1
|
Yavuz O, Dincel GC, Yildirim S, El-Ashram S, Al-Olayan E. Impact of apoptosis and oxidative stress on pancreatic beta cell pathophysiology in streptozotocin-induced Type 1 diabetes mellitus. Tissue Cell 2024; 91:102552. [PMID: 39255742 DOI: 10.1016/j.tice.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/21/2024] [Accepted: 08/31/2024] [Indexed: 09/12/2024]
Abstract
AIMS Hyperglycemia plays a crucial role in the islet cells, especially pancreatic beta cell death in type 1 diabetes mellitus (T1DM). However, a few research have concentrated on the pathophysiology of apoptosis and oxidative stress in T1DM. The aim of this study was to determine the expression of Caspase 3, Caspase 9, 8-OHdG, Glutathione Reductase, endothelial and inducible nitric oxide synthase in the pancreatic tissue of streptozotocin (STZ)-induced T1DM patients and to compare the cellular mechanisms underlying this metabolic disorder. METHODS For this purpose, a total of 20 Wistar albino rats were divided into two groups: Control (C) and Diabetes Mellitus (DM). In the DM group, T1DM was induced by STZ. Rats in the C group were injected intravenously with buffer solution. At the end of the day 20, rats were necropsied and immunohistochemical procedures were applied. RESULTS The immunohistochemical examination revealed, strong positive immunoreactions were observed in the islet cells of the DM groups, particularly when all antibody stains were considered. On the other hand, the C groups showed minimal changes. The difference between the C and DM groups in terms of all antibodies was statistically significant (p<0.01). CONCLUSIONS In the present study, it was concluded that apoptosis, oxidative stress and NOS expressions were involved in islet cell destruction in pancreatic tissue in STZ-induced T1DM.
Collapse
Affiliation(s)
- Orhan Yavuz
- Department of Pathology, Faculty of Veterinary Medicine, Dokuz Eylul University, 35890, Kiraz, Izmir, Türkiye
| | - Gungor Cagdas Dincel
- College of Life Science and Engineering, Foshan University, 18 Jiangwan Street, Foshan, 528231, Guangdong Province, China.
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Ataturk University, Erzurum, 25240, Türkiye
| | - Saeed El-Ashram
- College of Life Science and Engineering, Foshan University, 18 Jiangwan Street, Foshan, 528231, Guangdong Province, China; Zoology Department, Faculty of Science, Kafrelsheikh University, Kafr El-Sheikh, 33516, Egypt.
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
dos Santos TW, Pereira QC, Fortunato IM, Oliveira FDS, Alvarez MC, Ribeiro ML. Body Composition and Senescence: Impact of Polyphenols on Aging-Associated Events. Nutrients 2024; 16:3621. [PMID: 39519454 PMCID: PMC11547493 DOI: 10.3390/nu16213621] [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: 09/09/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Aging is a dynamic and progressive process characterized by the gradual accumulation of cellular damage. The continuous functional decline in the intrinsic capacity of living organisms to precisely regulate homeostasis leads to an increased susceptibility and vulnerability to diseases. Among the factors contributing to these changes, body composition-comprised of fat mass and lean mass deposits-plays a crucial role in the trajectory of a disability. Particularly, visceral and intermuscular fat deposits increase with aging and are associated with adverse health outcomes, having been linked to the pathogenesis of sarcopenia. Adipose tissue is involved in the secretion of bioactive factors that can ultimately mediate inter-organ pathology, including skeletal muscle pathology, through the induction of a pro-inflammatory profile such as a SASP, cellular senescence, and immunosenescence, among other events. Extensive research has shown that natural compounds have the ability to modulate the mechanisms associated with cellular senescence, in addition to exhibiting anti-inflammatory, antioxidant, and immunomodulatory potential, making them interesting strategies for promoting healthy aging. In this review, we will discuss how factors such as cellular senescence and the presence of a pro-inflammatory phenotype can negatively impact body composition and lead to the development of age-related diseases, as well as how the use of polyphenols can be a functional measure for restoring balance, maintaining tissue quality and composition, and promoting health.
