1
|
Funabashi H, Inoue H, Shigematsu R, Imae I, Amemiya Y, Ishida T, Ikeda T, Hirota R, Kuroda A. Electrochemical manipulation of the insulin secretion from pancreatic beta cells directly cultured on a PEDOT:PSS electrode. Biosens Bioelectron 2025; 281:117453. [PMID: 40215889 DOI: 10.1016/j.bios.2025.117453] [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: 12/06/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/04/2025]
Abstract
The development of cell-based devices using mammalian cells is becoming increasingly feasible. To remotely control such sophisticated devices, an interface between digital computer/internet networks and cellular/organ networks is essential. This study explores the electrochemical manipulation of insulin secretion-a regulatory hormone for the control of blood sugar levels-using pancreatic β cells as a model. iGL cells, expressing insulin fused with Gaussia Luciferase (INS-GLase), were directly cultured on a custom-made cell culture device coated with a transparent poly(3,4-ethylenedioxythiophene):poly(styrene sulfonate) electrode. Luminescence imaging was employed to evaluate insulin secretion in response to applied potentials. Results showed that insulin secretion could be induced by regulating membrane potential through an applied potential. The addition of nicardipine, an L-type voltage-dependent Ca2+ channel inhibitor, suppressed insulin secretion, suggesting the involvement of Ca2+ channels in this electrochemical system. Additionally, changes in membrane potential were directly visualized with the membrane potential-sensitive dye FluoVolt™, which confirmed both the forced depolarization and the forced restoration of the membrane potential to its non-excited state upon potential application to the electrode. The reported electrochemical technique, in which cells are directly cultured on an electrode, offers significant promise for designing advanced bio-hybrid systems that integrate cellular functions with digital networks.
Collapse
Affiliation(s)
- Hisakage Funabashi
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan.
| | - Hayate Inoue
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan
| | - Reiji Shigematsu
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan
| | - Ichiro Imae
- Department of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8527, Japan
| | - Yoshiteru Amemiya
- Research Institute for Semiconductor Engineering, Hiroshima University, 1-4-2 Kagamiyama, Higashihiroshima Hiroshima, 739-8527, Japan
| | - Takenori Ishida
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan
| | - Takeshi Ikeda
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan; Research Institute for Semiconductor Engineering, Hiroshima University, 1-4-2 Kagamiyama, Higashihiroshima Hiroshima, 739-8527, Japan
| | - Ryuichi Hirota
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan
| | - Akio Kuroda
- Division of Biological and Life Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-1 Kagamiyama, Higashihiroshima, Hiroshima, 739-8530, Japan
| |
Collapse
|
2
|
Schukarucha Gomes A, Ellis CE, Spigelman AF, Dos Santos T, Maghera J, Suzuki K, MacDonald PE. Molecular correlates of glycine receptor activity in human β cells. Mol Metab 2025; 96:102156. [PMID: 40258441 PMCID: PMC12059332 DOI: 10.1016/j.molmet.2025.102156] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025] Open
Abstract
OBJECTIVES Glycine acts in an autocrine positive feedback loop in human β cells through its ionotropic receptors (GlyRs). In type 2 diabetes (T2D), islet GlyR activity is impaired by unknown mechanisms. We sought to investigate if the GlyR dysfunction in T2D is replicated by hyperglycemia per se, and to further characterize its action in β cells and islets. METHODS GlyR-mediated currents were measured using whole-cell patch-clamp in human β cells from donors with or without T2D, or after high glucose (15 mM) culture. We also correlated glycine-induced current amplitude with transcript expression levels through patch-seq. The expression of the GlyR α1, α3, and β subunit mRNA splice variants was compared between islets from donors with and without T2D, and after high glucose culture. Insulin secretion from human islets was measured in the presence or absence of the GlyR antagonist strychnine. RESULTS Although gene expression of GlyRs was decreased in T2D islets, and β cell GlyR-mediated currents were smaller, we found no evidence for a shift in GlyR subunit splicing. Glycine-induced currents are also reduced after 48 h culture of islets from donors without diabetes in high glucose, where we also find the reduction of the α1 subunit expression, but an increase in the α3 subunit. We discovered that glycine-evoked currents are highly heterogeneous amongst β cells, inversely correlate with donor HbA1c, and are significantly correlated to the expression of 92 different transcripts and gene regulatory networks (GRNs) that include CREB3(+), RREB1(+) and ZNF697(+). Finally, glucose-stimulated insulin secretion is decreased in the presence of the GlyR antagonist strychnine. CONCLUSIONS We demonstrate that glucose can modulate GlyR expression, and that the current decrease in T2D is likely due to the receptor gene expression downregulation, and not a change in transcript splicing. Moreover, we define a previously unknown set of genes and regulons that are correlated to GlyR-mediated currents and could be involved in GlyR downregulation in T2D. Among those we validate the negative impact of EIF4EBP1 expression on GlyR activity.
