51
|
Ratajczak W, Jones AG, Atkinson SD, Kelly C. Type 1 Diabetes Risk Variants Reduce Beta Cell Function. Genes (Basel) 2025; 16:172. [PMID: 40004501 PMCID: PMC11855905 DOI: 10.3390/genes16020172] [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: 12/02/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
INTRODUCTION The variants rs10517086 and rs1534422 are predictive of type 1 diabetes mellitus (T1DM) development and poor residual β cell function within the first year of diagnosis. However, the mechanism by which risk is conferred is unknown. We explored the impact of both variants on β cell function in vitro and assessed their relationship with C-peptide in people with T1DM and type 2 diabetes mellitus (T2DM). METHODS Using CRISPR/Cas9, the variants were introduced into a β cell line (BRIN-BD11) and a T cell line (Jurkat cells) from which the conditioned media was applied to otherwise healthy β cells to model the inflammatory environment associated with these variants. RESULTS Both variants significantly reduced glucose-stimulated insulin secretion, increased production of pro-inflammatory cytokines and reduced expression of several β cell markers and transcription factors (KCNJ11, KCNQ1, SCL2A2, GCK, NKX6.1, Pdx1 NGN3). However, HNF1A was significantly upregulated in the presence of both variants. We subsequently silenced HNF1A in variant expressing BRIN-BD11 cells using siRNA and found that gene expression profiles were normalised. Induction of each variant significantly increased expression of the lncRNAs they encode, which was normalised upon HNF1A silencing. Analysis of the DARE (Diabetes Alliance for Research in England) study revealed an association of rs10517086_A genotype with C-peptide in 153 individuals with T1DM, but not in 417 people with T2DM. CONCLUSIONS These data suggest that rs1534422 and rs10517086 exert multiple insults on the β cell through excessive upregulation of HNF1A and induction of pro-inflammatory cytokines, and highlight their utility as prognostic markers of β cell function.
Collapse
Affiliation(s)
- Wiktoria Ratajczak
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital Campus, Glenshane Road, Derry BT48 7JL, UK;
| | - Angus G. Jones
- University of Exeter Medical School, RILD Building, RD&E Hospital Wonford, Barrack Road, Exeter EX1 2LU, UK;
| | - Sarah D. Atkinson
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK;
| | - Catriona Kelly
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital Campus, Glenshane Road, Derry BT48 7JL, UK;
| |
Collapse
|
52
|
Karampelias C, Liu KC, Tengholm A, Andersson O. Mechanistic insights and approaches for beta cell regeneration. Nat Chem Biol 2025:10.1038/s41589-024-01822-y. [PMID: 39881214 DOI: 10.1038/s41589-024-01822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
Diabetes is characterized by variable loss of insulin-producing beta cells, and new regenerative approaches to increasing the functional beta cell mass of patients hold promise for reversing disease progression. In this Review, we summarize recent chemical biology breakthroughs advancing our knowledge of beta cell regeneration. We present current chemical-based tools, sensors and mechanistic insights into pathways that can be targeted to enhance beta cell regeneration in model organisms. We group the pathways according to the cellular processes they affect, that is, proliferation, conversion of other mature cell types to beta cells and beta cell differentiation from progenitor-like populations. We also suggest assays for assessing the functionality of the regenerated beta cells. Although regeneration processes differ between animal models, such as zebrafish, mice and pigs, regenerative mechanisms identified in any one animal model may be translatable to humans. Overall, chemical biology-based approaches in beta cell regeneration give hope that specific molecular pathways can be targeted to enhance beta cell regeneration.
Collapse
Affiliation(s)
- Christos Karampelias
- Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, Neuherberg, Germany.
| | - Ka-Cheuk Liu
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Anders Tengholm
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | - Olov Andersson
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden.
| |
Collapse
|
53
|
Li J, Guo K, Qiu J, Xue S, Pi L, Li X, Huang G, Xie Z, Zhou Z. Epidemiological status, development trends, and risk factors of disability-adjusted life years due to diabetic kidney disease: A systematic analysis of Global Burden of Disease Study 2021. Chin Med J (Engl) 2025; 138:568-578. [PMID: 39863522 PMCID: PMC11882292 DOI: 10.1097/cm9.0000000000003428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Approximately 40% of individuals with diabetes worldwide are at risk of developing diabetic kidney disease (DKD), which is not only the leading cause of kidney failure, but also significantly increases the risk of cardiovascular disease, causing significant societal health and financial burdens. This study aimed to describe the burden of DKD and explore its cross-country epidemiological status, predict development trends, and assess its risk factors and sociodemographic transitions. METHODS Based on the Global Burden of Diseases (GBD) Study 2021, data on DKD due to type 1 diabetes (DKD-T1DM) and type 2 diabetes (DKD-T2DM) were analyzed by sex, age, year, and location. Numbers and age-standardized rates were used to compare the disease burden between DKD-T1DM and DKD-T2DM among locations. Decomposition analysis was used to assess the potential drivers. Locally weighted scatter plot smoothing and Frontier analysis were used to estimate sociodemographic transitions of DKD disability-adjusted life years (DALYs). RESULTS The DALYs due to DKD increased markedly from 1990 to 2021, with a 74.0% (from 2,228,000 to 3,876,000) and 173.6% (from 4,123,000 to 11,279,000) increase for DKD-T1DM and DKD-T2DM, respectively. In 2030, the estimated DALYs for DKD-T1DM surpassed 4.4 million, with that of DKD-T2DM exceeding 14.6 million. Notably, middle-sociodemographic index (SDI) quintile was responsible for the most significant DALYs. Decomposition analysis revealed that population growth and aging were major drivers for the increased DKD DALYs in most GBD regions. Interestingly, the most pronounced effect of positive DALYs change from 1990 to 2021 was presented in high-SDI quintile, while in low-SDI quintile, DALYs for DKD-T1DM and DKD-T2DM presented a decreasing trend over the past years. Frontiers analysis revealed that there was a negative association between SDI quintiles and age-standardized DALY rates (ASDRs) in DKD-T1DM and DKD-T2DM. Countries with middle-SDI shouldered disproportionately high DKD burden. Kidney dysfunction (nearly 100% for DKD-T1DM and DKD-T2DM), high fasting plasma glucose (70.8% for DKD-T1DM and 87.4% for DKD-T2DM), and non-optimal temperatures (low and high, 5.0% for DKD-T1DM and 5.1% for DKD-T2DM) were common risk factors for age-standardized DALYs in T1DM-DKD and T2DM-DKD. There were other specific risk factors for DKD-T2DM such as high body mass index (38.2%), high systolic blood pressure (10.2%), dietary risks (17.8%), low physical activity (6.2%), lead exposure (1.2%), and other environmental risks. CONCLUSIONS DKD markedly increased and varied significantly across regions, contributing to a substantial disease burden, especially in middle-SDI countries. The rise in DKD is primarily driven by population growth, aging, and key risk factors such as high fasting plasma glucose and kidney dysfunction, with projections suggesting continued escalation of the burden by 2030.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Keyu Guo
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Junlin Qiu
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Song Xue
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Department of Nephrology, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Linhua Pi
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xia Li
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhiguo Xie
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
54
|
Lu Y, Wang Y, Ruan T, Wang Y, Ju L, Zhou M, Liu L, Yao D, Yao M. Immunometabolism of Tregs: mechanisms, adaptability, and therapeutic implications in diseases. Front Immunol 2025; 16:1536020. [PMID: 39917294 PMCID: PMC11798928 DOI: 10.3389/fimmu.2025.1536020] [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: 11/28/2024] [Accepted: 01/06/2025] [Indexed: 02/09/2025] Open
Abstract
Immunometabolism is an emerging field that explores the intricate interplay between immune cells and metabolism. Regulatory T cells (Tregs), which maintain immune homeostasis in immunometabolism, play crucial regulatory roles. The activation, differentiation, and function of Tregs are influenced by various metabolic pathways, such as the Mammalian targets of rapamycin (mTOR) pathway and glycolysis. Correspondingly, activated Tregs can reciprocally impact these metabolic pathways. Tregs also possess robust adaptive capabilities, thus enabling them to adapt to various microenvironments, including the tumor microenvironment (TME). The complex mechanisms of Tregs in metabolic diseases are intriguing, particularly in conditions like MASLD, where Tregs are significantly upregulated and contribute to fibrosis, while in diabetes, systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA), they show downregulation and reduced anti-inflammatory capacity. These phenomena suggest that the differentiation and function of Tregs are influenced by the metabolic environment, and imbalances in either can lead to the development of metabolic diseases. Thus, moderate differentiation and inhibitory capacity of Tregs are critical for maintaining immune system balance. Given the unique immunoregulatory abilities of Tregs, the development of targeted therapeutic drugs may position them as novel targets in immunotherapy. This could contribute to restoring immune system balance, resolving metabolic dysregulation, and fostering innovation and progress in immunotherapy.
Collapse
|
55
|
Gao M, Liu Q, Zhang L, Tabak F, Hua Y, Shao W, Li Y, Qian L, Liu Y. Identification of crucial extracellular genes as potential biomarkers in newly diagnosed Type 1 diabetes via integrated bioinformatics analysis. PeerJ 2025; 13:e18660. [PMID: 39802181 PMCID: PMC11725270 DOI: 10.7717/peerj.18660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/17/2024] [Indexed: 01/16/2025] Open
Abstract
Purpose In this study, we aimed to study the role of extracellular proteins as biomarkers associated with newly diagnosed Type 1 diabetes (NT1D) diagnosis and prognosis. Patients and Methods We retrieved and analyzed the GSE55098 microarray dataset from the Gene Expression Omnibus (GEO) database. Using R software, we screened out the extracellular protein-differentially expressed genes (EP-DEGs) through several protein-related databases. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were applied to describe the role and function of these EP-DEGs. We used the STRING database to construct the interaction of proteins, Cytoscape software to visualize the protein-protein interaction (PPI) networks, and its plugin CytoHubba to identify the crucial genes between PPI networks. Finally, we used the comparative toxicogenomics database (CTD) to evaluate the connection between NT1D with the potential crucial genes and we validated our conclusions with another dataset (GSE33440) and some clinical samples. Results We identified 422 DEGs and 122 EP-DEGs from a dataset that includes (12) NT1D patients compared with (10) healthy people. Protein digestion and absorption, toll-like receptor signaling, and T cell receptor signaling were the most meaningful pathways defined by KEGG enrichment analyses. We recognized nine important extracellular genes: GZMB, CCL4, TNF, MMP9, CCL5, IFNG, CXCL1, GNLY, and LCN2. CTD analyses showed that LCN2, IFNG, and TNF had higher levels in NT1D and hypoglycemia; while TNF, IFNG and MMP9 increased in hyperglycemia. Further verification showed that LCN2, MMP9, TNF and IFNG were elevated in NT1D patients. Conclusion The nine identified key extracellular genes, particularly LCN2, IFNG, TNF, and MMP9, may be potential diagnostic biomarkers for NT1D. Our findings provide new insights into the molecular mechanisms and novel therapeutic targets of NT1D.
Collapse
Affiliation(s)
- Ming Gao
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Qing Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lingyu Zhang
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Fatema Tabak
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yifei Hua
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Wei Shao
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yangyang Li
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Li Qian
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu Liu
- Department of Endocrinology, Sir Run Run Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
56
|
Sundheim B, Hirani K, Blaschke M, Lemos JRN, Mittal R. Pre-Type 1 Diabetes in Adolescents and Teens: Screening, Nutritional Interventions, Beta-Cell Preservation, and Psychosocial Impacts. J Clin Med 2025; 14:383. [PMID: 39860389 PMCID: PMC11765808 DOI: 10.3390/jcm14020383] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
Type 1 Diabetes (T1D) is a progressive autoimmune disease often identified in childhood or adolescence, with early stages detectable through pre-diabetic markers such as autoantibodies and subclinical beta-cell dysfunction. The identification of the pre-T1D stage is critical for preventing complications, such as diabetic ketoacidosis, and for enabling timely interventions that may alter disease progression. This review examines the multifaceted approach to managing T1D risk in adolescents and teens, emphasizing early detection, nutritional interventions, beta-cell preservation strategies, and psychosocial support. Screening for T1D-associated autoantibodies offers predictive insight into disease risk, particularly when combined with education and family resources that promote lifestyle adjustments. Although nutritional interventions alone are not capable of preventing T1D, certain lifestyle interventions, such as weight management and specific nutritional choices, have shown the potential to preserve insulin sensitivity, reduce inflammation, and mitigate metabolic strain. Pharmacological strategies, including immune-modulating drugs like teplizumab, alongside emerging regenerative and cell-based therapies, offer the potential to delay disease onset by protecting beta-cell function. The social and psychological impacts of a T1D risk diagnosis are also significant, affecting adolescents' quality of life, family dynamics, and mental health. Supportive interventions, including counseling, cognitive-behavioral therapy (CBT), and group support, are recommended for managing the emotional burden of pre-diabetes. Future directions call for integrating universal or targeted screening programs within schools or primary care, advancing research into nutrition and psychosocial support, and promoting policies that enhance access to preventive resources. Advocacy for the insurance coverage of screening, nutritional counseling, and mental health services is also crucial to support families in managing T1D risk. By addressing these areas, healthcare systems can promote early intervention, improve beta-cell preservation, and support the overall well-being of adolescents at risk of T1D.