Collapse
Affiliation(s)
- Tanila Wood dos Santos
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Quélita Cristina Pereira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Isabela Monique Fortunato
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Fabrício de Sousa Oliveira
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| | - Marisa Claudia Alvarez
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
- Hematology and Transfusion Medicine Center, University of Campinas/Hemocentro, UNICAMP, Rua Carlos Chagas 480, Campinas 13083-878, SP, Brazil
| | - Marcelo Lima Ribeiro
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University, Av. Sao Francisco de Assis, 218, Braganca Paulista 12916-900, SP, Brazil; (T.W.d.S.); (Q.C.P.); (I.M.F.); (F.d.S.O.); (M.C.A.)
| |
Collapse
|
3
|
Motlagh RA, Pipella J, Thompson PJ. Exploring senescence as a modifier of β cell extracellular vesicles in type 1 diabetes. Front Endocrinol (Lausanne) 2024; 15:1422279. [PMID: 39239092 PMCID: PMC11374605 DOI: 10.3389/fendo.2024.1422279] [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: 04/23/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Type 1 Diabetes (T1D) is a chronic metabolic disease resulting from insulin deficiency due to autoimmune loss of pancreatic β cells. In addition to β cell destruction, it is now accepted that β cell stress and dysfunction, such as senescence, plays a crucial role in the development of the disease. Accumulation of senescent β cells occurs during development of T1D in humans and contributes to the progression of T1D in the nonobese diabetic (NOD) mouse model. Senescent β cells are thought to exacerbate the inflammatory response within the islets by production and secretion of senescence-associated secretory phenotype (SASP). Extracellular vesicles (EVs) from β cells have been shown to carry protein and microRNAs (miRNAs), influencing cellular signaling and may contribute to the development of T1D but it remains to be addressed how senescence impacts β cell EV cargo. In this minireview, we discuss emerging evidence that EV cargo proteins and miRNAs associated with senescence could contribute to the development of T1D and could suggest potential biomarkers and therapeutic targets for the regulation of SASP and elimination of senescent β cells in T1D. Future investigation exploring the intricate relationship between β cell senescence, EVs and miRNAs could pave the way for the development of novel diagnostic techniques and therapeutic interventions.
Collapse
Affiliation(s)
- Roozbeh Akbari Motlagh
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Jasmine Pipella
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| | - Peter J Thompson
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Theme, Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
4
|
Kamrul-Hasan ABM, Mondal S, Nagendra L, Yadav A, Aalpona FTZ, Dutta D. Role of Teplizumab, a Humanized Anti-CD3 Monoclonal Antibody, in Managing Newly Diagnosed Type 1 Diabetes: An Updated Systematic Review and Meta-Analysis. Endocr Pract 2024; 30:431-440. [PMID: 38519028 DOI: 10.1016/j.eprac.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/23/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE Teplizumab has emerged as a potential disease-modifying drug in type 1 diabetes (T1D). This meta-analysis sought to summarize the therapeutic effect of teplizumab in newly diagnosed patients with T1D. METHODS Randomized controlled trials involving patients with T1D receiving teplizumab in the intervention arm and placebo (or no active intervention) in the control arm were searched throughout the electronic databases. The primary outcome was the change in area under the curve of C-peptide levels from baseline. RESULTS Seven reports from 6 studies involving 834 subjects met the inclusion criteria. Compared to teplizumab, greater reductions in area under the curve of C-peptide from the baseline values were observed in the control group after 6 months (mean difference [MD] 0.07 nmol/L [0.01, 0.13], P = .02), after 12 months (MD 0.07 nmol/L [0.04, 0.11], P = .0001), after 18 months (MD 0.10 nmol/L [0.06, 0.14], P < .00001), and after 24 months (MD 0.07 nmol/L [0.01, 0.14], P = .03) of interventions. Moreover, fewer patients treated with teplizumab had a decreased C-peptide response after 6 months (odds ratio [OR] 0.21), after 12 months (OR 0.17), after 18 months (OR 0.30), and after 24 months (OR 0.12) of treatment. The preservation of endogenous insulin production was supported by reduced use of exogenous insulin with maintenance of comparable glycemic control for up to 18 months post-treatment. Teplizumab imparted higher risks of grade 3 or higher adverse events, adverse events leading to study medication discontinuation, nausea, rash, and lymphopenia. CONCLUSION The results of the meta-analysis support teplizumab as a promising disease-modifying therapy for newly diagnosed T1D.
Collapse
Affiliation(s)
- A B M Kamrul-Hasan
- Department of Endocrinology, Mymensingh Medical College, Mymensingh, Bangladesh.
| | - Sunetra Mondal
- Department of Endocrinology, NRS Medical College, Kolkata, India
| | - Lakshmi Nagendra
- Department of Endocrinology, JSS Medical College, JSS Academy of Higher Education and Research, Mysore, India
| | - Ashmita Yadav
- Department of Neurosciences, Nobel Medical College and Teaching Hospital, Biratnagar, Nepal
| | | | - Deep Dutta
- Department of Endocrinology, CEDAR Superspeciality Healthcare, New Delhi, India
| |
Collapse
|
5
|
Thompson PJ, Pipella J, Rutter GA, Gaisano HY, Santamaria P. Islet autoimmunity in human type 1 diabetes: initiation and progression from the perspective of the beta cell. Diabetologia 2023; 66:1971-1982. [PMID: 37488322 PMCID: PMC10542715 DOI: 10.1007/s00125-023-05970-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 07/26/2023]
Abstract
Type 1 diabetes results from the poorly understood process of islet autoimmunity, which ultimately leads to the loss of functional pancreatic beta cells. Mounting evidence supports the notion that the activation and evolution of islet autoimmunity in genetically susceptible people is contingent upon early life exposures affecting the islets, especially beta cells. Here, we review some of the recent advances and studies that highlight the roles of these changes as well as antigen presentation and stress response pathways in beta cells in the onset and propagation of the autoimmune process in type 1 diabetes. Future progress in this area holds promise for advancing islet- and beta cell-directed therapies that could be implemented in the early stages of the disease and could be combined with immunotherapies.