Collapse
Affiliation(s)
- Amanda Schukarucha Gomes
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Cara E Ellis
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Aliya F Spigelman
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Theodore Dos Santos
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Jasmine Maghera
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Kunimasa Suzuki
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada
| | - Patrick E MacDonald
- Alberta Diabetes Institute, Department of Pharmacology, University of Alberta, Edmonton, Alberta, T6G 2R3, Canada.
| |
Collapse
|
3
|
Millan-Pacheco C, Serratos IN, Félix-Martínez GJ, Blancas-Flores G, Osorno A, Godínez R. Cholesterol Concentration in Cell Membranes and its Impact on Receptor-Ligand Interaction: A Computational Study of ATP-Sensitive Potassium Channels and ATP Binding. J Membr Biol 2025; 258:225-236. [PMID: 40137942 DOI: 10.1007/s00232-025-00345-4] [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: 11/28/2024] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
This work describes a computer study that looks at how different amounts of cholesterol (0%, 25%, and 50%) in cell membranes change the relationship between ATP and the KATP channel. This could explain why pancreatic beta-cells secrete insulin differently. We use computer simulations of molecular dynamics, calculations of binding free energy, and an integrated oscillator model to look at the electrical activity of beta-cells. There is a need for this kind of multiscale approach right now because cholesterol plays a part in metabolic syndrome and early type 2 diabetes. Our results showed that the increase in cholesterol concentration in the cell membrane affects the electrostatic interactions between ATP and the KATP channel, especially with charged residues in the binding site. Cholesterol can influence the properties of a membrane, including its local charge distribution near the channel. This affects the electrostatic environment around the ATP-binding site, increasing the affinity of ATP for the channel as our results indicated from 0 to 25 and 50% cholesterol (- 141 to - 113 kJ/mol, respectively). Simulating this change in the affinity to ATP of the KATP channels in a model of the electrical activity of the pancreatic beta-cell indicates that even a minimal increase could produce hyperinsulism. The study answers an important research question about how the structure of the membrane affects the function of KATP and, in turn, insulin releases a common feature of metabolic syndrome and early stages of type 2 diabetes.
Collapse
Affiliation(s)
- Cesar Millan-Pacheco
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Morelos. Av. Universidad No. 1001, Colonia Chamilpa, 62209, Morelos, México
| | - Iris N Serratos
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México.
| | - Gerardo J Félix-Martínez
- Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Gerardo Blancas-Flores
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Av. San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Alejandra Osorno
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México
| | - Rafael Godínez
- Departamento de Ingeniería Eléctrica, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina. Iztapalapa, C. P. 09340, Ciudad de Mexico, México.
| |
Collapse
|
4
|
Maharwal N, Shrivastava R, Majumder SK. Insight into Optogenetics for Diabetes Management. ACS Synth Biol 2025; 14:1324-1335. [PMID: 40279455 DOI: 10.1021/acssynbio.4c00549] [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] [Indexed: 04/27/2025]
Abstract
Optogenetics is an interdisciplinary field wherein optical and genetic engineering methods are employed together to impart photounresponsive cells (usually of higher animals) the ability to respond to light through expression of light-sensitive proteins sourced generally from algae or bacteria. It enables precise spatiotemporal control of various cellular activities through light stimulation. Recently, emerging as a synthetic biology-based approach for diabetes management, optogenetics can provide user-control of hormonal secretion by photoactivation of a suitably modified cell. For around a decade, studies have been performed on the applicability of various light-sensitive proteins and their incorporation into pancreatic and nonpancreatic cells for photoinduced insulin secretion. Further, in vivo studies demonstrated amelioration of diabetes in mouse models through photoactivation of the implanted engineered cells. Here, we attempt to highlight the various optogenetic approaches explored in terms of influencing the insulin secretion pathway at different points in light of the natural insulin secretion pathway in pancreatic β cells. We also discuss how transgenic cells of both pancreatic as well as nonpancreatic origin are exploited for photoinduced secretion of insulin. Recent advances on integration of "smart" technologies for remote control of light irradiation and thereby insulin secretion from implanted engineered cells in preclinical models are also described. Additionally, the need for further comprehensive studies on irradiation parameters, red-shifted opsins, and host-cell interaction is stressed to realize the full potential of optogenetics as a clinically applicable modality providing user-controlled "on demand" hormonal secretion for better management of diabetes.