Collapse
Affiliation(s)
- Brody Sundheim
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Ransom Everglades High School, 3575 Main Hwy, Miami, FL 33133, USA
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Krish Hirani
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- American Heritage School, 12200 W Broward Blvd, Plantation, FL 33325, USA
| | - Mateo Blaschke
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Coral Gables High School, 450 Bird Rd, Coral Gables, FL 33146, USA
| | - Joana R. N. Lemos
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rahul Mittal
- Young Leaders Advocacy Group, Diabetes Research Institute Foundation, Hollywood, FL 33021, USA; (B.S.); (K.H.); (M.B.); (J.R.N.L.)
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
57
|
Ullsten S, Østnes Hansen K, Petit GA, Hansen EH, Andersen JH. Promotion of beta cell proliferation through DYRK kinase inhibition using the marine natural product breitfussin C. Sci Rep 2025; 15:1247. [PMID: 39774736 PMCID: PMC11706957 DOI: 10.1038/s41598-025-85178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Pro-inflammatory cytokines, like interleukin-1 beta and interferon gamma, are known to activate signalling pathways causing pancreatic beta cell death and dysfunction, contributing to the onset of diabetes. Targeting cytokine signalling pathways offers a potential strategy to slow or even halt disease progression, reducing reliance on exogenous insulin and improving glucose regulation. This study explores the protective and proliferative effects of breitfussin C (BfC), a natural compound isolated from the Arctic marine hydrozoan Thuiaria breitfussi, on pancreatic beta cells exposed to pro-inflammatory cytokines. Using the beta cell line RIN-M5F, we assessed the protective effects of BfC through a MTS assay for cell viability, caspase 3/7 activity for apoptosis, and EdU incorporation and cell cycle distribution for proliferation. Additionally, we investigated BfC's inhibitory effects on the DYRK family of kinases using kinase activity and binding assays, western blotting, and docking simulations. Our findings reveal that BfC treatment effectively increases beta cell proliferation and counteracts cytokine-induced decrease in proliferation. The proliferative effect is associated with inhibition of DYRK kinases and a subsequent decrease in the cell cycle inhibitor p27KIP. These results suggest that BfC mediates beta cell-protective effect by promoting proliferation through DYRK inhibition, highlighting its potential as a molecular starting point for the development of a therapeutic agent against diabetes.
Collapse
Affiliation(s)
- Sara Ullsten
- MARBIO, UiT - The Arctic University of Norway, Breivika, 9037, Tromsø, Norway
| | - Kine Østnes Hansen
- MARBIO, UiT - The Arctic University of Norway, Breivika, 9037, Tromsø, Norway
| | | | - Espen Holst Hansen
- MARBIO, UiT - The Arctic University of Norway, Breivika, 9037, Tromsø, Norway
| | | |
Collapse
|
58
|
Smith F, Kotowska AM, Fiedler B, Cerny E, Cheung K, Rutland CS, Chowdhury F, Segal J, Rawson FJ, Marlow M. Using Oscillation to Improve the Insertion Depth and Consistency of Hollow Microneedles for Transdermal Insulin Delivery with Mechanistic Insights. Mol Pharm 2025; 22:316-329. [PMID: 39625848 PMCID: PMC11707737 DOI: 10.1021/acs.molpharmaceut.4c00942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 01/07/2025]
Abstract
Microneedles (MNs) offer the potential for discrete and painless transdermal drug delivery, yet poor insertion and dosing consistency have hindered their clinical translation. Specifically, hollow MNs are appropriate for the administration of liquid modalities, including insulin, which could prove to be beneficial for patients with type 1 diabetes mellitus. This work aimed to design and manufacture a hollow MN with an improved insertion and delivery profile suitable for insulin administration. Ex vivo insertion studies demonstrated that oscillation of MNs upon insertion into skin produced a favorable insertion profile, with reduced variation, compared to static MN insertion. Histological staining showed that this could be due to the repeated motion of the oscillating MN disrupting elastic fibers in the dermis. Additionally, permeation studies demonstrated that increased quantities of insulin were able to permeate the skin when oscillation was employed compared to static MN insertion. This study has shown that oscillation is a valuable tool in improving the transdermal delivery of insulin via a single hollow MN in vitro. Moving forward, in vivo studies should be completed to gain a fuller understanding of the benefits of the oscillation of MNs on transdermal drug delivery.
Collapse
Affiliation(s)
- Fiona Smith
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Anna M. Kotowska
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Benjamin Fiedler
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
- School
of Pharmacy, University College London, London WC1N 1AX, United Kingdom
| | - Edward Cerny
- Advanced
Technology Centre, Oakwood Drive, Nemaura
Pharma Limited, Loughborough, Leicestershire LE11 3QF, United Kingdom
| | - Karmen Cheung
- Advanced
Technology Centre, Oakwood Drive, Nemaura
Pharma Limited, Loughborough, Leicestershire LE11 3QF, United Kingdom
| | - Catrin S. Rutland
- School
of Veterinary Medicine and Science, The
University of Nottingham, Nottingham NG7 2RD, U.K.
| | - Faz Chowdhury
- Advanced
Technology Centre, Oakwood Drive, Nemaura
Pharma Limited, Loughborough, Leicestershire LE11 3QF, United Kingdom
| | - Joel Segal
- Department
of Mechanical, Materials and Manufacturing Engineering, Faculty of
Engineering, University of Nottingham, Nottingham NG8 1BB, United Kingdom
| | - Frankie J. Rawson
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Maria Marlow
- School
of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
59
|
Mallone R, Sims E, Achenbach P, Mathieu C, Pugliese A, Atkinson M, Dutta S, Evans-Molina C, Klatzmann D, Koralova A, Long SA, Overbergh L, Rodriguez-Calvo T, Ziegler AG, You S. Emerging Concepts and Success Stories in Type 1 Diabetes Research: A Road Map for a Bright Future. Diabetes 2025; 74:12-21. [PMID: 39446565 DOI: 10.2337/db24-0439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
Type 1 diabetes treatment stands at a crucial and exciting crossroad since the 2022 U.S. Food and Drug Administration approval of teplizumab to delay disease development. In this article, we discuss four major conceptual and practical issues that emerged as key to further advancement in type 1 diabetes research and therapies. First, collaborative networks leveraging the synergy between the type 1 diabetes research and care community members are key to fostering innovation, know-how, and translation into the clinical arena worldwide. Second, recent clinical trials in presymptomatic stage 2 and recent-onset stage 3 disease have shown the promise, and potential pitfalls, of using immunomodulatory and/or β-cell protective agents to achieve sustained remission or prevention. Third, the increasingly appreciated heterogeneity of clinical, immunological, and metabolic phenotypes and disease trajectories is of critical importance to advance the decision-making process for tailored type 1 diabetes care and therapy. Fourth, the clinical benefits of early diagnosis of β-cell autoimmunity warrant consideration of general population screening for islet autoantibodies, which requires further efforts to address the technical, organizational, and ethical challenges inherent to a sustainable program. Efforts are underway to integrate these four concepts into the future directions of type 1 diabetes research and therapy. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Roberto Mallone
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
- Service de Diabétologie et Immunologie Clinique, Hôpital Cochin, Assistance Publique Hôpitaux de Paris, Paris, France
- Indiana Biosciences Research Institute, Indianapolis, IN
| | - Emily Sims
- Division of Pediatric Endocrinology and Diabetology, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
| | - Peter Achenbach
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Alberto Pugliese
- Department of Diabetes Immunology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA
| | - Mark Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL
- Department of Pediatrics, University of Florida, Gainesville, FL
| | | | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Department of Medicine, and Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
- Richard L. Roudebush Veterans' Administration Medical Center, Indianapolis, IN
| | - David Klatzmann
- Clinical Investigation Center for Biotherapies and Inflammation-Immunopathology-Biotherapy Department (i2B), Sorbonne Université, Pitié-Salpêtrière Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
- Immunology-Immunopathology-Immunotherapy (i3), INSERM UMRS 959, Sorbonne UniversitéParis, France
| | - Anne Koralova
- The Leona M. and Harry B. Helmsley Charitable Trust, New York, NY
| | - S Alice Long
- Translational Immunology, Benaroya Research Institute, Seattle, WA
| | - Lut Overbergh
- Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
| | - Teresa Rodriguez-Calvo
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany
| | - Sylvaine You
- Institut Cochin, CNRS, INSERM, Université Paris Cité, Paris, France
- Indiana Biosciences Research Institute, Indianapolis, IN
| |
Collapse
|
60
|
de Klerk E, Deacon E, van Rensburg E. Reviewing identity development in young people living with Type 1 Diabetes Mellitus. J Adolesc 2025; 97:73-84. [PMID: 39327821 DOI: 10.1002/jad.12412] [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: 08/07/2023] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
INTRODUCTION Type 1 Diabetes Mellitus (T1DM) is a chronic condition increasing among young people. Identity development occurs during an individual's life and can be significantly influenced by a chronic disease such as T1DM. We have critically reviewed the relevant scientific literature to understand young people's identity development with T1DM. METHODS A critical review design was employed to answer the research question: "What does scientific literature state regarding identity development in young people living with Type 1 Diabetes Mellitus?" Numerous databases were searched to include the most relevant scientific literature to answer the research questions. Boolean operator phrases were ultimately used to search for the literature. The initial screening produced 1319 scientific literature, among which seven articles were analyzed thematically. RESULTS Analysis revealed several significant themes: The identity of young people with T1DM develops differently from those without this chronic condition; young people can either incorporate or contain their T1DM in their identity development, highlighting the complex nature of this process; and numerous external factors significantly influence identity development in young people living with a chronic condition such as T1DM. CONCLUSION The study underscores that identity develops differently in young people living with T1DM than in those without chronic conditions. However, the existing studies and literature exploring the experiences of young people living with T1DM have relied on information from caregivers and health professionals. This reliance on secondary sources underscores the urgent need for more research to obtain data from young people living with T1DM as a primary source. Such a shift in research practices is crucial to gaining a more comprehensive and accurate understanding of their experiences.
Collapse
Affiliation(s)
- Elinda de Klerk
- Community Psychosocial Research (COMPRES), School of Psychosocial Health, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| | - Elmarí Deacon
- Optentia Research Focus Area, Faculty of Humanities, North-West University, Potchefstroom, South Africa
| | - Esmé van Rensburg
- Community Psychosocial Research (COMPRES), School of Psychosocial Health, Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
| |
Collapse
|
61
|
Yokomichi H, Mochizuki M, Suzuki S, Ito Y, Hotsubo T, Matsuura N. Slowly progressive subtype of childhood-onset type 1 diabetes as a high-risk factor for end-stage renal disease: A cohort study in Japan. J Diabetes Complications 2025; 39:108922. [PMID: 39616658 DOI: 10.1016/j.jdiacomp.2024.108922] [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: 06/16/2024] [Revised: 11/01/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
AIM To compare the incidence of end-stage renal disease (ESRD) between slowly progressive type 1 diabetes and acute-onset type 1 diabetes. METHODS This cohort study enrolled all 521 patients with childhood-onset type 1 diabetes with the year of onset from 1959 to 1996 in Hokkaido Prefecture, Japan. We calculated the ESRD incidence rate per 100,000 person-years by sex, onset year, onset age, and type 1 diabetes subtype (slowly progressive or acute-onset). We also constructed a Kaplan-Meier curve for ESRD by these risk factors. RESULTS The data of 391 patients were gathered, among whom 66 developed ESRD. The ESRD incidence rate per 100,000 person-years was 525 among all patients; 538 and 503 among women (n = 235) and men (n = 156); 893, 413, and 225 for onset year of 1959-1979 (n = 107), 1980-1989 (n = 201), and 1990-1996 (n = 83); 420 and 715 for onset before (n = 243) and after (n = 148) puberty; and 1388 and 432 for the slowly progressive (n = 41) and acute-onset (n = 350) subtypes, respectively. The Kaplan-Meier curve also indicated a significantly higher incidence of ESRD in slowly progressive than in acute-onset type 1 diabetes. CONCLUSION The incidence of ESRD was higher in slowly progressive than acute-onset type 1 diabetes.