Collapse
Affiliation(s)
- Peter J Thompson
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada.
| | - Jasmine Pipella
- Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Guy A Rutter
- CRCHUM and Department of Medicine, Université de Montréal, Montréal, QC, Canada.
- Department of Diabetes, Endocrinology and Medicine, Faculty of Medicine, Imperial College, London, UK.
- LKC School of Medicine, Nanyang Technological College, Singapore, Republic of Singapore.
| | - Herbert Y Gaisano
- Departments of Medicine and Physiology, University of Toronto, Toronto, ON, Canada
| | - Pere Santamaria
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Institut D'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
6
|
Varghese SS, Dhawan S. Senescence: a double-edged sword in beta-cell health and failure? Front Endocrinol (Lausanne) 2023; 14:1196460. [PMID: 37229454 PMCID: PMC10203573 DOI: 10.3389/fendo.2023.1196460] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/19/2023] [Indexed: 05/27/2023] Open
Abstract
Cellular senescence is a complex process marked by permanent cell-cycle arrest in response to a variety of stressors, and acts as a safeguard against the proliferation of damaged cells. Senescence is not only a key process underlying aging and development of many diseases, but has also been shown to play a vital role in embryogenesis as well as tissue regeneration and repair. In context of the pancreatic beta-cells, that are essential for maintaining glucose homeostasis, replicative senescence is responsible for the age-related decline in regenerative capacity. Stress induced premature senescence is also a key early event underlying beta-cell failure in both type 1 and type 2 diabetes. Targeting senescence has therefore emerged as a promising therapeutic avenue for diabetes. However, the molecular mechanisms that mediate the induction of beta-cell senescence in response to various stressors remain unclear. Nor do we know if senescence plays any role during beta-cell growth and development. In this perspective, we discuss the significance of senescence in beta-cell homeostasis and pathology and highlight emerging directions in this area that warrant our attention.
Collapse
Affiliation(s)
| | - Sangeeta Dhawan
- Department of Translational Research and Cellular Therapeutics, Arthur Riggs Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, United States
| |
Collapse
|
7
|
Ahmadi SF, Mansour RN, Hassannia H, Enderami SE, Abediankenari S, Hosseini-Khah Z. Generation of glucose sensitive insulin-secreting cells from human induced pluripotent stem cells on optimized polyethersulfone hybrid nanofibrous scaffold. Artif Organs 2023; 47:502-511. [PMID: 36287200 DOI: 10.1111/aor.14431] [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: 07/01/2022] [Revised: 09/19/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND In the realm of diabetes treatment, various strategies have been tried, including islet transplantation and common drug therapies, but the limitations of these procedures and lack of responsive to the high number of patients have prompted researchers to develop a new method. In recent decades, the use of stem cells and three-dimonsional (3D) scaffold to produce insulin-secreting cells is one of the most promising new approaches. Meanwhile, human-induced pluripotent stem cells (iPSCs) propose due to advantages such as autologousness and high pluripotency in cell therapy. This study aimed to evaluate the differentiation of iPSCs into pancreatic islet insuli-producing cells (IPCs) on Silk/PES (polyethersulfone) nanofibers as a 3D scaffold and compare it with a two-dimonsional (2D) cultured group. METHODS Investigating the functional, morphological, molecular, and cellular characteristics of differentiated iPSCs on control cultures (without differentiation medium), 2D and 3D were measured by various methods such as electron microscopy, Q-PCR, immunofluorescence, western blot, and ELISA. RESULTS This investigation revealed that differentiated cells on the 3D Silk/PES scaffold expressed pancreatic specific-markers such as insulin and pdx1 at higher levels than the control and 2D groups, with a significant difference between the two groups. All results of Q-PCR, immunocytochemistry, and western blot showed that IPCs in the silk/PES 3D group was more efficient than in the 2D group. In the face of these cases, the release of insulin and C-peptide in response to several concentrations of glucose in the 3D group was significantly higher than in the 2D culture. CONCLUSION Finally, our findings displayed that optimized Silk/PES 3D scaffolds can enhance the differentiation of IPCs from iPSCs compared to the 2D culture group.