Collapse
Affiliation(s)
- Nidhi Maharwal
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| | - Rashmi Shrivastava
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| | - Shovan Kumar Majumder
- Laser Biomedical Applications Division, Laser R&D Block-A1, Raja Ramanna Centre for Advanced Technology, Indore 452013, India
- Homi Bhabha National Institute, 2nd floor, BARC Training School Complex, Anushakti Nagar, Mumbai 400 094, India
| |
Collapse
|
5
|
Davison A, Reimann F, Gribble FM. Molecular mechanisms of stimulus detection and secretion in enteroendocrine cells. Curr Opin Neurobiol 2025; 92:103045. [PMID: 40378579 DOI: 10.1016/j.conb.2025.103045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 05/19/2025]
Abstract
Enteroendocrine cells (EECs) secrete over 20 different gut hormones in response to changes to the gut environment. They detect a range of nutritional stimuli through activation of a host of nutrient-sensing G-protein-coupled receptors and electrogenic nutrient cotransport. These activate intracellular signalling pathways which converge on membrane depolarisation and action potential generation, which elicit secretion. Emerging evidence has demonstrated that EECs also respond to non-nutritional stimuli, including mechanosensation, pH changes, and metabolites produced by the gut microbiome. EECs are polyhormonal cells, in which hormone expression is plastic and dependent on location in the gut. Hormones and small-molecule neurotransmitters secreted by EECs can activate extrinsic vagal afferents, modulating central processes such as appetite and food preference. While neuronal afferents are sometimes found in close proximity to EECs, the extent to which EEC/neuronal connections recapitulate traditional synaptic connections remains undefined.
Collapse
Affiliation(s)
- Adam Davison
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Frank Reimann
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| | - Fiona M Gribble
- Institute of Metabolic Science Metabolic Research Laboratories, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
6
|
Krell S, Hamburg A, Gover O, Molakandov K, Leibowitz G, Sharabi K, Walker MD, Helman A. Beta cells intrinsically sense and limit their secretory activity via mTORC1-RhoA signaling. Cell Rep 2025; 44:115647. [PMID: 40347471 DOI: 10.1016/j.celrep.2025.115647] [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: 10/10/2024] [Revised: 02/24/2025] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Precise regulation of insulin secretion by pancreatic β cells is essential to prevent excessive insulin release. Here, we show that the nutrient sensor mechanistic Target of Rapamycin Complex 1 (mTORC1) is rapidly activated by glucose in β cells via the insulin secretion machinery, positioning mTORC1 as a sensor of β cell activity. Acute pharmacological inhibition of mTORC1 during glucose stimulation enhances insulin release, suggesting that mTORC1 acts as an intrinsic feedback regulator that restrains insulin secretion. Phosphoproteomic profiling reveals that mTORC1 modulates the phosphorylation of proteins involved in actin remodeling and vesicle trafficking, with a prominent role in the RhoA-GTPase pathway. Mechanistically, mTORC1 promotes RhoA activation and F-actin polymerization, limiting vesicle movement and dampening the second phase of insulin secretion. These findings identify a glucose-mTORC1-RhoA signaling axis that forms an autonomous feedback loop to constrain insulin exocytosis, providing insight into how β cells prevent excessive insulin release and maintain metabolic balance.
Collapse
Affiliation(s)
- Saar Krell
- Department of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel.
| | - Amit Hamburg
- Department of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Ofer Gover
- Department of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | | | - Gil Leibowitz
- Diabetes Unit, Department of Endocrinology and Metabolism, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kfir Sharabi
- Department of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel
| | - Michael D Walker
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Aharon Helman
- Department of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, Hebrew University of Jerusalem, Rehovot, Israel.
| |
Collapse
|
7
|
Akter M, Moghimianavval H, Luker GD, Liu AP. Light-Triggered Protease-Mediated Release of Actin-Bound Cargo from Synthetic Cells. Adv Biol (Weinh) 2025; 9:e2400539. [PMID: 39825686 PMCID: PMC12078867 DOI: 10.1002/adbi.202400539] [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: 09/13/2024] [Revised: 12/19/2024] [Indexed: 01/20/2025]
Abstract
Synthetic cells offer a versatile platform for addressing biomedical and environmental challenges, due to their modular design and capability to mimic cellular processes such as biosensing, intercellular communication, and metabolism. Constructing synthetic cells capable of stimuli-responsive secretion is vital for applications in targeted drug delivery and biosensor development. Previous attempts at engineering secretion for synthetic cells have been confined to non-specific cargo release via membrane pores, limiting the spatiotemporal precision and specificity necessary for selective secretion. Here, a protein-based platform termed TEV Protease-mediated Releasable Actin-binding Protein (TRAP) is designed and constructed for selective, rapid, and triggerable secretion in synthetic cells. TRAP is designed to bind tightly to reconstituted actin networks and is proteolytically released from bound actin, followed by secretion via cell-penetrating peptide membrane translocation. TRAP's efficacy in facilitating light-activated secretion of both fluorescent and luminescent proteins is demonstrated. By equipping synthetic cells with a controlled secretion mechanism, TRAP paves the way for the development of stimuli-responsive biomaterials, versatile synthetic cell-based biosensing systems, and therapeutic applications through the integration of synthetic cells with living cells for targeted delivery of protein therapeutics.