Collapse
Affiliation(s)
- Hiroshi Yokomichi
- Department of Epidemiology and Environmental Medicine, University of Yamanashi, 1110 Shimokato, Chuo City, Yamanashi 409-3898, Japan.
| | - Mie Mochizuki
- Department of Paediatrics, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, Japan; Department of Paediatrics, NHO Kofu National Hospital, 11-35 Tenjincho, Kofu, Yamanashi, Japan
| | - Shigeru Suzuki
- Department of Paediatrics, Asahikawa Medical University, 2-1-1-1 Midorigaoka-higashi, Asahikawa, Hokkaido, Japan
| | - Yoshiya Ito
- Division of Clinical Medicine, Japanese Red Cross Hokkaido College of Nursing, 664-1 Akibonocho, Kitami, Hokkaido, Japan
| | - Tomoyuki Hotsubo
- Sapporo Children's Endocrine Clinic, 14-291-81-2F Minami-ichijo-nishi, Chuo, Sapporo, Hokkaido, Japan
| | - Nobuo Matsuura
- Bibai City Hospital, 1-1-1 Kita, Nishi-nijo, Bibai, Hokkaido, Japan
| |
Collapse
|
62
|
Okamura T, Kitagawa N, Kitagawa N, Sakai K, Sumi M, Kobayashi G, Imai D, Matsui T, Hamaguchi M, Fukui M. Single-cell analysis reveals islet autoantigen's immune activation in type 1 diabetes patients. J Clin Biochem Nutr 2025; 76:64-84. [PMID: 39896168 PMCID: PMC11782777 DOI: 10.3164/jcbn.24-86] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 02/04/2025] Open
Abstract
In this study, we used single-cell sequencing, which can comprehensively detect the type and number of transcripts per cell, to efficiently stimulate peripheral blood mononuclear cells of type 1 diabetic patients with overlapping peptides of GAD, IA-2, and insulin antigens, and performed gene expression analysis by single-cell variable-diversity-joining sequencing and T-cell receptor repertoire analysis. Twenty male patients with type 1 diabetes mellitus participating in the KAMOGAWA-DM cohort were included. Four of them were randomly selected for BD Rhapsody system after reacting peripheral blood mononuclear cells with overlapping peptides of GAD, IA-2, and insulin antigen. Peripheral blood mononuclear cells were clustered into CD8+ T cells, CD4+ T cells, granulocytes, natural killer cells, dendritic cells, monocytes, and B cells based on Seurat analysis. In the insulin group, gene expression of inflammatory cytokines was elevated in cytotoxic CD8+ T cells and Th1 and Th17 cells, and gene expression related to exhaustion was elevated in regulatory T cells. In T cell receptors of various T cells, the T cell receptor β chain was monoclonally increased in the TRBV28/TRBJ2-7 pairs. This study provides insights into the pathogenesis of type 1 diabetes and provides potential targets for the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Takuro Okamura
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Noriyuki Kitagawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Diabetology, Kameoka Municipal Hospital, 1-1 Shinonoda, Shino-cho, Kameoka 621-8585, Japan
| | - Nobuko Kitagawa
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kimiko Sakai
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Madoka Sumi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Genki Kobayashi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Dan Imai
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takaaki Matsui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, Kawaramachi-Hirokoji, Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
63
|
Valenzise M, Bombaci B, Lombardo F, Passanisi S, Lombardo C, Lugarà C, D'Amico F, Grasso L, Aguennouz M, Catalano A, Salzano G. Association between osteocalcin and residual β-cell function in children and adolescents newly diagnosed with type 1 diabetes: a pivotal study. J Endocrinol Invest 2025; 48:227-232. [PMID: 38965181 DOI: 10.1007/s40618-024-02414-2] [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: 02/19/2024] [Accepted: 05/29/2024] [Indexed: 07/06/2024]
Abstract
PURPOSE This pivotal study aimed to evaluate circulating levels of bone remodeling markers in children and adolescents at the onset of type 1 diabetes (T1D). Additionally, we assessed their correlation with glucose control, residual β-cell function, and the severity of presentation. METHODS In this single-center cross-sectional study, we recruited children and adolescents newly diagnosed with T1D at our tertiary-care Diabetes Centre. Anamnestic, anthropometric, clinical, and biochemical data at T1D diagnosis were collected. Basal and stimulated C-peptide levels were assessed, along with the following bone remodeling biomarkers: osteocalcin (OC), alkaline phosphatase (ALP), parathormone (PTH), 25-OH Vitamin D (25OH-D), and the C-terminal cross-linked telopeptide of type 1 collagen (CTX). RESULTS We enrolled 29 individuals newly diagnosed with T1D, with a slight male prevalence (51.7%). The mean age was 8.4 ± 3.7 years. A positive correlation between OC and stimulated C-peptide (R = 0.538; p = 0.026) and between PTH and serum HCO3- (R = 0.544; p = 0.025) was found. No other correlations between bone remodeling biomarkers and clinical variables were detected. CONCLUSION Our data showed a positive correlation between OC levels and residual β-cell function in children and adolescents at T1D presentation. Further longitudinal studies evaluating OC levels in pediatric subjects with T1D are needed to better understand the complex interaction between bone and glucose metabolisms.
Collapse
Affiliation(s)
- M Valenzise
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy.
| | - B Bombaci
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - F Lombardo
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - S Passanisi
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - C Lombardo
- Department of Biomedical Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - C Lugarà
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - F D'Amico
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| | - L Grasso
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - M Aguennouz
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - A Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - G Salzano
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina, Messina, Italy
| |
Collapse
|
64
|
Abhale K, Addepalli V, Desai S, Sanap A, Bhonde R. Effects of Mesenchymal Stem Cell-conditioned Media with Natural Immunomodulatory Agent Resveratrol on Type 1 Diabetes. Curr Drug Discov Technol 2025; 22:e080324227818. [PMID: 38468534 DOI: 10.2174/0115701638276524240305054259] [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: 10/03/2023] [Revised: 01/18/2024] [Accepted: 01/29/2024] [Indexed: 03/13/2024]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is a condition marked by elevated blood sugar levels and primarily recognized by the destruction of beta cells caused by an autoimmune attack, which is a significant characteristic of T1DM. Recent studies have demonstrated the regenerative potential of conditioned medium therapy. In light of this, the current research sought to assess the impact of Mesenchymal Stem Cell conditioned media (CM) and CM with resveratrol (CM+ Resveratrol) on the management of T1DM in Swiss albino mice. By leveraging and modifying existing conditioned medium therapy, this study aims to evaluate its effectiveness in treating T1DM. MATERIALS & METHODS Diabetes was induced in animals using the diabetes-inducing agent streptozotocin (STZ). The animals were then divided into five groups: Normal control, Disease Control, Resveratrol, Condition Media, and CM + Resveratrol. Treatments were given to the animals accordingly. The study period was 28 days. During this time, the animals were monitored for foodwater intake twice a week, blood glucose levels, and body weight. At the conclusion of the 28-day study period, biochemical estimations were performed for serum insulin levels, C-peptide levels, anti-inflammatory cytokines levels and pro-inflammatory cytokines levels. Additionally, histopathology of the pancreas was performed. RESULTS The test groups showed a significant decrease in blood glucose levels, an increase in Cpeptide levels, and a decrease in pro-inflammatory cytokine levels compared to the disease group. However, no statistically significant change within groups was observed in terms of serum insulin and anti-inflammatory cytokine levels. The improvement in diabetic symptoms, such as polyphagia, polydipsia, and weight loss, was observed in the treatment group, along with pancreatic regeneration, which indicated improved insulin secretion. CONCLUSION In the current investigation, we concluded that CM and CM+ Resveratrol, as natural immunomodulators, have the capacity to regenerate injured pancreatic beta cells and have antidiabetic action, together with immunomodulating impact. Nonetheless, future studies on this therapy appear to be promising.
Collapse
Affiliation(s)
- Krushna Abhale
- Department of Pharmacology, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune, India
| | | | - Shivani Desai
- Clinical Research and Pharmacovigilance, Serum Institute of India Pvt. Ltd., Hadapsar, Pune
| | - Avinash Sanap
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| | - Ramesh Bhonde
- Regenerative Medicine Laboratory, Dr. D. Y. Patil Dental College and Hospital, Pimpri, Pune, India
| |
Collapse
|
65
|
Rangraze IR, El-Tanani M, Arman Rabbani S, Babiker R, Matalka II, Rizzo M. Diabetes and its Silent Partner: A Critical Review of Hyperinsulinemia and its Complications. Curr Diabetes Rev 2025; 21:e15733998311738. [PMID: 39192649 DOI: 10.2174/0115733998311738240813110032] [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: 03/19/2024] [Revised: 06/09/2024] [Accepted: 06/21/2024] [Indexed: 08/29/2024]
Abstract
In this complex realm of diabetes, hyperinsulinemia is no longer regarded as just a compensatory response to insulin resistance but rather has evolved into an integral feature. This comprehensive review provides a synthesis of the current literature, including various aspects associated with hyperinsulinemia in diabetic complications. Hyperinsulinemia has been shown to be more than just a compensatory mechanism, and the key findings demonstrate how hyperinsulinism affects the development of cardiovascular events as well as microvascular complications. Additionally, recognizing hyperinsulinemia as a modifiable factor, the diabetes management paradigm shifts towards cognitive ones that consider the use of lifestyle modifications in combination with newer pharmacotherapies and precision medicine approaches. These findings have crucial implications for the clinical work, requiring a careful appreciation of hyperinsulinemia's changing aspects as well as incorporation in personalized treatment protocol. In addition, the review focuses on bigger issues related to public health, showing that prevention and early diagnosis will help reduce the burden of complications. Research implications favor longitudinal studies, biomarker discovery, and the study of emerging treatment modalities; clinical practice should adopt global evaluations, patient education, and precision medicine adaptation. Finally, this critical review provides an overview of the underlying processes of hyperinsulinemia in diabetes and its overall health effects.
Collapse
Affiliation(s)
- Imran Rashid Rangraze
- Internal Medicine Department, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Rasal- Khaimah, United Arab Emirates
| | - Mohamed El-Tanani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Syed Arman Rabbani
- College of Pharmacy, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Rasha Babiker
- Physiology Department, RAK College of Medical Sciences, RAK Medical and Health Sciences University, Ras-al-Khaimah, United Arab Emirates
| | - Ismail I Matalka
- Department of Pathology, Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Childcare, Internal Medicine and Medical Specialties, School of Medicine, University of Palermo, Palermo, Italy
| |
Collapse
|
66
|
Öztaş G, Aydın Aİ, Atak M, Özyazıcıoğlu N, Sağlam H. Relationship among nursing support for parents of adolescents diagnosed with Type 1 Diabetes, parental monitoring of adolescents and the self-efficacy of adolescents with Type 1 Diabetes. J Pediatr Nurs 2025; 80:9-15. [PMID: 39536382 DOI: 10.1016/j.pedn.2024.10.040] [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: 08/30/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND This study aims to explore the connection between parental monitoring in the treatment of adolescents with type 1 diabetes mellitus (T1DM), the self-efficacy of adolescents with diabetes, and nurse assistance provided to parents of adolescents with T1DM. METHODS This cross-sectional descriptive study consisted of 115 adolescents aged 10-18 with T1DM and their parents. Study data were collected through the "Diabetes Management Self-Efficacy Scale in Adolescents with T1DM", "Introductory Information Form", "Nurse-Parent Support Tool (NPST)" and "Parental Monitoring of Diabetes Care Questionnaire in Adolescents with T1DM". RESULTS Only 31.3 % of the study adolescent participants reported monitoring their blood sugar less than four times per day, and the frequency of blood sugar monitoring increased with increasing parental supervision (p = 0.021). When diabetes was managed only by parents, adolescents' perceptions of their self-management were statistically significant and high (p = 0.015). Neither the level of nurse-parent support nor the connection between parental monitoring and adolescents' self-efficacy was statistically significant. CONCLUSIONS Adolescents with T1DM should receive support from both parents and medical professionals. In particular, nurses should closely inquire about the adolescent's and family's disease management strategies during hospital visits and health monitoring.
Collapse
Affiliation(s)
- Gülay Öztaş
- Faculty of Health Science, Department of Nursing, Yozgat Bozok University, Yozgat, Turkiye.
| | - Ayla İrem Aydın
- Faculty of Health Science, Department of Nursing, Bursa Uludag University, Bursa, Turkiye.
| | - Meryem Atak
- Faculty of Health Science, Department of Nursing, Bursa Uludag University, Bursa, Turkiye.
| | - Nurcan Özyazıcıoğlu
- Faculty of Health Science, Department of Nursing, Bursa Uludag University, Bursa, Turkiye.
| | - Halil Sağlam
- Faculty of Medicine, Department of Pediatric Endocrinology, Bursa Uludag University, Bursa, Turkiye.
| |
Collapse
|
67
|
Carro Vázquez D, Emini L, Rauner M, Hofbauer C, Skalicky S, Wagner A, Grillari J, Diendorfer AB, Hofbauer LC, Hackl M. Differential microRNA expression patterns between TallyHo/JngJ mice and non-diabetic Swiss Webster Random/Jackson mice. JBMR Plus 2025; 9:ziae121. [PMID: 39664932 PMCID: PMC11631062 DOI: 10.1093/jbmrpl/ziae121] [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: 02/22/2024] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 12/13/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases the susceptibility of bone fragility. The underlying mechanisms have, however, remained largely unknown. MicroRNAs (miRNAs) are short single-stranded non-coding RNA molecules with utility as biomarkers due to their easy accessibility and stability in bodily fluids. Here, we aimed to use an unbiased approach to identify miRNAs dysregulated in a polygenic mouse model of T2DM. Genome-wide analysis of miRNAs in serum, BM, and bone from the polygenic TallyHo/JngJ (TH) mice, which recapitulate T2DM in humans, was performed. This analysis was compared to the recommended control Swiss Webster Random/Jackson (SWR/J) and a strain-matched non-diabetic control (TH-ND). When comparing TH mice with TH-ND using an adjusted p-value false discovery rate (FDR) cut-off of 0.2 to identify differentially expressed miRNAs, mmu-miR-466i-5p and mmu-miR-1195 were found to be up-regulated in both serum and in BM. Dysregulated miRNAs were not found in bone tissue. When comparing TH-ND mice with SWR/J using the same FDR cut-off, mmu-miR-351-5p, and mmu-miR-322-3p were upregulated in both BM and serum, while mmu-miR-449a-5p and mmu-miR-6240 were downregulated in BM and serum. Dysregulated miRNAs in BM or cortical bone compared to serum between TH-ND mice and SWR/J were investigated for their cell-type enrichment to identify putative donor cells and their gene target networks. Gene target network analysis revealed genes involved in diabetes-related signaling pathways as well as in diabetic bone disease. Cell-type enrichment analysis identified hsa-miR-449a enriched in immune cells, hsa-miR-592 in hepatocytes and endothelial cells, while hsa-miR-424-3p, hsa-miR-1-3p, and hsa-miR-196b-5p were enriched in mesenchymal stem cells and their derived tissues. In conclusion, our comparative miRNA profiling sheds light on differential expression patterns between SWR/J and both subgroups of TH. No differences were observed between TH and TH-ND, suggesting the genetic background of SWR/J may be responsible for the change of dysregulated miRNA.