Collapse
Affiliation(s)
- Seyedeh Fatemeh Ahmadi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Hadi Hassannia
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Ehsan Enderami
- Immunogenetics Research Center, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Saeid Abediankenari
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Hosseini-Khah
- Diabetes Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
8
|
Kang SL, Hwang YN, Kwon JY, Kim SM. Effectiveness and safety of a model predictive control (MPC) algorithm for an artificial pancreas system in outpatients with type 1 diabetes (T1D): systematic review and meta-analysis. Diabetol Metab Syndr 2022; 14:187. [PMID: 36494830 PMCID: PMC9733359 DOI: 10.1186/s13098-022-00962-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this study was to assess the effectiveness and safety of a model predictive control (MPC) algorithm for an artificial pancreas system in outpatients with type 1 diabetes. METHODS We searched PubMed, EMBASE, Cochrane Central, and the Web of Science to December 2021. The eligibility criteria for study selection were randomized controlled trials comparing artificial pancreas systems (MPC, PID, and fuzzy algorithms) with conventional insulin therapy in type 1 diabetes patients. The heterogeneity of the overall results was identified by subgroup analysis of two factors including the intervention duration (overnight and 24 h) and the follow-up periods (< 1 week, 1 week to 1 month, and > 1 month). RESULTS The meta-analysis included a total of 41 studies. Considering the effect on the percentage of time maintained in the target range between the MPC-based artificial pancreas and conventional insulin therapy, the results showed a statistically significantly higher percentage of time maintained in the target range in overnight use (10.03%, 95% CI [7.50, 12.56] p < 0.00001). When the follow-up period was considered, in overnight use, the MPC-based algorithm showed a statistically significantly lower percentage of time maintained in the hypoglycemic range (-1.34%, 95% CI [-1.87, -0.81] p < 0.00001) over a long period of use (> 1 month). CONCLUSIONS Overnight use of the MPC-based artificial pancreas system statistically significantly improved glucose control while increasing time maintained in the target range for outpatients with type 1 diabetes. Results of subgroup analysis revealed that MPC algorithm-based artificial pancreas system was safe while reducing the time maintained in the hypoglycemic range after an overnight intervention with a long follow-up period (more than 1 month).
Collapse
Affiliation(s)
- Su Lim Kang
- Department of Medical Device and Healthcare, Dongguk University-Seoul, 26, Pil-Dong 3-Ga, Seoul, Jung-Gu 04620 Republic of Korea
| | - Yoo Na Hwang
- Department of Medical Device and Healthcare, Dongguk University-Seoul, 26, Pil-Dong 3-Ga, Seoul, Jung-Gu 04620 Republic of Korea
| | - Ji Yean Kwon
- Department of Medical Device and Healthcare, Dongguk University-Seoul, 26, Pil-Dong 3-Ga, Seoul, Jung-Gu 04620 Republic of Korea
| | - Sung Min Kim
- Department of Medical Device and Healthcare, Dongguk University-Seoul, 26, Pil-Dong 3-Ga, Seoul, Jung-Gu 04620 Republic of Korea
- Department of Medical Device Regulatory Science, Dongguk University-Seoul, 26, Pil-dong 3-Ga, Seoul, Jung-Gu 04620 Republic of Korea
| |
Collapse
|
9
|
Kulkarni A, Muralidharan C, May SC, Tersey SA, Mirmira RG. Inside the β Cell: Molecular Stress Response Pathways in Diabetes Pathogenesis. Endocrinology 2022; 164:bqac184. [PMID: 36317483 PMCID: PMC9667558 DOI: 10.1210/endocr/bqac184] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 11/05/2022]
Abstract
The pathogeneses of the 2 major forms of diabetes, type 1 and type 2, differ with respect to their major molecular insults (loss of immune tolerance and onset of tissue insulin resistance, respectively). However, evidence suggests that dysfunction and/or death of insulin-producing β-cells is common to virtually all forms of diabetes. Although the mechanisms underlying β-cell dysfunction remain incompletely characterized, recent years have witnessed major advances in our understanding of the molecular pathways that contribute to the demise of the β-cell. Cellular and environmental factors contribute to β-cell dysfunction/loss through the activation of molecular pathways that exacerbate endoplasmic reticulum stress, the integrated stress response, oxidative stress, and impaired autophagy. Whereas many of these stress responsive pathways are interconnected, their individual contributions to glucose homeostasis and β-cell health have been elucidated through the development and interrogation of animal models. In these studies, genetic models and pharmacological compounds have enabled the identification of genes and proteins specifically involved in β-cell dysfunction during diabetes pathogenesis. Here, we review the critical stress response pathways that are activated in β cells in the context of the animal models.
