:7476. [PMID: 39000583 PMCID: PMC11242587 DOI: 10.3390/ijms25137476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Cancer is one of the major causes of mortality and is the second leading cause of death. Diabetes mellitus is a serious and growing problem worldwide, and its prevalence continues to grow; it is the 12th leading cause of death. An association between diabetes mellitus and cancer has been suggested for more than 100 years. Diabetes is a common disease diagnosed among patients with cancer, and evidence indicates that approximately 8-18% of patients with cancer have diabetes, with investigations suggesting an association between diabetes and some particular cancers, increasing the risk for developing cancers such as pancreatic, liver, colon, breast, stomach, and a few others. Breast and colorectal cancers have increased from 20% to 30% and there is a 97% increased risk of intrahepatic cholangiocarcinoma or endometrial cancer. On the other hand, a number of cancers and cancer therapies increase the risk of diabetes mellitus. Complications due to diabetes in patients with cancer may influence the choice of cancer therapy. Unfortunately, the mechanisms of the associations between diabetes mellitus and cancer are still unknown. The aim of this review is to summarize the association of diabetes mellitus with selected cancers and update the evidence on the underlying mechanisms of this association.
Collapse
Affiliation(s)
- Monika Pliszka
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| | - Leszek Szablewski
- Chair and Department of General Biology and Parasitology, Medical University of Warsaw, Chałubińskiego Str. 5, 02-004 Warsaw, Poland
| |
Collapse
|
48
|
Zhang J, Li Y, Yang L, Ma N, Qian S, Chen Y, Duan Y, Xiang X, He Y. New advances in drug development for metabolic dysfunction-associated diseases and alcohol-associated liver disease. Cell Biosci 2024; 14:90. [PMID: 38971765 PMCID: PMC11227172 DOI: 10.1186/s13578-024-01267-9] [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: 03/14/2024] [Accepted: 06/19/2024] [Indexed: 07/08/2024] Open
Abstract
Metabolic disorders are currently threatening public health worldwide. Discovering new targets and developing promising drugs will reduce the global metabolic-related disease burden. Metabolic disorders primarily consist of lipid and glucose metabolic disorders. Specifically, metabolic dysfunction-associated steatosis liver disease (MASLD) and alcohol-associated liver disease (ALD) are two representative lipid metabolism disorders, while diabetes mellitus is a typical glucose metabolism disorder. In this review, we aimed to summarize the new drug candidates with promising efficacy identified in clinical trials for these diseases. These drug candidates may provide alternatives for patients with metabolic disorders and advance the progress of drug discovery for the large disease burden.
Collapse
Affiliation(s)
- Jinming Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yixin Li
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, Anhui, China
| | - Liu Yang
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ningning Ma
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shengying Qian
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yingfen Chen
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yajun Duan
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, 230001, Anhui, China.
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yong He
- Shanghai Institute of Materia Medica (SIMM), Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
49
|
Maestas MM, Ishahak M, Augsornworawat P, Veronese-Paniagua DA, Maxwell KG, Velazco-Cruz L, Marquez E, Sun J, Shunkarova M, Gale SE, Urano F, Millman JR. Identification of unique cell type responses in pancreatic islets to stress. Nat Commun 2024; 15:5567. [PMID: 38956087 PMCID: PMC11220140 DOI: 10.1038/s41467-024-49724-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
Diabetes involves the death or dysfunction of pancreatic β-cells. Analysis of bulk sequencing from human samples and studies using in vitro and in vivo models suggest that endoplasmic reticulum and inflammatory signaling play an important role in diabetes progression. To better characterize cell type-specific stress response, we perform multiplexed single-cell RNA sequencing to define the transcriptional signature of primary human islet cells exposed to endoplasmic reticulum and inflammatory stress. Through comprehensive pair-wise analysis of stress responses across pancreatic endocrine and exocrine cell types, we define changes in gene expression for each cell type under different diabetes-associated stressors. We find that β-, α-, and ductal cells have the greatest transcriptional response. We utilize stem cell-derived islets to study islet health through the candidate gene CIB1, which was upregulated under stress in primary human islets. Our findings provide insights into cell type-specific responses to diabetes-associated stress and establish a resource to identify targets for diabetes therapeutics.
Collapse
Affiliation(s)
- Marlie M Maestas
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Matthew Ishahak
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Punn Augsornworawat
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Daniel A Veronese-Paniagua
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Kristina G Maxwell
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, USA
| | - Leonardo Velazco-Cruz
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Erica Marquez
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, USA
| | - Jiameng Sun
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Mira Shunkarova
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Sarah E Gale
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
| | - Fumihiko Urano
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Jeffrey R Millman
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, MSC 8127-057-08, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, MSC 8127-057-08, St. Louis, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, USA.
| |
Collapse
|
50
|
Qiu M, Chang L, Tang G, Ye W, Xu Y, Tulufu N, Dan Z, Qi J, Deng L, Li C. Activation of the osteoblastic HIF-1α pathway partially alleviates the symptoms of STZ-induced type 1 diabetes mellitus via RegIIIγ. Exp Mol Med 2024; 56:1574-1590. [PMID: 38945950 PMCID: PMC11297314 DOI: 10.1038/s12276-024-01257-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 02/04/2024] [Accepted: 03/19/2024] [Indexed: 07/02/2024] Open
Abstract
The hypoxia-inducible factor-1α (HIF-1α) pathway coordinates skeletal bone homeostasis and endocrine functions. Activation of the HIF-1α pathway increases glucose uptake by osteoblasts, which reduces blood glucose levels. However, it is unclear whether activating the HIF-1α pathway in osteoblasts can help normalize glucose metabolism under diabetic conditions through its endocrine function. In addition to increasing bone mass and reducing blood glucose levels, activating the HIF-1α pathway by specifically knocking out Von Hippel‒Lindau (Vhl) in osteoblasts partially alleviated the symptoms of streptozotocin (STZ)-induced type 1 diabetes mellitus (T1DM), including increased glucose clearance in the diabetic state, protection of pancreatic β cell from STZ-induced apoptosis, promotion of pancreatic β cell proliferation, and stimulation of insulin secretion. Further screening of bone-derived factors revealed that islet regeneration-derived protein III gamma (RegIIIγ) is an osteoblast-derived hypoxia-sensing factor critical for protection against STZ-induced T1DM. In addition, we found that iminodiacetic acid deferoxamine (SF-DFO), a compound that mimics hypoxia and targets bone tissue, can alleviate symptoms of STZ-induced T1DM by activating the HIF-1α-RegIIIγ pathway in the skeleton. These data suggest that the osteoblastic HIF-1α-RegIIIγ pathway is a potential target for treating T1DM.
Collapse
Affiliation(s)
- Minglong Qiu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Leilei Chang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Guoqing Tang
- Kunshan Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangzhou University, 388 Zuchongzhi Road, Kunshan, 215300, Jiangsu, China
| | - Wenkai Ye
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Yiming Xu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Nijiati Tulufu
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhou Dan
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Jin Qi
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Lianfu Deng
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Changwei Li
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|