Collapse
Affiliation(s)
| | - Lejla Emini
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Christine Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Susanna Skalicky
- TAmiRNA GmbH, Department of Research and Development, 1110 Vienna, Austria
| | - Alisia Wagner
- TAmiRNA GmbH, Department of Research and Development, 1110 Vienna, Austria
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, 1200 Vienna, Austria
- Department of Biotechnology, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, 1190 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| | | | - Lorenz C Hofbauer
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden Medical Center, 01307 Dresden, Germany
| | - Matthias Hackl
- TAmiRNA GmbH, Department of Research and Development, 1110 Vienna, Austria
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
68
|
Kang S, Penaloza Aponte JD, Elashkar O, Morales JF, Waddington N, Lamb DG, Ju H, Campbell-Thompson M, Kim S. Leveraging pre-trained machine learning models for islet quantification in type 1 diabetes. J Pathol Inform 2025; 16:100406. [PMID: 39720415 PMCID: PMC11665367 DOI: 10.1016/j.jpi.2024.100406] [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: 09/13/2024] [Revised: 10/23/2024] [Accepted: 11/01/2024] [Indexed: 12/26/2024] Open
Abstract
Human islets display a high degree of heterogeneity in terms of size, number, architecture, and endocrine cell-type compositions. An ever-increasing number of immunohistochemistry-stained whole slide images (WSIs) are available through the online pathology database of the Network for Pancreatic Organ donors with Diabetes (nPOD) program at the University of Florida (UF). We aimed to develop an enhanced machine learning-assisted WSI analysis workflow to utilize the nPOD resource for analysis of endocrine cell heterogeneity in the natural history of type 1 diabetes (T1D) in comparison to donors without diabetes. To maximize usability, the user-friendly open-source software QuPath was selected for the main interface. The WSI data were analyzed with two pre-trained machine learning models (i.e., Segment Anything Model (SAM) and QuPath's pixel classifier), using the UF high-performance-computing cluster, HiPerGator. SAM was used to define precise endocrine cell and cell grouping boundaries (with an average quality score of 0.91 per slide), and the artificial neural network-based pixel classifier was applied to segment areas of insulin- or glucagon-stained cytoplasmic regions within each endocrine cell. An additional script was developed to automatically count CD3+ cells inside and within 20 μm of each islet perimeter to quantify the number of islets with inflammation (i.e., CD3+ T-cell infiltration). Proof-of-concept analysis was performed to test the developed workflow in 12 subjects using 24 slides. This open-source machine learning-assisted workflow enables rapid and high throughput determinations of endocrine cells, whether as single cells or within groups, across hundreds of slides. It is expected that the use of this workflow will accelerate our understanding of endocrine cell and islet heterogeneity in the context of T1D endotypes and pathogenesis.
Collapse
Affiliation(s)
- Sanghoon Kang
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, Intelligent Critical Care Center, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Jesus D. Penaloza Aponte
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Omar Elashkar
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, Intelligent Critical Care Center, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Juan Francisco Morales
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, Intelligent Critical Care Center, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Nicholas Waddington
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, Intelligent Critical Care Center, College of Pharmacy, University of Florida, Orlando, FL, USA
| | - Damon G. Lamb
- Departments of Psychiatry, Neuroscience, Biomedical Engineering, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville, FL, USA
- Malcom Randall VAMC, Gainesville, FL, USA
| | - Huiwen Ju
- NVIDIA Corporation, Santa Clara, CA, USA
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sarah Kim
- Department of Pharmaceutics, Center for Pharmacometrics and Systems Pharmacology, Intelligent Critical Care Center, College of Pharmacy, University of Florida, Orlando, FL, USA
| |
Collapse
|
69
|
Park M, Lee H, Jang Y, Kim MJ, Cho Y, Liu SS, Lee J, Shim S, Jung HD, Seong H, Yang K. Macroencapsulation Device with Anti-inflammatory Membrane Modification Enhances Long-Term Viability and Function of Transplanted β Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:70218-70230. [PMID: 39665438 DOI: 10.1021/acsami.4c14057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Treating type 1 diabetes (T1D) through β-cell macroencapsulation is a promising long-term solution, but it faces challenges such as immune-mediated fibrosis on the capsule surface, which impairs cell functionality and compromises longevity and effectiveness. This study presents an approach for including an anti-inflammatory molecule on the macroencapsulation device (MED) using initiated chemical vapor deposition for the surface modification of poly(tetrafluoroethylene) (PTFE) membranes. The surface-modified MEDs significantly reduced fibrosis, improved β-cell viability and functionality, and promoted M2 macrophage polarization, which is associated with anti-inflammatory effects. This MED displayed improved glycemic control in a streptozotocin-induced diabetic mouse model for 45 days. The findings underscore the potential of surface-modified MEDs for improving T1D management by mitigating inflammation and enhancing the therapeutic efficacy of β-cell encapsulation.
Collapse
Affiliation(s)
- MinJi Park
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Hyun Lee
- Research Institute of Intelligent Manufacturing & Materials Technology, Korea Institute of Industrial Technology, Incheon 21999, Republic of Korea
| | - Yerim Jang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Min Ji Kim
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sophie S Liu
- Department of Chemical Engineering, University of Toronto, Toronto ON M5S 3E5, Canada
| | - JungEun Lee
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Surim Shim
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Hyejeong Seong
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Kisuk Yang
- Department of Bioengineering and Nano-Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Division of Bioengineering, College of Life Sciences and Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
70
|
Wang YN, Li R, Huang Y, Chen H, Nie H, Liu L, Zou X, Zhong J, Zheng B, Gong Q. The role of B cells in the pathogenesis of type 1 diabetes. Front Immunol 2024; 15:1450366. [PMID: 39776900 PMCID: PMC11703732 DOI: 10.3389/fimmu.2024.1450366] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Type 1 diabetes (T1D) is a metabolic disorder caused by a complete lack of insulin, primarily manifested by hyperglycemia. The mechanisms underlying the onset of T1D are complex, involving genetics, environment, and various unknown factors, leading to the infiltration of various immune components into the islets. Besides T cells, B cells are now considered important contributors to the pathogenesis of T1D, according to recent studies. In non-obese diabetic (NOD) mice, the absence of B cells prevents the development of T1D, and B-cell depletion can even restore the function of pancreatic β cells, emphasizing their involvement in the development of T1D. Naturally, besides pathogenic B cells, regulatory B cells (Bregs) might have a protective function in T1D. This article examines the mechanisms behind B-cell tolerance and the defects in B-cell tolerance checkpoints in T1D. We explored possible functions of B cells in T1D, including the role of islet autoantibodies in T1D, T-B cell interactions, and the role of Bregs in the pathogenesis of T1D. We also summarized the advances of B cell-targeted therapy, exploring new methods for intervention and treatment of T1D.
Collapse
Affiliation(s)
- Ya-nan Wang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Ruihua Li
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Yaxuan Huang
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Hui Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Hao Nie
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Lian Liu
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Xiaoting Zou
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bing Zheng
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
71
|
Kim J, Shim IK, Lee YN, Kim M, Hwang DG, Kim J, Jo Y, Chae S, Kim J, Kim SC, Cho DW, Jang J. Engineering pore-enriched and pre-vascularized volumetric constructs for enhanced blood glucose regulation in type 1 diabetes therapy. Biofabrication 2024; 17:015034. [PMID: 39622163 DOI: 10.1088/1758-5090/ad998e] [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: 05/21/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Managing type 1 diabetes mellitus (T1DM) presents significant challenges because of the complexity of replicating the microenvironment of pancreatic islets and ensuring the long-term viability and function of transplanted insulin-producing cells (IPCs). This study developed a functional approach that utilizes 3D bioprinting technology to create pore-enriched and pre-vascularized tissue constructs incorporating a pancreatic tissue-derived decellularized extracellular matrix and human-induced pluripotent stem cells (hiPSCs) aimed at enhancing blood glucose regulation in T1DM. We designed a volumetric 3D pancreatic tissue construct that supported the engraftment, survival, and insulin-producing functionality of hiPSC-derived IPCs. The construct's porosity was optimized to enhance IPC delivery efficiency. Additionally, human umbilical vein endothelial cells co-cultured with IPCs in a patterned structure facilitated pre-vascularization, improving construct integration with host tissues and accelerating revascularization post-transplantation. Our results demonstrate high cell viability and sustained insulin production in diabetic rodent models, indicating the constructs' effectiveness in regulating blood glucose levels over an extended period. The findings of this study not only underscore the potential of 3D bioprinting for creating functional tissue constructs for T1DM treatment but also offer efficient cell delivery techniques applicable to other areas of regenerative medicine.
Collapse
Affiliation(s)
- Jaewook Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - In Kyong Shim
- Department of Biomedical Engineering, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Yu Na Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - Myungji Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Dong Gyu Hwang
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Jihwan Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | | | - Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Song Cheol Kim
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Brain Korea 21 Project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Center for 3D Organ Printing and Stem Cells, Pohang University of Science and Technology, Pohang, Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology, Pohang, Republic of Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
72
|
Altabas V, Bulum T. Current Challenges in Pancreas and Islet Transplantation: A Scoping Review. Biomedicines 2024; 12:2853. [PMID: 39767759 PMCID: PMC11673013 DOI: 10.3390/biomedicines12122853] [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: 10/22/2024] [Revised: 12/07/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Type 1 diabetes mellitus is an autoimmune condition characterized by the destruction of pancreatic β-cells, necessitating insulin therapy to prevent life-threatening complications such as diabetic ketoacidosis. Despite advancements in glucose monitoring and pharmacological treatments, managing this disease remains challenging, often leading to long-term complications and psychological burdens, including diabetes distress. Advanced treatment options, such as whole-pancreas transplantation and islet transplantation, aim to restore insulin production and improve glucose control in selected patients with diabetes. The risk of transplant rejection necessitates immunosuppressive therapy, which increases susceptibility to infections and other adverse effects. Additionally, surgical complications, including infection and bleeding, are significant concerns, particularly for whole-pancreas transplantation. Recently, stem cell-derived therapies for type 1 diabetes have emerged as a promising alternative, offering potential solutions to overcome the limitations of formerly established transplantation methods. The purpose of this scoping review was to: (1) summarize the current evidence on achieved insulin independence following various transplantation methods of insulin-producing cells in patients with type 1 diabetes; (2) compare insulin independence rates among whole-pancreas transplantation, islet cell transplantation, and stem cell transplantation; and (3) identify limitations, challenges and potential future directions associated with these techniques. We systematically searched three databases (PubMed, Scopus, and Web of Science) from inception to November 2024, focusing on English-language, peer-reviewed clinical studies. The search terms used were 'transplantation' AND 'type 1 diabetes' AND 'insulin independence'. Studies were included if they reported on achieved insulin independence, involved more than 10 patients with type 1 diabetes, and had a mean follow-up period of at least one year. Reviewers screened citations and extracted data on transplant type, study population size, follow-up duration, and insulin independence rates. We identified 1380 papers, and after removing duplicates, 705 papers remained for title and abstract screening. A total of 139 English-language papers were retrieved for full-text review, of which 48 studies were included in this review. The findings of this scoping review indicate a growing body of literature on transplantation therapy for type 1 diabetes. However, significant limitations and challenges, like insufficient rates of achieved insulin independence, risks related to immunosuppression, malignant diseases, and ethical issues remain with each of the established techniques, highlighting the need for innovative approaches such as stem cell-derived islet transplantation to promote β-cell regeneration and protection.
Collapse
Affiliation(s)
- Velimir Altabas
- Department of Endocrinology, Diabetes and Metabolic Diseases Mladen Sekso, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Tomislav Bulum
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Vuk Vrhovac University Clinic for Diabetes, Endocrinology and Metabolic Diseases, Merkur University Hospital, 10000 Zagreb, Croatia
| |
Collapse
|
73
|
Warnick JL, Darling KE, Swartz Topor L, Jelalian E. Barriers to healthy behaviors: perspectives from teens with comorbid Type 1 diabetes and overweight/obesity, caregivers, and pediatric endocrinologists. J Pediatr Psychol 2024; 49:874-881. [PMID: 39449679 DOI: 10.1093/jpepsy/jsae086] [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: 02/28/2024] [Revised: 09/16/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
OBJECTIVES This study aimed to understand barriers to engagement in healthy lifestyle behaviors among adolescents with comorbid Type 1 diabetes (T1D) and overweight/obesity (n = 12), their caregivers (n = 12), and pediatric endocrinologists (n = 9). METHODS Participants (n = 33) completed individual, semi-structured interviews. Interviews were transcribed verbatim, and applied thematic analysis was used to analyze the interview data. RESULTS Results from adolescents, caregivers, and pediatric endocrinologists revealed three thematic barriers to healthy lifestyle behaviors for adolescents with T1D and overweight/obesity: (1) discomfort with aspects of T1D that draw attention to the self; (2) T1D as a barrier to engagement in healthy lifestyle behaviors; and (3) physiological dysregulation due to T1D impacting health behaviors. CONCLUSIONS Results identify perceived limitations to engaging in recommended healthy lifestyle behaviors and diabetes management concurrently. Results may assist research and clinical care in identifying supports and guidance needed to support adolescents in meeting behavioral recommendations for their health.