Collapse
Affiliation(s)
- Abhishek Kulkarni
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Charanya Muralidharan
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah C May
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| |
Collapse
|
10
|
Brawerman G, Pipella J, Thompson PJ. DNA damage to β cells in culture recapitulates features of senescent β cells that accumulate in type 1 diabetes. Mol Metab 2022; 62:101524. [PMID: 35660116 PMCID: PMC9213768 DOI: 10.1016/j.molmet.2022.101524] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/24/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE Type 1 Diabetes (T1D) is characterized by progressive loss of insulin-producing pancreatic β cells as a result of autoimmune destruction. In addition to β cell death, recent work has shown that subpopulations of β cells acquire dysfunction during T1D. We previously reported that β cells undergoing a DNA damage response (DDR) and senescence accumulate during the pathogenesis of T1D. However, the question of how senescence develops in β cells has not been investigated. METHODS Here, we tested the hypothesis that unrepaired DNA damage in the context of genetic susceptibility triggers β cell senescence using culture models including the mouse NIT1 β cell line derived from the T1D-susceptible nonobese diabetic (NOD) strain, human donor islets and EndoC β cells. DNA damage was chemically induced using etoposide or bleomycin and cells or islets were analyzed by a combination of molecular assays for senescence phenotypes including Western blotting, qRT-PCR, Luminex assays, flow cytometry and histochemical staining. RNA-seq was carried out to profile global transcriptomic changes in human islets undergoing DDR and senescence. Insulin ELISAs were used to quantify glucose-stimulated insulin secretion from chemically-induced senescent human islets, EndoC β cells and mouse β cell lines in culture. RESULTS Sub-lethal DNA damage in NIT1 cells led to several classical hallmarks of senescence including sustained DDR activation, growth arrest, enlarged flattened morphology and a senescence-associated secretory phenotype (SASP) resembling what occurs in primary β cells during T1D in NOD mice. These phenotypes differed between NIT1 cells and the MIN6 β cell line derived from a non-T1D susceptible mouse strain. RNA-seq analysis of DNA damage-induced senescence in human islets from two different donors revealed a p53 transcriptional program and upregulation of prosurvival and SASP genes, with inter-donor variability in this response. Inter-donor variability in human islets was also apparent in the extent of persistent DDR activation and SASP at the protein level. Notably, chemically induced DNA damage also led to DDR activation and senescent phenotypes in EndoC-βH5 human β cells, confirming that this response can occur directly in a human β cell line. Finally, DNA damage led to different effects on glucose-stimulated insulin secretion in mouse β cell lines as compared with human islets and EndoC β cells. CONCLUSIONS Taken together, these findings suggest that some of the phenotypes of senescent β cells that accumulate during the development of T1D in the NOD mouse and humans can be modeled by chemically induced DNA damage to mouse β cell lines, human islets and EndoC β cells in culture. The differences between β cells from different mouse strains and different human islet donors and EndoC β cells highlights species differences and the role for genetic background in modifying the β cell response to DNA damage and its effects on insulin secretion. These culture models will be useful tools to understand some of the mechanisms of β cell senescence in T1D.
Collapse
Affiliation(s)
- Gabriel Brawerman
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, 715 McDermot ave, Winnipeg, MB R3E 3P4, Canada
| | - Jasmine Pipella
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, 715 McDermot ave, Winnipeg, MB R3E 3P4, Canada
| | - Peter J Thompson
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada; Children's Hospital Research Institute of Manitoba, 715 McDermot ave, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
11
|
Nanotechnology in Immunotherapy for Type 1 Diabetes: Promising Innovations and Future Advances. Pharmaceutics 2022; 14:pharmaceutics14030644. [PMID: 35336018 PMCID: PMC8955746 DOI: 10.3390/pharmaceutics14030644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/05/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Diabetes is a chronic condition which affects the glucose metabolism in the body. In lieu of any clinical “cure,” the condition is managed through the administration of pharmacological aids, insulin supplements, diet restrictions, exercise, and the like. The conventional clinical prescriptions are limited by their life-long dependency and diminished potency, which in turn hinder the patient’s recovery. This necessitated an alteration in approach and has instigated several investigations into other strategies. As Type 1 diabetes (T1D) is known to be an autoimmune disorder, targeting the immune system in activation and/or suppression has shown promise in reducing beta cell loss and improving insulin levels in response to hyperglycemia. Another strategy currently being explored is the use of nanoparticles in the delivery of immunomodulators, insulin, or engineered vaccines to endogenous immune cells. Nanoparticle-assisted targeting of immune cells holds substantial potential for enhanced patient care within T1D clinical settings. Herein, we summarize the knowledge of etiology, clinical scenarios, and the current state of nanoparticle-based immunotherapeutic approaches for Type 1 diabetes. We also discuss the feasibility of translating this approach to clinical practice.