Collapse
Affiliation(s)
- Mousumi Akter
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMI48109USA
| | | | - Gary D. Luker
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Department of RadiologyUniversity of MichiganAnn ArborMI48109USA
| | - Allen P. Liu
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMI48109USA
- Cellular and Molecular Biology ProgramUniversity of MichiganAnn ArborMI48109USA
- Department of BiophysicsUniversity of MichiganAnn ArborMI48109USA
| |
Collapse
|
8
|
Gojani EG, Wang B, Li DP, Kovalchuk O, Kovalchuk I. The Impact of Major and Minor Phytocannabinoids on the Maintenance and Function of INS-1 β-Cells Under High-Glucose and High-Lipid Conditions. Molecules 2025; 30:1991. [PMID: 40363798 PMCID: PMC12073157 DOI: 10.3390/molecules30091991] [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: 01/18/2025] [Revised: 04/22/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Type 2 diabetes mellites (T2DM) is the most common form of diabetes and affects a significant portion of the population. Obesity-related increases in free fatty acids and glucose in the diet contribute to β-cell dysfunction and loss, ultimately leading to the onset of T2DM. The endocannabinoid system, which is present throughout the body, plays a vital role in regulating various physiological processes, including those in the pancreas. This system has been implicated in metabolic disorders like obesity and diabetes, as it helps to regulate appetite, food intake, and fat production. Phytocannabinoids from Cannabis sativa have the potential to influence the endocannabinoid system, offering a promising therapeutic approach for diabetes and its complications. Using high-glucose-high-lipid (HGHL)-induced INS-1 β-cells, we investigated the protective effects of two major (THC and CBD) and three minor (THCV, CBC, and CBG) phytocannabinoids on high glucose-high lipid (HGHL)-induced apoptosis, cell cycle disruption, and impaired function of beta-cells. Our results showed that all five phytocannabinoids reduced HGHL-induced apoptosis, likely by decreasing TXNIP protein levels. Additionally, THC and all three minor phytocannabinoids provided protective effects against functional impairments caused by HGHL exposure.
Collapse
Affiliation(s)
| | | | | | - Olga Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.-P.L.)
| | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.-P.L.)
| |
Collapse
|
9
|
Pant S, Tam SW, Long SB. The pentameric chloride channel BEST1 is activated by extracellular GABA. Proc Natl Acad Sci U S A 2025; 122:e2424474122. [PMID: 40249777 PMCID: PMC12037058 DOI: 10.1073/pnas.2424474122] [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: 11/22/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025] Open
Abstract
Bestrophin-1 (BEST1) is a chloride channel expressed in the eye and other tissues of the body. A link between BEST1 and the principal inhibitory neurotransmitter γ-aminobutyric acid (GABA) has been proposed. The most appreciated receptors for extracellular GABA are the GABAB G-protein-coupled receptors and the pentameric GABAA chloride channels, both of which have fundamental roles in the central nervous system. Here, we demonstrate that BEST1 is directly activated by GABA. Through functional studies and atomic-resolution structures of human and chicken BEST1, we identify a GABA binding site on the channel's extracellular side and determine the mechanism by which GABA binding stabilizes opening of the channel's central gate. This same gate, "the neck," is activated by intracellular [Ca2+], indicating that BEST1 is controlled by ligands from both sides of the membrane. The studies demonstrate that BEST1, which shares no structural homology with GABAA receptors, is a GABA-activated chloride channel. The physiological implications of this finding remain to be studied.