Collapse
Affiliation(s)
- Jennifer L Warnick
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, United States
- The Weight Control and Diabetes Research Center, The Miriam Hospital, Providence, RI, United States
| | - Katherine E Darling
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, United States
- The Weight Control and Diabetes Research Center, The Miriam Hospital, Providence, RI, United States
| | - Lisa Swartz Topor
- Department of Pediatrics, Alpert Medical School of Brown University, Providence, RI, United States
| | - Elissa Jelalian
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, RI, United States
- The Weight Control and Diabetes Research Center, The Miriam Hospital, Providence, RI, United States
| |
Collapse
|
74
|
Lee JM, Pop-Busui R, Lee JM, Fleischer J, Wiens J. Shortcomings in the Evaluation of Blood Glucose Forecasting. IEEE Trans Biomed Eng 2024; 71:3424-3431. [PMID: 38990742 PMCID: PMC11724010 DOI: 10.1109/tbme.2024.3424665] [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] [Indexed: 07/13/2024]
Abstract
OBJECTIVE Recent years have seen an increase in machine learning (ML)-based blood glucose (BG) forecasting models, with a growing emphasis on potential application to hybrid or closed-loop predictive glucose controllers. However, current approaches focus on evaluating the accuracy of these models using benchmark data generated under the behavior policy, which may differ significantly from the data the model may encounter in a control setting. This study challenges the efficacy of such evaluation approaches, demonstrating that they can fail to accurately capture an ML-based model's true performance in closed-loop control settings. METHODS Forecast error measured using current evaluation approaches was compared to the control performance of two forecasters - a ML-based model (LSTM) and a rule-based model (Loop) - in silico when the forecasters were utilized with a model-based controller in a hybrid closed-loop setting. RESULTS Under current evaluation standards, LSTM achieves a significantly lower (better) forecast error than Loop with a root mean squared error (RMSE) of vs. at the 30-minute prediction horizon. Yet in a control setting, LSTM led to significantly worse control performance with only 77.14% (IQR 66.57-84.03) time-in-range compared to 86.20% (IQR 78.28-91.21) for Loop. CONCLUSION Prevailing evaluation methods can fail to accurately capture the forecaster's performance when utilized in closed-loop settings. SIGNIFICANCE Our findings underscore the limitations of current evaluation standards and the need for alternative evaluation metrics and training strategies when developing BG forecasters for closed-loop control systems.
Collapse
|
75
|
Ning X, Munir KM, Davis SN. Drugs stimulating insulin secretion in early clinical development for the treatment of type 1 diabetes: what's new? Expert Opin Investig Drugs 2024; 33:1199-1208. [PMID: 39645243 DOI: 10.1080/13543784.2024.2439501] [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: 01/25/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Type 1 diabetes is a chronic autoimmune condition characterized by the selective destruction of insulin-producing beta cells in the pancreas. The etiology of T1D is multifactorial, with a combination of genetic susceptibility and environmental triggers believed to underlie beta-cell destruction. Preserving and prolonging beta-cell function in T1D is a pivotal therapeutic objective that can mitigate disease progression and improve glycemic control. AREAS COVERED Insulin secretagogues have long been used in the management of type 2 diabetes, but do not have a significant beneficial effect in individuals with long-standing type 1 diabetes. Enhancement of beta-cell function early in the course of type 1 diabetes may offer important benefits in glycemic control and reduced hypoglycemia risk. Glucagon-like peptide-1 receptor agonists, glucokinase activators, free fatty acid receptor agonists, and glimins are drug classes which may offer benefit in enhancing insulin secretion in individuals with type 1 diabetes. EXPERT OPINION Drugs which enhance insulin secretion in individuals may offer clinical benefits to individuals with type 1 diabetes. However, the lack of beta-cell capacity introduces a challenge without regeneration of insulin-producing cells. Stem cell therapies combined with regulation of islet autoimmunity may offer the best prospect of increased insulin secretion in individuals with T1D.
Collapse
Affiliation(s)
- Xinyuan Ning
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Kashif M Munir
- Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Center for Diabetes and Endocrinology, Baltimore, MD, USA
| | - Stephen N Davis
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
76
|
Wych J, Brunner M, Stenson R, Chmura PJ, Danne T, Mander AP, Mathieu C, Dayan C, Pieber TR. Investigating the effect of verapamil on preservation of beta-cell function in adults with newly diagnosed type 1 diabetes mellitus (Ver-A-T1D): protocol for a randomised, double-blind, placebo-controlled, parallel-group, multicentre trial. BMJ Open 2024; 14:e091597. [PMID: 39613428 PMCID: PMC11605811 DOI: 10.1136/bmjopen-2024-091597] [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: 07/24/2024] [Accepted: 10/25/2024] [Indexed: 12/01/2024] Open
Abstract
INTRODUCTION Type 1 diabetes mellitus (T1DM) is a disorder that arises following the selective autoimmune destruction of the insulin-producing beta cells. Beta-cell protective or beta-cell regenerative approaches have gained wider attention, and pharmacological approaches to protect the patient's own insulin-producing beta-cell mass have been proposed. Verapamil is an L-type calcium channel blocker that has been reported to effectively lowers beta-cell thioredoxin-interacting protein expression in rodent beta cells and islets, as well as in human islets, and thus promotes functional beta-cell mass. METHODS AND ANALYSIS The trial is a multicentre, randomised, double-blind, placebo-controlled trial in participants with T1DM, investigating the effect of verapamil on preservation of beta-cell function (Ver-A-T1D). A total of 120 participants will be randomised in a 2:1 ratio between 360 mg verapamil and placebo, administered orally once daily. T1DM patients aged ≥18 and <45 years will be eligible for recruitment within 6 weeks of diagnosis (defined as day of starting insulin therapy). The primary objective will be to determine the changes in stimulated C-peptide response during the first 2 hours of a mixed meal tolerance test at baseline and after 12 months for 360 mg verapamil administered orally once daily versus placebo. Secondary objectives include the effects of 360 mg verapamil on (1) fasting C-peptide, (2) dried blood spot C-peptide, (3) glycated haemoglobin, (4) daily total insulin dose, (5) time in range by intermittent continuous glucose monitoring measures, (6) other biomarkers related to immunological changes and beta-cell death and (6) safety (vital signs, ECG). ETHICS AND DISSEMINATION Ethics approval was sought from the research ethics committee of all participating countries. All participants provided written informed consent before joining the study. Ver-A-T1D received first regulatory and ethical approvals in Austria. The publication policy is set in the innovative approach towards understanding and arresting type 1 diabetes grant agreement (www.innodia.eu). TRIAL REGISTRATION NUMBER EudraCT, 2020-000435-45; ClinicalTrials.gov, NCT04545151. PROTOCOL VERSION Version 8.0 (08 November 2021).
Collapse
Affiliation(s)
- Julie Wych
- Centre for Trials Research, Cardiff University, Cardiff, UK
| | - Martina Brunner
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| | - Rachel Stenson
- Diabetes Research Group, Cardiff University, Cardiff, UK
| | - Piotr Jaroslaw Chmura
- Center for Protein Research, Kobenhavns Universitet Sundhedsvidenskabelige Fakultet, Kobenhavn, Denmark
| | - Thomas Danne
- Department of General Pediatrics Endocrinology/Diabetology & Clinical Research, Auf der Bult Children's Hospital, Hannover, Germany
| | | | | | - Colin Dayan
- Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| | - Thomas R Pieber
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Medical University of Graz, Graz, Austria
| |
Collapse
|
77
|
Antikainen AA, Mutter S, Harjutsalo V, Thorn LM, Groop PH, Sandholm N. Urinary metabolomics provide insights into coronary artery disease in individuals with type 1 diabetes. Cardiovasc Diabetol 2024; 23:425. [PMID: 39593124 PMCID: PMC11590341 DOI: 10.1186/s12933-024-02512-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Type 1 diabetes increases the risk of coronary artery disease (CAD). High-throughput metabolomics may be utilized to identify metabolites associated with disease, thus, providing insight into disease pathophysiology, and serving as predictive markers in clinical practice. Urine is less tightly regulated than blood, and therefore, may enable earlier discovery of disease-associated markers. We studied urine metabolomics in relation to incident CAD in individuals with type 1 diabetes. METHODS We prospectively studied CAD in 2501 adults with type 1 diabetes from the Finnish Diabetic Nephropathy Study. 209 participants experienced incident CAD within the 10-year follow-up. We analyzed the baseline urine samples with a high-throughput targeted urine metabolomics platform, which yielded 54 metabolites. With the data, we performed metabolome-wide survival analyses, correlation network analyses, and metabolomic state profiling for prediction of incident CAD. RESULTS Urinary 3-hydroxyisobutyrate was associated with decreased 10-year incident CAD, which according to the network analysis, likely reflects younger age and improved kidney function. Urinary xanthosine was associated with 10-year incident CAD. In the network analysis, xanthosine correlated with baseline urinary allantoin, which is a marker of oxidative stress. In addition, urinary trans-aconitate and 4-deoxythreonate were associated with decreased 5-year incident CAD. Metabolomic state profiling supported the usage of CAD-associated urinary metabolites to improve prediction accuracy, especially during shorter follow-up. Furthermore, urinary trans-aconitate and 4-deoxythreonate were associated with decreased 5-year incident CAD. The network analysis further suggested glomerular filtration rate to influence the urinary metabolome differently between individuals with and without future CAD. CONCLUSIONS We have performed the first high-throughput urinary metabolomics analysis on CAD in individuals with type 1 diabetes and found xanthosine, 3-hydroxyisobutyrate, trans-aconitate, and 4-deoxythreonate to be associated with incident CAD. In addition, metabolomic state profiling improved prediction of incident CAD.
Collapse
Affiliation(s)
- Anni A Antikainen
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Stefan Mutter
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Valma Harjutsalo
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
| | - Lena M Thorn
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, 00014, Helsinki, Finland
| | - Per-Henrik Groop
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
| | - Niina Sandholm
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, 00290, Helsinki, Finland.
- Department of Nephrology, University of Helsinki and Helsinki University Hospital, 00290, Helsinki, Finland.
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290, Helsinki, Finland.
| |
Collapse
|
78
|
Carroll J, Chen J, Mittal R, Lemos JRN, Mittal M, Juneja S, Assayed A, Hirani K. Decoding the Significance of Alpha Cell Function in the Pathophysiology of Type 1 Diabetes. Cells 2024; 13:1914. [PMID: 39594662 PMCID: PMC11593172 DOI: 10.3390/cells13221914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Alpha cells in the pancreas, traditionally known for their role in secreting glucagon to regulate blood glucose levels, are gaining recognition for their involvement in the pathophysiology of type 1 diabetes (T1D). In T1D, autoimmune destruction of beta cells results in insulin deficiency, which in turn may dysregulate alpha cell function, leading to elevated glucagon levels and impaired glucose homeostasis. This dysfunction is characterized by inappropriate glucagon secretion, augmenting the risk of life-threatening hypoglycemia. Moreover, insulin deficiency and autoimmunity alter alpha cell physiological responses, further exacerbating T1D pathophysiology. Recent studies suggest that alpha cells undergo transdifferentiation and interact with beta cells through mechanisms involving gamma-aminobutyric acid (GABA) signaling. Despite these advances, the exact pathways and interactions remain poorly understood and are often debated. Understanding the precise role of alpha cells in T1D is crucial, as it opens up avenues for developing new therapeutic strategies for T1D. Potential strategies include targeting alpha cells to normalize glucagon secretion, utilizing glucagon receptor antagonists, enhancing GABA signaling, and employing glucagon-like peptide-1 (GLP-1) receptor agonists. These approaches aim to improve glycemic control and reduce the risk of hypoglycemic events in individuals with T1D. This review provides an overview of alpha cell function in T1D, highlighting the emerging focus on alpha cell dysfunction in the context of historically well-developed beta cell research.