Collapse
|
12
|
El-Shafey M, El-Agawy MSED, Eldosoky M, Ebrahim HA, Elsherbini DMA, El-Sherbiny M, Asseri SM, Elsherbiny NM. Role of Dapagliflozin and Liraglutide on Diabetes-Induced Cardiomyopathy in Rats: Implication of Oxidative Stress, Inflammation, and Apoptosis. Front Endocrinol (Lausanne) 2022; 13:862394. [PMID: 35370937 PMCID: PMC8972060 DOI: 10.3389/fendo.2022.862394] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The current study aims to assess the protective effects of dapagliflozin (Dapa; a sodium-glucose cotransporter-2 inhibitor) and/or liraglutide (Lira; a glucagon-like peptide 1 agonist) in an experimental model of diabetic cardiomyopathy (DCM). A single dose of streptozotocin (STZ) was administrated to male Sprague-Dawley rats by intraperitoneal injection at a dose of 50 mg/kg to induce diabetes mellitus (DM). Dapa (1 mg/kg, orally), Lira (0.4 mg/kg, s.c.), and Dapa-Lira combination were administrated for 8 weeks once-daily. Blood samples were evaluated for glucose level and biochemical markers of cardiac functions. Cardiac tissue was dissected and assessed for redox homeostasis (malondialdehyde (MDA), glutathione (GSH), and catalase (CAT)), pro-inflammatory mediators (NF-κB and tumor necrosis factor-α (TNF-α)), and apoptotic effectors (caspase-3). Moreover, the effect of treatments on the cardiac cellular structure was studied. Dapa and/or Lira administration resulted in significant improvement of biochemical indices of cardiac function. Additionally, all treatment groups demonstrated restoration of oxidant/antioxidant balance. Moreover, inflammation and apoptosis key elements were markedly downregulated in cardiac tissue. Also, histological studies demonstrated attenuation of diabetes-induced cardiac tissue injury. Interestingly, Dapa-Lira combination treatment produced a more favorable protective effect as compared to a single treatment. These data demonstrated that Dapa, Lira, and their combination therapy could be useful in protection against DM-accompanied cardiac tissue injury, shedding the light on their possible utilization as adjuvant therapy for the management of DM patients.
Collapse
Affiliation(s)
- Mohamed El-Shafey
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Physiological Sciences Department, Fakeeh College for Medical Sciences, Jeddah, Saudi Arabia
| | | | - Mohamed Eldosoky
- Department of Neuroscience Technology-College of Applied Sciences, Jubail Imam Abdulraman bin Faisal University, Dammam, Saudi Arabia
| | - Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Dalia Mahmoud Abdelmonem Elsherbini
- Department of Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- *Correspondence: Mohamed El-Sherbiny, ; Nehal M. Elsherbiny,
| | - Saad Mohamed Asseri
- Department of Clinical Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
| | - Nehal M. Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia
- *Correspondence: Mohamed El-Sherbiny, ; Nehal M. Elsherbiny,
| |
Collapse
|
13
|
Cefalu WT, Andersen DK, Arreaza-Rubín G, Pin CL, Sato S, Verchere CB, Woo M, Rosenblum ND. Heterogeneity of Diabetes: β-Cells, Phenotypes, and Precision Medicine: Proceedings of an International Symposium of the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases. Diabetes Care 2022; 45:3-22. [PMID: 34782355 PMCID: PMC8753760 DOI: 10.2337/dci21-0051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 02/03/2023]
Abstract
One hundred years have passed since the discovery of insulin-an achievement that transformed diabetes from a fatal illness into a manageable chronic condition. The decades since that momentous achievement have brought ever more rapid innovation and advancement in diabetes research and clinical care. To celebrate the important work of the past century and help to chart a course for its continuation into the next, the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases recently held a joint international symposium, bringing together a cohort of researchers with diverse interests and backgrounds from both countries and beyond to discuss their collective quest to better understand the heterogeneity of diabetes and thus gain insights to inform new directions in diabetes treatment and prevention. This article summarizes the proceedings of that symposium, which spanned cutting-edge research into various aspects of islet biology, the heterogeneity of diabetic phenotypes, and the current state of and future prospects for precision medicine in diabetes.