Collapse
Affiliation(s)
| | | | - Rahul Mittal
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.C.); (J.C.); (J.R.N.L.); (M.M.); (S.J.); (A.A.)
| | | | | | | | | | - Khemraj Hirani
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.C.); (J.C.); (J.R.N.L.); (M.M.); (S.J.); (A.A.)
| |
Collapse
|
79
|
Su Z, Bian L, Zhao H, Yang C, Gu Y, Cai Y, Yang T, Xu X. KIR2DL5 +CD8 + T cells associate with dietary lipid intake and are active in type 1 diabetes. Int Immunopharmacol 2024; 141:112971. [PMID: 39178517 DOI: 10.1016/j.intimp.2024.112971] [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: 08/06/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
BACKGROUND Recent studies have shown that KIR+CD8+ T cells play a role in suppressing autoimmunity by eliminating pathogenic CD4+ T cells. However, their specific role in type 1 diabetes (T1D) remains unclear. METHODS In this study, we enrolled 108 patients diagnosed with T1D and 86 healthy individuals. We conducted flow cytometric analysis to examine the various subtypes of KIR+CD8+ T cells derived from peripheral blood mononuclear cells. Additionally, CD8+ T cells were isolated from the peripheral blood of T1D patients to assess the functions of different KIR+CD8+ T cell subtypes. To investigate the influence of lipids on the characteristics and activities of these T cell subtypes, the isolated CD8+ T cells were cultured with varying concentrations of palmitic acid (PA). Furthermore, we utilized an NSG (NOD scid gamma) mouse adoptive transfer model to assess the impact of dietary lipid intake on the functionality of KIR2DL5+CD8+ T cells in vivo. RESULTS We observed variations in circulating KIR+CD8+ T cell subtypes between patients with T1D and healthy controls. Notably, we observed a significant negative correlation between the frequencies of circulating KIR+CD8+ T cells and the titers of ZnT8 autoantibodies in individuals with T1D. Among these subtypes, KIR2DL5+CD8+ T cells demonstrated a positive association with dietary fat intake, characterized by increased perforin expression and reduced PD-1 expression. Importantly, KIR2DL5+CD8+ T cells exhibited enhanced proliferative capacity compared to other KIR+CD8+ T cell subsets. Palmitic acid (PA) was found to enhance the activation of KIR2DL5+CD8+ T cells and strengthened their ability to suppress CD4+ T cell proliferation in T1D patients. Moreover, dietary lipid intake significantly enhanced the functionality of KIR2DL5+CD8+ T cells in an NSG mouse adoptive transfer model. CONCLUSION Our findings suggest that lipid intake enhances the functionality of human KIR2DL5+CD8+ T cells and may offer implications for immunotherapy in T1D.
Collapse
Affiliation(s)
- Zhangyao Su
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Lingling Bian
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China; Department of Endocrinology, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, China
| | - Hang Zhao
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Chun Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yong Gu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yun Cai
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Tao Yang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Xinyu Xu
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
80
|
Chuang ST, Alcazar O, Watts B, Abdulreda MH, Buchwald P. Small-molecule inhibitors of the CD40-CD40L costimulatory interaction are effective in pancreatic islet transplantation and prevention of type 1 diabetes models. Front Immunol 2024; 15:1484425. [PMID: 39606229 PMCID: PMC11599200 DOI: 10.3389/fimmu.2024.1484425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
As part of our work to develop small-molecule inhibitors (SMIs) of the CD40-CD40L(CD154) costimulatory protein-protein interaction, here, we describe the ability of two of our most promising SMIs, DRI-C21041 and DRI-C21095, to prolong the survival and function of islet allografts in two murine models of islet transplantation (under the kidney capsule and in the anterior chamber of the eye) and to prevent autoimmune type 1 diabetes (T1D) onset in NOD mice. In both transplant models, a significant portion of islet allografts (50%-80%) remained intact and functional long after terminating treatment, suggesting the possibility of inducing operational immune tolerance via inhibition of the CD40-CD40L axis. SMI-treated mice maintained the structural integrity and function of their islet allografts with concomitant reduction in immune cell infiltration as evidenced by direct longitudinal imaging in situ. Furthermore, in female NODs, three-month SMI treatment reduced the incidence of diabetes from 80% to 60% (DRI-C21041) and 25% (DRI-C21095). These results (i) demonstrate the susceptibility of this TNF superfamily protein-protein interaction to small-molecule inhibition, (ii) confirm the in vivo therapeutic potential of these SMIs of a critical immune checkpoint, and (iii) reaffirm the therapeutic promise of CD40-CD40L blockade in islet transplantation and T1D prevention. Thus, CD40L-targeting SMIs could ultimately lead to alternative immunomodulatory therapeutics for transplant recipients and prevention of autoimmune diseases that are safer, less immunogenic, more controllable (shorter half-lives), and more patient-friendly (i.e., suitable for oral administration, which makes them easier to administer) than corresponding antibody-based interventions.
Collapse
Affiliation(s)
- Sung-Ting Chuang
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Oscar Alcazar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brandon Watts
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Midhat H. Abdulreda
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
81
|
Laptev DN, Bezlepkina OB, Sheshko EL, Aleksandrova GA, Chumakova OV, Krestovskaya NM, Kulaev AS, Peterkova VA. [Main epidemiological indicators of type 1 diabetes mellitus in children in the Russian Federation for 2014-2023]. PROBLEMY ENDOKRINOLOGII 2024; 70:76-83. [PMID: 39509639 DOI: 10.14341/probl13515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/29/2024] [Indexed: 11/15/2024]
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) is the most common form of diabetes mellitus in childhood, where, unlike in adults, it accounts for more than 90% of all cases of diabetes. The constant change in the epidemiology of T1DM with significant differences in populations and regions requires systematic data collection and analysis for timely monitoring of T1DM trends. AIM Analysis of the main epidemiological indicators of T1DM in children in the Russian Federation over the past 10 years - from 2014 to 2023. MATERIALS AND METHODS The object of the study was the data obtained from the federal statistical observation form No. 12 «Information on the number of diseases registered in patients living in the service area of a medical organization» for the period from 2014 to 2023. The prevalence (total number of registered cases) and incidence (cases with a diagnosis established for the first time) of T1DM (ICD-10 code: E10) were analyzed in children in three age groups: from 0 to 14 years, from 15 to 17 years, and combined from 0 to 17 years (inclusive). RESULTS Over the analyzed period, the prevalence of T1DM increased steadily from 238.6 in 2014 to 374.2 cases per 100,000 children in 2023. The prevalence of T1DM in adolescents from 15 to 17 years was higher than in children and amounted to 120.3-203.2 cases per 100,000 adolescents, while in children under 14 years of age, the prevalence was 100.1-172.2 cases per 100,000 children. The annual increase in the prevalence of T1DM averaged 6.3% (95% CI 4.9-7.8). The incidence of T1DM during the analyzed period was 19.1-27.2 cases per 100,000 children and also had a general tendency toward an annual increase in new cases. At the same time, over the past three years, there has been a relative stabilization of incidence rates at 26.5-27.2 per 100,000 children. The annual increase in incidence averaged 4.9% (95% CI 0.9-8.9). The greatest increase in the incidence of T1DM was observed in regions with low incidence. CONCLUSION The epidemiology of T1DM in the Russian Federation is characterized by significant regional and dynamic changes. Over the period 2014-2023, the incidence of type 1 diabetes in children has increased significantly, increasing annually by an average of 5%, while there has been a relative stabilization of incidence rates over the past three years.
Collapse
Affiliation(s)
| | | | - E L Sheshko
- Department of Medical Care for Children, Maternity Services and Public Health
| | - G A Aleksandrova
- Department of Monitoring, Analysis and Strategic Development of Healthcare
| | - O V Chumakova
- Department of Medical Care for Children, Maternity Services and Public Health
| | - N M Krestovskaya
- Department of Medical Care for Children, Maternity Services and Public Health
| | - A Sh Kulaev
- Department of Medical Care for Children, Maternity Services and Public Health
| | | |
Collapse
|
82
|
Wong JM, Pepper AR. Status of islet transplantation and innovations to sustainable outcomes: novel sites, cell sources, and drug delivery strategies. FRONTIERS IN TRANSPLANTATION 2024; 3:1485444. [PMID: 39553396 PMCID: PMC11565603 DOI: 10.3389/frtra.2024.1485444] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024]
Abstract
Islet transplantation (ITx) is an effective means to restore physiologic glycemic regulation in those living with type 1 diabetes; however, there are a handful of barriers that prevent the broad application of this functionally curative procedure. The restricted cell supply, requisite for life-long toxic immunosuppression, and significant immediate and gradual graft attrition limits the procedure to only those living with brittle diabetes. While intraportal ITx is the primary clinical site, portal vein-specific factors including low oxygen tension and the instant blood-mediated inflammatory reaction are detrimental to initial engraftment and long-term function. These factors among others prevent the procedure from granting recipients long-term insulin independence. Herein, we provide an overview of the status and limitations of ITx, and novel innovations that address the shortcomings presented. Despite the marked progress highlighted in the review from as early as the initial islet tissue transplantation in 1893, ongoing efforts to improve the procedure efficacy and success are also explored. Progress in identifying unlimited cell sources, more favourable transplant sites, and novel drug delivery strategies all work to broaden ITx application and reduce adverse outcomes. Exploring combination of these approaches may uncover synergies that can further advance the field of ITx in providing sustainable functional cures. Finally, the potential of biomaterial strategies to facilitate immune evasion and local immune modulation are featured and may underpin successful application in alternative transplant sites.
Collapse
Affiliation(s)
| | - Andrew R. Pepper
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
83
|
Vasileva LA, Gaynanova GA, Romanova EA, Petrov KA, Feng C, Zakharova LY, Sinyashin OG. Supramolecular approach to the design of nanocarriers for antidiabetic drugs: targeted patient-friendly therapy. RUSSIAN CHEMICAL REVIEWS 2024; 93:RCR5150. [DOI: 10.59761/rcr5150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Diabetes and its complications derived are among serious global health concerns that critically deteriorate the quality of life of patients and, in some cases, result in lethal outcome. Herein, general information on the pathogenesis, factors aggravating the course of the disease and drugs used for the treatment of two types of diabetes are briefly discussed. The aim of the review is to introduce supramolecular strategies that are currently being developed for the treatment of diabetes mellitus and that present a very effective alternative to chemical synthesis, allowing the fabrication of nanocontainers with switchable characteristics that meet the criteria of green chemistry. Particular attention is paid to organic (amphiphilic and polymeric) formulations, including those of natural origin, due to their biocompatibility, low toxicity, and bioavailability. The advantages and limitations of different nanosystems are discussed, with emphasis on their adaptivity to noninvasive administration routes.<br>The bibliography includes 378 references.
Collapse
Affiliation(s)
- L. A. Vasileva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - G. A. Gaynanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - E. A. Romanova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - K. A. Petrov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - Ch. Feng
- Shanghai Jiao Tong University, Shanghai, China
| | - L. Ya. Zakharova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| | - O. G. Sinyashin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russian Federation
| |
Collapse
|
84
|
You L, Ferrat LA, Oram RA, Parikh HM, Steck AK, Krischer J, Redondo MJ, Type 1 Diabetes TrialNet Study Group. Identification of type 1 diabetes risk phenotypes using an outcome-guided clustering analysis. Diabetologia 2024; 67:2507-2517. [PMID: 39103721 PMCID: PMC12103260 DOI: 10.1007/s00125-024-06246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024]
Abstract
AIMS/HYPOTHESIS Although statistical models for predicting type 1 diabetes risk have been developed, approaches that reveal the heterogeneity of the at-risk population by identifying clinically meaningful clusters are lacking. We aimed to identify and characterise clusters of islet autoantibody-positive individuals who share similar characteristics and type 1 diabetes risk. METHODS We tested a novel outcome-guided clustering method in initially non-diabetic autoantibody-positive relatives of individuals with type 1 diabetes, using the TrialNet Pathway to Prevention study data (n=1123). The outcome of the analysis was the time to development of type 1 diabetes, and variables in the model included demographic characteristics, genetics, metabolic factors and islet autoantibodies. An independent dataset (the Diabetes Prevention Trial of Type 1 Diabetes Study) (n=706) was used for validation. RESULTS The analysis revealed six clusters with varying type 1 diabetes risks, categorised into three groups based on the hierarchy of clusters. Group A comprised one cluster with high glucose levels (median for glucose mean AUC 9.48 mmol/l; IQR 9.16-10.02) and high risk (2-year diabetes-free survival probability 0.42; 95% CI 0.34, 0.51). Group B comprised one cluster with high IA-2A titres (median 287 DK units/ml; IQR 250-319) and elevated autoantibody titres (2-year diabetes-free survival probability 0.73; 95% CI 0.67, 0.80). Group C comprised four lower-risk clusters with lower autoantibody titres and glucose levels (with 2-year diabetes-free survival probability ranging from 0.84-0.99 in the four clusters). Within group C, the clusters exhibit variations in characteristics such as glucose levels, C-peptide levels and age. A decision rule for assigning individuals to clusters was developed. Use of the validation dataset confirmed that the clusters can identify individuals with similar characteristics. CONCLUSIONS/INTERPRETATION Demographic, metabolic, immunological and genetic markers may be used to identify clusters of distinctive characteristics and different risks of progression to type 1 diabetes among autoantibody-positive individuals with a family history of type 1 diabetes. The results also revealed the heterogeneity in the population and complex interactions between variables.