Collapse
Affiliation(s)
- William T. Cefalu
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Dana K. Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Guillermo Arreaza-Rubín
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Christopher L. Pin
- Departments of Physiology and Pharmacology, Paediatrics, and Oncology, University of Western Ontario, and Genetics and Development Division, Children’s Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Sheryl Sato
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - C. Bruce Verchere
- Departments of Surgery and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children’s Hospital, Vancouver, British Columbia, Canada
- UBC Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Minna Woo
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, University Health Network and Sinai Health System, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Norman D. Rosenblum
- Canadian Institutes of Health Research Institute of Nutrition, Metabolism and Diabetes, Toronto, Ontario, Canada
- Division of Nephrology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada
- Program in Stem Cell and Developmental Biology, Research Institute, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
14
|
Cefalu WT, Andersen DK, Arreaza-Rubín G, Pin CL, Sato S, Verchere CB, Woo M, Rosenblum ND. Heterogeneity of Diabetes: β-Cells, Phenotypes, and Precision Medicine: Proceedings of an International Symposium of the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases. Can J Diabetes 2021; 45:697-713. [PMID: 34794897 DOI: 10.1016/j.jcjd.2021.09.126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 10/19/2022]
Abstract
One hundred years have passed since the discovery of insulin-an achievement that transformed diabetes from a fatal illness into a manageable chronic condition. The decades since that momentous achievement have brought ever more rapid innovation and advancement in diabetes research and clinical care. To celebrate the important work of the past century and help to chart a course for its continuation into the next, the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases recently held a joint international symposium, bringing together a cohort of researchers with diverse interests and backgrounds from both countries and beyond to discuss their collective quest to better understand the heterogeneity of diabetes and thus gain insights to inform new directions in diabetes treatment and prevention. This article summarizes the proceedings of that symposium, which spanned cutting-edge research into various aspects of islet biology, the heterogeneity of diabetic phenotypes, and the current state of and future prospects for precision medicine in diabetes.
Collapse
Affiliation(s)
- William T Cefalu
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States.
| | - Dana K Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Guillermo Arreaza-Rubín
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - Christopher L Pin
- Departments of Physiology and Pharmacology, Paediatrics, and Oncology, University of Western Ontario, and Genetics and Development Division, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Sheryl Sato
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States
| | - C Bruce Verchere
- Departments of Surgery and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital, Vancouver, British Columbia, Canada; UBC Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Minna Woo
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada; Division of Endocrinology and Metabolism, University Health Network and Sinai Health System, Toronto, Ontario, Canada; Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | - Norman D Rosenblum
- Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes, Toronto, Ontario, Canada; Division of Nephrology, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada; Program in Stem Cell and Developmental Biology, Research Institute, The Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
15
|
Cefalu WT, Andersen DK, Arreaza-Rubín G, Pin CL, Sato S, Verchere CB, Woo M, Rosenblum ND. Heterogeneity of Diabetes: β-Cells, Phenotypes, and Precision Medicine: Proceedings of an International Symposium of the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases. Diabetes 2021; 71:db210777. [PMID: 34782351 PMCID: PMC8763877 DOI: 10.2337/db21-0777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 11/13/2022]
Abstract
One hundred years have passed since the discovery of insulin-an achievement that transformed diabetes from a fatal illness into a manageable chronic condition. The decades since that momentous achievement have brought ever more rapid innovation and advancement in diabetes research and clinical care. To celebrate the important work of the past century and help to chart a course for its continuation into the next, the Canadian Institutes of Health Research's Institute of Nutrition, Metabolism and Diabetes and the U.S. National Institutes of Health's National Institute of Diabetes and Digestive and Kidney Diseases recently held a joint international symposium, bringing together a cohort of researchers with diverse interests and backgrounds from both countries and beyond to discuss their collective quest to better understand the heterogeneity of diabetes and thus gain insights to inform new directions in diabetes treatment and prevention. This article summarizes the proceedings of that symposium, which spanned cutting-edge research into various aspects of islet biology, the heterogeneity of diabetic phenotypes, and the current state of and future prospects for precision medicine in diabetes.