Collapse
Affiliation(s)
- Lu You
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
| | - Lauric A Ferrat
- Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
- Faculty of Medicine, Department of Genetic Medicine and Development, University of Geneva, Geneva, Switzerland
| | - Richard A Oram
- Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Hemang M Parikh
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Andrea K Steck
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jeffrey Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Maria J Redondo
- Baylor College of Medicine, Texas Children's Hospital, Houston, TX, USA
| | | |
Collapse
|
85
|
Kelly CS, Nguyen H, Chapman KS, Wolf WA. The emotional burden of type 1 diabetes: A cross-sectional study to understand associations between diabetes distress and glucose metrics in adulthood. Diabet Med 2024; 41:e15425. [PMID: 39149967 DOI: 10.1111/dme.15425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/01/2024] [Indexed: 08/17/2024]
Abstract
AIMS Advancements in type 1 diabetes (T1D) management, such as continuous glucose monitoring (CGM), have helped people achieve narrower glucose ranges, but associations between CGM and diabetes distress are unclear. Although higher HbA1c is associated with higher distress, associations with other glucose metrics are unknown. To better understand this relationship, we characterized diabetes distress in a sample of CGM users and compared differences in glucose metrics (measured via CGM) between those with higher versus lower distress. METHODS CGM users with T1D from the T1D Exchange Registry completed an online survey including diabetes distress (DDS-2) and shared CGM data (N = 199). CGM metrics were computed from all available data within 3 months prior to survey completion. Participants were grouped by distress level: lower (DDS-2 < 3, n = 120) or higher (DDS-2 ≥ 3, n = 79). Welch's t-tests were used to compare mean differences in CGM metrics between groups and MANCOVA was used to further probe mean differences. RESULTS Approximately 39.7% participants reported higher diabetes distress. Welch's t-tests revealed participants with higher distress spent significantly more time in higher glucose ranges (above 180 mg/dL and above 250 mg/dL), less time in target glucose ranges (between 70 and 180 mg/dL and between 70 and 140 mg/dL) and had higher glucose management index values compared to those with lower distress (p < 0.01). MANCOVA models showed similar results. CONCLUSIONS CGM users continue to experience diabetes distress. Moreover, higher distress appears to be associated with hyperglycaemia. These findings provide support for broader screening efforts for diabetes distress.
Collapse
|
86
|
Rundell TB, Baranski TJ. Insect Models to Study Human Lipid Metabolism Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39405006 DOI: 10.1007/5584_2024_827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Disorders of lipid metabolism such as obesity have become some of the most significant diseases of the twenty-first century. Despite these metabolic diseases affecting more than a third of the population in highly industrialized nations, the mechanisms underlying disease development remain poorly understood. Insect models, such as Drosophila melanogaster, offer a means of systematically examining conserved lipid metabolism and its pathology. Over the past several decades, Drosophila melanogaster has been used to greatly expand on our knowledge of metabolic disease, often taking advantage of the extensive genetic tools available to researchers. Additionally, Drosophila melanogaster has served and will continue to serve as a powerful tool for validating the results of genome-wide approaches to the study of diseases. This chapter explores the advancements of insect models in the study of lipid metabolism disorders as well as highlight opportunities for future areas of research.
Collapse
Affiliation(s)
- Thomas B Rundell
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Thomas J Baranski
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
87
|
Maestas MM, Bui MH, Millman JR. Recent progress in modeling and treating diabetes using stem cell-derived islets. Stem Cells Transl Med 2024; 13:949-958. [PMID: 39159002 PMCID: PMC11465181 DOI: 10.1093/stcltm/szae059] [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: 05/02/2024] [Accepted: 07/12/2024] [Indexed: 08/21/2024] Open
Abstract
Stem cell-derived islets (SC-islets) offer the potential to be an unlimited source of cells for disease modeling and the treatment of diabetes. SC-islets can be genetically modified, treated with chemical compounds, or differentiated from patient derived stem cells to model diabetes. These models provide insights into disease pathogenesis and vulnerabilities that may be targeted to provide treatment. SC-islets themselves are also being investigated as a cell therapy for diabetes. However, the transplantation process is imperfect; side effects from immunosuppressant use have reduced SC-islet therapeutic potential. Alternative methods to this include encapsulation, use of immunomodulating molecules, and genetic modification of SC-islets. This review covers recent advances using SC-islets to understand different diabetes pathologies and as a cell therapy.
Collapse
Affiliation(s)
- Marlie M Maestas
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Maggie H Bui
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Jeffrey R Millman
- Roy and Diana Vagelos Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO 63110, United States
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
88
|
Fogarasi M, Dima S. Immunomodulatory Functions of TNF-Related Apoptosis-Inducing Ligand in Type 1 Diabetes. Cells 2024; 13:1676. [PMID: 39451194 PMCID: PMC11506310 DOI: 10.3390/cells13201676] [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: 09/02/2024] [Revised: 10/01/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF protein superfamily and was initially identified as a protein capable of inducing apoptosis in cancer cells. In addition, TRAIL can promote pro-survival and proliferation signaling in various cell types. Subsequent studies have demonstrated that TRAIL plays several important roles in immunoregulation, immunosuppression, and immune effector functions. Type 1 diabetes (T1D) is an autoimmune disease characterized by hyperglycemia due to the loss of insulin-producing β-cells, primarily driven by T-cell-mediated pancreatic islet inflammation. Various genetic, epigenetic, and environmental factors, in conjunction with the immune system, contribute to the initiation, development, and progression of T1D. Recent reports have highlighted TRAIL as an important immunomodulatory molecule with protective effects on pancreatic islets. Experimental data suggest that TRAIL protects against T1D by reducing the proliferation of diabetogenic T cells and pancreatic islet inflammation and restoring normoglycemia in animal models. In this review, we aimed to summarize the consequences of TRAIL action in T1D, focusing on and discussing its signaling mechanisms, role in the immune system, and protective effects in T1D.
Collapse
Affiliation(s)
- Marton Fogarasi
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Simona Dima
- Center of Excellence in Translational Medicine, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| |
Collapse
|
89
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
90
|
Luppi S, Aldegheri L, Azzalini E, Pacetti E, Barucca Sebastiani G, Fabiani C, Robino A, Comar M. Unravelling the Role of Gut and Oral Microbiota in the Pediatric Population with Type 1 Diabetes Mellitus. Int J Mol Sci 2024; 25:10611. [PMID: 39408940 PMCID: PMC11477131 DOI: 10.3390/ijms251910611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Type 1 Diabetes Mellitus (T1DM) is a chronic autoimmune disease that results in the destruction of pancreatic β cells, leading to hyperglycaemia and the need for lifelong insulin therapy. Although genetic predisposition and environmental factors are considered key contributors to T1DM, the exact causes of the disease remain partially unclear. Recent evidence has focused on the relationship between the gut, the oral cavity, immune regulation, and systemic inflammation. In individuals with T1DM, changes in the gut and oral microbial composition are commonly observed, indicating that dysbiosis may contribute to immune dysregulation. Gut dysbiosis can influence the immune system through increased intestinal permeability, altered production of short chain fatty acids (SCFAs), and interactions with the mucosal immune system, potentially triggering the autoimmune response. Similarly, oral dysbiosis may contribute to the development of systemic inflammation and thus influence the progression of T1DM. A comprehensive understanding of these relationships is essential for the identification of biomarkers for early diagnosis and monitoring, as well as for the development of therapies aimed at restoring microbial balance. This review presents a synthesis of current research on the connection between T1DM and microbiome dysbiosis, with a focus on the gut and oral microbiomes in pediatric populations. It explores potential mechanisms by which microbial dysbiosis contributes to the pathogenesis of T1DM and examines the potential of microbiome-based therapies, including probiotics, prebiotics, synbiotics, and faecal microbiota transplantation (FMT). This complex relationship highlights the need for longitudinal studies to monitor microbiome changes over time, investigate causal relationships between specific microbial species and T1DM, and develop personalised medicine approaches.
Collapse
Affiliation(s)
- Stefania Luppi
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Luana Aldegheri
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Eros Azzalini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Emanuele Pacetti
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| | - Giulia Barucca Sebastiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Carolina Fabiani
- Medicine of Services Department, Clinical Analysis Laboratory, Azienda Sanitaria Universitaria Giuliano Isontina, 34125 Trieste, Italy; (G.B.S.); (C.F.)
| | - Antonietta Robino
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, 65/1 Via dell’Istria, 34137 Trieste, Italy; (S.L.); (L.A.); (M.C.)
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy; (E.A.); (E.P.)
| |
Collapse
|
91
|
Escobar Vasco MA, Fantaye SH, Raghunathan S, Solis-Herrera C. The potential role of finerenone in patients with type 1 diabetes and chronic kidney disease. Diabetes Obes Metab 2024; 26:4135-4146. [PMID: 39021345 DOI: 10.1111/dom.15773] [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: 04/05/2024] [Revised: 06/20/2024] [Accepted: 06/22/2024] [Indexed: 07/20/2024]
Abstract
Chronic kidney disease (CKD) represents a global health concern, associated with an increased risk of cardiovascular morbidity and mortality and decreased quality of life. Many patients with type 1 diabetes (T1D) will develop CKD over their lifetime. Uncontrolled glucose levels, which occur in patients with T1D as well as type 2 diabetes (T2D), are associated with substantial mortality and cardiovascular disease burden. T2D and T1D share common pathological features of CKD, which is thought to be driven by haemodynamic dysfunction, metabolic disturbances, and subsequently an influx of inflammatory and profibrotic mediators, both of which are major interrelated contributors to CKD progression. The mineralocorticoid receptor is also involved, and, under conditions of oxidative stress, salt loading and hyperglycaemia, it switches from homeostatic regulator to pathophysiological mediator by promoting oxidative stress, inflammation and fibrosis. Progressive glomerular and tubular injury leads to macroalbuminuria a progressive reduction in the glomerular filtration rate and eventually end-stage renal disease. Finerenone, a non-steroidal, selective mineralocorticoid receptor antagonist, is approved for treatment of patients with CKD associated with T2D; however, the benefit of finerenone in patients with T1D has yet to be determined. This narrative review will discuss treatment of CKD in T1D and the potential future role of finerenone in this setting.
Collapse
Affiliation(s)
| | - Samuel H Fantaye
- Division of Endocrinology, University of Texas Health, San Antonio, Texas, USA
| | - Sapna Raghunathan
- Division of Endocrinology, University of Texas Health, San Antonio, Texas, USA
| | | |
Collapse
|
92
|
Ling EM, Lemos JRN, Hirani K, von Herrath M. Type 1 diabetes: immune pathology and novel therapeutic approaches. Diabetol Int 2024; 15:761-776. [PMID: 39469552 PMCID: PMC11512973 DOI: 10.1007/s13340-024-00748-z] [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: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 10/30/2024]
Abstract
Type 1 diabetes (T1D) is characterized by the progressive destruction of insulin-producing beta cells in the pancreas. Despite improvements in insulin monitoring techniques, there remains no cure for T1D. Individuals with T1D require lifelong insulin therapy and some develop life-threatening complications. T1D is a complex, multifactorial, autoimmune condition. Understanding why people get T1D and how it progresses has advanced our knowledge of the disease and led to the discovery of specific targets that can be therapeutically manipulated to halt or reverse the course of T1D. Scientists investigating the potential of immunotherapy treatment for the treatment have recently had some encouraging results. Teplizumab, an anti-CD3 monoclonal antibody that has been approved by the FDA, delays the onset of clinical T1D in patients ≥ 8 years of age with preclinical T1D and improves beta cell function. Therapies targeting beta cell health, vitality, and function are now thought to be an essential component of successful combination therapy for T1D. The idea that the beta cells themselves may influence their own destruction during the development of T1D is a notion that has recently been gaining acceptance in the field. Researchers have recently made remarkable strides in beta cell replacement therapy and beta cell regeneration techniques. This review offers a detailed exploration of the pathophysiological mechanisms of T1D. It discusses the intricate interplay of factors leading to T1D development and the innovative approaches being explored to discover new treatments and a cure for the millions of people living with T1D worldwide.
Collapse
Affiliation(s)
- Eleanor M. Ling
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
| | - Joana R. N. Lemos
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
| | - Khemraj Hirani
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
| | - Matthias von Herrath
- Diabetes Research Institute (DRI), University of Miami Miller School of Medicine, Miami, FL USA
- Division of Endocrine, Diabetes, and Metabolism, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL USA
- Global Chief Medical Office, Novo Nordisk A/S, Søborg, Denmark
| |
Collapse
|
93
|
Debuysschere C, Nekoua MP, Alidjinou EK, Hober D. The relationship between SARS-CoV-2 infection and type 1 diabetes mellitus. Nat Rev Endocrinol 2024; 20:588-599. [PMID: 38890459 DOI: 10.1038/s41574-024-01004-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
Environmental factors, in particular viral infections, are thought to have an important role in the pathogenesis of type 1 diabetes mellitus (T1DM). The COVID-19 pandemic reinforced this hypothesis as many observational studies and meta-analyses reported a notable increase in the incidence of T1DM following infection with SARS-CoV-2 as well as an association between SARS-CoV-2 infection and the risk of new-onset T1DM. Experimental evidence suggests that human β-cells express SARS-CoV-2 receptors and that SARS-CoV-2 can infect and replicate in β-cells, resulting in structural or functional alterations of these cells. These alterations include reduced numbers of insulin-secreting granules, impaired pro-insulin (or insulin) secretion, and β-cell transdifferentiation or dedifferentiation. The inflammatory environment induced by local or systemic SARS-CoV-2 infection might result in a set of signals (such as pro-inflammatory cytokines) that lead to β-cell alteration or apoptosis or to a bystander activation of T cells and disruption of peripheral tolerance that triggers autoimmunity. Other mechanisms, such as viral persistence, molecular mimicry and activation of endogenous human retroviruses, are also likely to be involved in the pathogenesis of T1DM following SARS-CoV-2 infection. This Review addresses the issue of the involvement of SARS-CoV-2 infection in the development of T1DM using evidence from epidemiological, clinical and experimental studies.