Collapse
Affiliation(s)
- William T Cefalu
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Dana K Andersen
- Division of Digestive Diseases and Nutrition, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Guillermo Arreaza-Rubín
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - Christopher L Pin
- Departments of Physiology and Pharmacology, Paediatrics, and Oncology, University of Western Ontario, and Genetics and Development Division, Children's Health Research Institute, Lawson Health Research Institute, London, Ontario, Canada
| | - Sheryl Sato
- Division of Diabetes, Endocrinology and Metabolic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD
| | - C Bruce Verchere
- Departments of Surgery and Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital, Vancouver, British Columbia, Canada
- UBC Centre for Molecular Medicine and Therapeutics, Vancouver, British Columbia, Canada
| | - Minna Woo
- Departments of Medicine and Immunology, University of Toronto, Toronto, Ontario, Canada
- Division of Endocrinology and Metabolism, University Health Network and Sinai Health System, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
16
|
Khalil RG, Abdel-Moneim A, Yousef AI, Abdel-Rahman H, Zanaty MI, El-Sayed A. Association of interleukin-2, interleukin-21 and interleukin-23 with hyperlipidemia in pediatric type 1 diabetes. Mol Biol Rep 2021; 48:5421-5433. [PMID: 34328597 DOI: 10.1007/s11033-021-06545-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND In type 1 diabetes mellitus (T1DM), cytokines have a central role in orchestrating multicellular relations between β-cells and immune cells. This study aims to investigate the role of interleukin (IL)-21, IL-23, and IL-2, and their association with dyslipidemia in T1DM children. METHODS The sample population consisted of 30 healthy controls and 70 children with T1DM, the latter of which were split into two groups according to the duration of their T1DM diagnosis: recent (≤ 1 year; n = 21) and older (> 1 year; n = 49) diagnoses. RESULTS Fasting blood sugar and glycated hemoglobin levels in all diabetic children were significantly (P < 0.001) higher, whereas levels of plasma C-peptide were markedly (P < 0.001) lower in children with T1DM compared to healthy controls. In older T1DM diagnosis children, the levels of creatinine were noticeably (P < 0.05) increased relative to healthy controls. In all diabetic children, levels of total triglyceride, cholesterol, and low-density lipoprotein were increased significantly (P < 0.001) than those of healthy controls. Furthermore, the IL-21 and IL-23 mRNA expressions of all children with T1DM were elevated significantly (P < 0.001) relative to healthy controls, whereas IL-2 levels revealed a significant (P < 0.001) decrease in all diabetic children. CONCLUSION There was a synergistic interplay between IL-21 and IL-23 with an antagonistic action of IL-2 in T1DM patients, and all three interleukins were associated with dyslipidemia in diabetic children. Importantly, therapies targeting IL-21 and IL-23 are promising targets for preventive strategies against the development of T1DM and its complications.
Collapse
Affiliation(s)
- Rehab G Khalil
- Immunology Division, Faculty of Science, Beni-Suef University, Beni Suef, Egypt
| | - Adel Abdel-Moneim
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Salah Salim St., Beni Suef, 62511, Egypt.
| | - Ahmed I Yousef
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Salah Salim St., Beni Suef, 62511, Egypt
| | - Hanan Abdel-Rahman
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Science, Beni-Suef University, Beni Suef, Egypt
| | - Mohamed I Zanaty
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Science, Beni-Suef University, Beni Suef, Egypt
| | - Amr El-Sayed
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Science, Beni-Suef University, Beni Suef, Egypt
| |
Collapse
|
17
|
Esakov E, Nandedkar-Kulkarni N, Al-Dieri AG, Hafner H, Gregg B, McInerney MF. Islet Dysfunction in a Novel Transgenic Model of T Cell Insulitis. Biomolecules 2021; 11:biom11040552. [PMID: 33918805 PMCID: PMC8070091 DOI: 10.3390/biom11040552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/26/2021] [Accepted: 04/06/2021] [Indexed: 11/25/2022] Open
Abstract
The newly established CD3FLAG-mIR transgenic mouse model on a C57Bl/6 background has a FLAG tag on the mouse Insulin Receptor (mIR), specifically on T cells, as the FLAG-tagged mIR gene was engineered behind CD3 promoter and enhancer. The IR is a chemotactic molecule for insulin and the Flag-tagged mIR T cells in the BL/6-CD3FLAGmIR transgenic mice can migrate into the pancreas, as shown by immunofluorescent staining. While the transgenic mice do not become diabetic, there are phenotypic and metabolic changes in the islets. The transgenic islets become enlarged and disorganized by 15 weeks and those phenotypes continue out to 35 weeks of age. We examined the islets by RT-PCR for cell markers, ER stress markers, beta cell proliferation markers, and cytokines, as well as measuring serum insulin and insulin content in the pancreas at 15, 25, and 35 weeks of age. In transgenic mice, insulin in serum was increased at 15 weeks of age and glucose intolerance developed by 25 weeks of age. Passage of transgenic spleen cells into C57Bl/6 RAG−/− mice resulted in enlarged and disorganized islets with T infiltration by 4 to 5 weeks post-transfer, replicating the transgenic mouse studies. Therefore, migration of non-antigen-specific T cells into islets has ramifications for islet organization and function.
Collapse
Affiliation(s)
- Emily Esakov
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Neha Nandedkar-Kulkarni
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Ali G. Al-Dieri
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
| | - Hannah Hafner
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (H.H.); (B.G.)
| | - Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (H.H.); (B.G.)
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marcia F. McInerney
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA; (E.E.); (N.N.-K.); (A.G.A.-D.)
- Center for Diabetes and Endocrine Research, Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
- Correspondence: ; Tel.: +1-419-517-3638
| |
Collapse
|