Collapse
Affiliation(s)
- Cyril Debuysschere
- Université de Lille, CHU Lille, Laboratoire de virologie ULR3610, Lille, France
| | | | | | - Didier Hober
- Université de Lille, CHU Lille, Laboratoire de virologie ULR3610, Lille, France.
| |
Collapse
|
94
|
Liu Z, Schaeffer NE, Wang X. Differential signaling effects of blood glucose on delay discounting in individuals with and without type 1 diabetes. J Behav Med 2024; 47:839-848. [PMID: 38853222 DOI: 10.1007/s10865-024-00500-7] [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: 02/17/2024] [Accepted: 05/23/2024] [Indexed: 06/11/2024]
Abstract
Based on the signaling hypothesis of blood glucose (BG), a rise in BG levels signals a positive energy budget for healthy individuals but cellular starvation for individuals with type 1 diabetes. We examined this novel prediction and its intervention implications in the context of delay discounting, the degree to which delayed rewards are discounted, and the regulatory effects of insulin ingestion. We recruited 44 adults with type 1 diabetes (mean age 30.8 years, diabetes duration 15.4 years) and recorded their BG levels. The delay discounting rate was measured using the intertemporal choice task, where participants were required to choose between sets of smaller-and-sooner (SS) and larger-and-later (LL) rewards. In addition, 82 age-matched healthy participants were recruited to provide a baseline comparison on delay discounting. Random forest analysis showed that among many diagnostic factors, delay discounting was most dominating in differentiating the individuals with type 1 diabetes from the control participants. A hierarchical linear mixed model revealed that participants with type 1 diabetes had a stronger preference for SS rewards (p < .001) after controlling for covariates. Participants who had insulin delivered before the last meal exhibited a stronger preference for LL rewards compared to after-meal delivery. In contrast, subjective measures (e.g., self-reported hunger) failed to predict the participants' actual BG levels and delay discounting rates. In sum, individuals with type 1 diabetes tend to discount future rewards excessively compared to the control participants. Pre-meal insulin ingestion was associated with a higher LL preference for future rewards.
Collapse
Affiliation(s)
- Zheng Liu
- School of Humanities and Social Science, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China
| | | | - XiaoTian Wang
- School of Humanities and Social Science, The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
| |
Collapse
|
95
|
Bakery HH, Hussein HAA, Ahmed OM, Abuelsaad ASA, Khalil RG. The potential therapeutic role of IL-35 in pathophysiological processes in type 1 diabetes mellitus. Cytokine 2024; 182:156732. [PMID: 39126765 DOI: 10.1016/j.cyto.2024.156732] [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: 05/01/2024] [Revised: 07/01/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
A chronic autoimmune condition known as type 1 diabetes mellitus (T1DM) has characteristics marked by a gradual immune-mediated deterioration of the β-cells that produce insulin and causes overt hyperglycemia. it affects more than 1.2 million kids and teenagers (0-19 years old). In both, the initiation and elimination phases of T1DM, cytokine-mediated immunity is crucial in controlling inflammation. T regulatory (Treg) cells, a crucial anti-inflammatory CD4+ T cell subset, secretes interleukin-35 (IL-35). The IL-35 has immunomodulatory properties by inhibiting pro-inflammatory cells and cytokines, increasing the secretion of interleukin-10 (IL-10) as well as transforming Growth Factor- β (TGF-β), along with stimulating the Treg and B regulatory (Breg) cells. IL-35, it is a possible target for cutting-edge therapies for cancers, inflammatory, infectious, and autoimmune diseases, including TIDM. Unanswered questions surround IL-35's function in T1DM. Increasing data suggests Treg cells play a crucial role in avoiding autoimmune T1DM. Throughout this review, we will explain the biological impacts of IL-35 and highlight the most recently progresses in the roles of IL-35 in treatment of T1DM; the knowledge gathered from these findings might lead to the development of new T1DM treatments. This review demonstrates the potential of IL-35 as an effective autoimmune diabetes inhibitor and points to its potential therapeutic value in T1DM clinical trials.
Collapse
Affiliation(s)
- Heba H Bakery
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt
| | - Heba A A Hussein
- Faculty of Medicine, Egyptian Fellowship of Radiology, Beni-Suef University, Egypt
| | - Osama M Ahmed
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Egypt
| | | | - Rehab G Khalil
- Immunology Division, Faculty of Science, Beni-Suef University, Egypt.
| |
Collapse
|
96
|
Ye Z, Li L, Yang L, Zhuang L, Aspatwar A, Wang L, Gong W. Impact of diabetes mellitus on tuberculosis prevention, diagnosis, and treatment from an immunologic perspective. EXPLORATION (BEIJING, CHINA) 2024; 4:20230138. [PMID: 39439490 PMCID: PMC11491313 DOI: 10.1002/exp.20230138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 02/02/2024] [Indexed: 10/25/2024]
Abstract
The coexistence of diabetes mellitus (DM) and tuberculosis (TB) presents a significant global burden, with DM being recognized as a major risk factor for TB. This review comprehensively analyzes the immunological aspects of DM-TB comorbidity, shedding light on the impact of DM on TB pathogenesis and immune responses. It reveals that high blood glucose levels in TB patients contribute to reduced innate immune cell count, compromised phagocytic function, and delayed antigen presentation. These factors ultimately impair the clearance of Mycobacterium tuberculosis (MTB) and delay adaptive immune responses. With the interaction between TB and DM, there is an increase in inflammation and elevated secretion of pro-inflammatory cytokines by immune cells. This exacerbates the inflammatory response and contributes to poor treatment outcomes in TB. Moreover, the review explores the effects of DM on TB prevention, diagnosis, and treatment. It highlights how poor glycemic control, insulin resistance (IR), DM complications, and genetic factors increase the risk of MTB infection in individuals with DM. Additionally, DM-related immune suppression adversely affects the sensitivity of traditional diagnostic tests for TB, potentially resulting in underdiagnosis and delayed intervention. To mitigate the burden of TB in DM patients, the review emphasizes the need for further research on the mechanisms underlying DM reactivation in latent TB infection (LTBI). It shows how important it is to find and treat LTBI in DM patients as soon as possible and suggests looking into biomarkers that are specific to DM to make diagnosis more accurate.
Collapse
Affiliation(s)
- Zhaoyang Ye
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and TreatmentSenior Department of TuberculosisThe Eighth Medical Center of PLA General HospitalBeijingChina
- Hebei North UniversityZhangjiakouHebeiChina
- Department of GeriatricsThe Eighth Medical Center of PLA General HospitalBeijingChina
| | | | - Ling Yang
- Hebei North UniversityZhangjiakouHebeiChina
| | - Li Zhuang
- Hebei North UniversityZhangjiakouHebeiChina
| | - Ashok Aspatwar
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Liang Wang
- Department of GeriatricsThe Eighth Medical Center of PLA General HospitalBeijingChina
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and TreatmentSenior Department of TuberculosisThe Eighth Medical Center of PLA General HospitalBeijingChina
| |
Collapse
|
97
|
Fernandez Trigo N, Kalbermatter C, Yilmaz B, Ganal-Vonarburg SC. The protective effect of the intestinal microbiota in type-1 diabetes in NOD mice is limited to a time window in early life. Front Endocrinol (Lausanne) 2024; 15:1425235. [PMID: 39391872 PMCID: PMC11464356 DOI: 10.3389/fendo.2024.1425235] [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: 04/29/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Introduction The incidence of type-1 diabetes is on the rise, particularly in developed nations, and predominantly affects the youth. While genetic predisposition plays a substantial role, environmental factors, including alterations in the gut microbiota, are increasingly recognized as significant contributors to the disease. Methods In this study, we utilized germ-free non-obese diabetic mice to explore the effects of microbiota colonization during early life on type-1 diabetes susceptibility. Results Our findings reveal that microbiota introduction at birth, rather than at weaning, significantly reduces the risk of type-1 diabetes, indicating a crucial window for microbiota-mediated modulation of immune responses. This protective effect was independent of alterations in intestinal barrier function but correlated with testosterone levels in male mice. Additionally, early life colonization modulated T cell subset frequencies, particularly T helper cells and regulatory T cells, in the intestine, potentially shaping type-1 diabetes predisposition. Discussion Our findings underscore the pivotal role of early-life microbial interactions in immune regulation and the development of autoimmune diseases.
Collapse
Affiliation(s)
- Nerea Fernandez Trigo
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Cristina Kalbermatter
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Stephanie C. Ganal-Vonarburg
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
98
|
Hernández-Munive AK, Molina-Leonor MB, Ayala-González BD, Vázquez-Andrade J, Medina-Nieto A, Fernández-Guasti A. Diabetes mellitus and female sexual response: what do animal models tell us? Sex Med Rev 2024; 12:542-550. [PMID: 38879347 DOI: 10.1093/sxmrev/qeae044] [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: 02/19/2024] [Revised: 05/14/2024] [Accepted: 05/30/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND One of the less explored effects of diabetes mellitus (DM) is female sexual dysfunction. Females of different species have been used as models. AIM To analyze the information of animal models of DM and female sexual response (FSR). METHODS The literature of FSR in models of DM was reviewed. OUTCOMES Paradigm- and diabetes-dependent changes have been found in various aspects of the FSR. RESULTS Females in a type 1 DM (DM1) model show a decrease in the number of proestrus events, and ovariectomized females treated with sex hormones have been used. In these females, a reduction in lordosis has been reported; in proceptivity, the data are contradictory. These females present a decrease in sexual motivation that was restored after exogenous insulin. In the type 2 DM (DM2) model, females show regular estrous cycles, normal levels of lordosis behavior, and, depending on the paradigm, decreased proceptivity. These females display normal preference for sexually active males or their olfactory cues when having free physical contact; they lose this preference when tested in paradigms where physical interaction is precluded. CLINICAL TRANSLATION Preclinical data showing the high deleterious effects of a DM1 model and the less drastic effects under a DM2 model are in accordance with clinical data revealing a much higher prevalence of sexual dysfunction in women with DM1 than DM2. STRENGTHS AND LIMITATIONS The main strength is the analysis of the changes in various components of FSR in 2 models of DM. The main limitation is the difficulty in extrapolating the data on FSR from rats to women and that most studies focus on evaluating the impact of severe or chronic-moderate hyperglycemia/hyperinsulinemia on the sexual response, without considering other pathophysiologic alterations generated by DM. CONCLUSION Females with severe hyperglycemia have a decrease in FSR, while those with moderate hyperglycemia show much less drastic effects.
Collapse
Affiliation(s)
- Abigail Karina Hernández-Munive
- Departamento de Farmacobiología, Centro de investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 14330, México City, México
- División de Neurociencias, Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, 04510, México City, México
| | | | - Brenda Denisse Ayala-González
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Joanna Vázquez-Andrade
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Alberto Medina-Nieto
- Departamento de Sistemas Biológicos, Unidad Xochimilco, Universidad Autónoma Metropolitana, 04960 México City, México
| | - Alonso Fernández-Guasti
- Departamento de Farmacobiología, Centro de investigación y de Estudios Avanzados del Instituto Politécnico Nacional, 14330, México City, México
| |
Collapse
|
99
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590798. [PMID: 39345402 PMCID: PMC11429609 DOI: 10.1101/2024.04.23.590798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maria Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
100
|
Citarella A, Cavinato M, Rosini E, Shehi H, Ballabio F, Camilloni C, Fasano V, Silvani A, Passarella D, Pollegioni L, Nardini M. Nicotinic Acid Derivatives As Novel Noncompetitive α-Amylase and α-Glucosidase Inhibitors for Type 2 Diabetes Treatment. ACS Med Chem Lett 2024; 15:1474-1481. [PMID: 39291031 PMCID: PMC11403757 DOI: 10.1021/acsmedchemlett.4c00190] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 09/19/2024] Open
Abstract
A library of novel nicotinic acid derivatives, focusing on the modification of position 6 of the pyridine ring with (thio)ether functionalities, was mostly produced through an innovative green synthetic approach (Cyrene-based) and evaluated for their α-amylase and α-glucosidase inhibitory activity. Compounds 8 and 44 demonstrated micromolar inhibition against α-amylase (IC50 of 20.5 and 58.1 μM, respectively), with 44 exhibiting a remarkable ∼72% enzyme inactivation level, surpassing the efficacy of the control compound, acarbose. Conversely, 35 and 39 exhibited comparable inhibition values to acarbose against α-glucosidase (IC50 of 32.9 and 26.4 μM, respectively) and a significant enhancement in enzyme inhibition at saturation (∼80-90%). Mechanistic studies revealed that the most promising compounds operated through a noncompetitive inhibition mechanism for both α-amylase and α-glucosidase, offering advantages for function regulation over competitive inhibitors. These inhibitors may open a new perspective for the development of improved hypoglycemic agents for type 2 diabetes treatment.
Collapse
Affiliation(s)
- Andrea Citarella
- Department of Chemistry, University of Milan, Via Golgi 19 20133 Milano, Italy
| | - Miriam Cavinato
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy
| | - Elena Rosini
- Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Haidi Shehi
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy
| | - Federico Ballabio
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy
| | - Carlo Camilloni
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy
| | - Valerio Fasano
- Department of Chemistry, University of Milan, Via Golgi 19 20133 Milano, Italy
| | - Alessandra Silvani
- Department of Chemistry, University of Milan, Via Golgi 19 20133 Milano, Italy
| | - Daniele Passarella
- Department of Chemistry, University of Milan, Via Golgi 19 20133 Milano, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100 Varese, Italy
| | - Marco Nardini
- Department of Biosciences, University of Milan, Via Celoria 26, 20133 Milano, Italy
| |
Collapse
|