Collapse
Affiliation(s)
- Swati Pant
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Biochemistry and Structural Biology, Cell and Developmental Biology, and Molecular Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephanie W. Tam
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
- Graduate Program in Physiology, Biophysics, and Systems Biology, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY10065
| | - Stephen B. Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY10065
| |
Collapse
|
10
|
Bal T. Scaffold-free endocrine tissue engineering: role of islet organization and implications in type 1 diabetes. BMC Endocr Disord 2025; 25:107. [PMID: 40259265 PMCID: PMC12010671 DOI: 10.1186/s12902-025-01919-y] [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: 08/09/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic hyperglycemia disorder emerging from beta-cell (insulin secreting cells of the pancreas) targeted autoimmunity. As the blood glucose levels significantly increase and the insulin secretion is gradually lost, the entire body suffers from the complications. Although various advances in the insulin analogs, blood glucose monitoring and insulin application practices have been achieved in the last few decades, a cure for the disease is not obtained. Alternatively, pancreas/islet transplantation is an attractive therapeutic approach based on the patient prognosis, yet this treatment is also limited mainly by donor shortage, life-long use of immunosuppressive drugs and risk of disease transmission. In research and clinics, such drawbacks are addressed by the endocrine tissue engineering of the pancreas. One arm of this engineering is scaffold-free models which often utilize highly developed cell-cell junctions, soluble factors and 3D arrangement of islets with the cellular heterogeneity to prepare the transplant formulations. In this review, taking T1D as a model autoimmune disease, techniques to produce so-called pseudoislets and their applications are studied in detail with the aim of understanding the role of mimicry and pointing out the promising efforts which can be translated from benchside to bedside to achieve exogenous insulin-free patient treatment. Likewise, these developments in the pseudoislet formation are tools for the research to elucidate underlying mechanisms in pancreas (patho)biology, as platforms to screen drugs and to introduce immunoisolation barrier-based hybrid strategies.
Collapse
Affiliation(s)
- Tugba Bal
- Department of Bioengineering, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, 34662, Turkey.
| |
Collapse
|
11
|
Engström M, Westholm E, Wendt A, Eliasson L. The role of islet CFTR in the development of cystic fibrosis-related diabetes: A semi-systematic review. J Cyst Fibros 2025:S1569-1993(25)00772-6. [PMID: 40254519 DOI: 10.1016/j.jcf.2025.04.006] [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: 10/01/2024] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND Cystic fibrosis related diabetes (CFRD) is the most common comorbidity of cystic fibrosis (CF) still, its pathogenesis is poorly understood. Recent studies have suggested that although pancreatic insufficiency is an important explanation for CFRD development, inherent pancreatic islet cell dysfunction may play a role. This study aimed to systematically compile current data regarding the impact of pancreatic islet cell dysfunction on the development of CFRD. METHODS A systematic search was conducted in PubMed and Embase. The resulting articles were screened for relevant experimental design and outcomes. Articles underwent data extraction and quality assessment before compilation and analysis of the results. RESULTS A total of 268 articles were initially screened and 19 studies conducted between 2006-2022 were finally included in this review. Half of the studies in human tissue and most of the studies in animal tissue could detect CFTR in the islets. Similarly, half of the publications in human islets and most studies in animal islets detect decreased insulin secretion with inhibition/mutation of CFTR. CONCLUSIONS The literature on the role of islet CFTR is contradictory. However, a pattern emerges where CFTR loss-of-function mutations have the potential to negatively affect islet cell function in a way that, together with previously described exocrine damage occurring in CF, could play a part in the development of CFRD.
Collapse
Affiliation(s)
- Matilda Engström
- Islet Cell Exocytosis, Lund University Diabetes Centre (LUDC), Department of Clinical Sciences-Malmö, Lund University, Malmö, Sweden; Clinical Research Centre (CRC), Skåne University Hospital, Malmö, Sweden
| | - Efraim Westholm
- Islet Cell Exocytosis, Lund University Diabetes Centre (LUDC), Department of Clinical Sciences-Malmö, Lund University, Malmö, Sweden; Clinical Research Centre (CRC), Skåne University Hospital, Malmö, Sweden
| | - Anna Wendt
- Islet Cell Exocytosis, Lund University Diabetes Centre (LUDC), Department of Clinical Sciences-Malmö, Lund University, Malmö, Sweden; Clinical Research Centre (CRC), Skåne University Hospital, Malmö, Sweden
| | - Lena Eliasson
- Islet Cell Exocytosis, Lund University Diabetes Centre (LUDC), Department of Clinical Sciences-Malmö, Lund University, Malmö, Sweden; Clinical Research Centre (CRC), Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
12
|
Murao N, Morikawa R, Seino Y, Shimomura K, Maejima Y, Ohno T, Yokoi N, Yamada Y, Suzuki A. Pyruvate kinase modulates the link between β-cell fructose metabolism and insulin secretion. FASEB J 2025; 39:e70500. [PMID: 40151947 PMCID: PMC11950909 DOI: 10.1096/fj.202401912rr] [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: 08/16/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The intricate link between glucose metabolism, ATP production, and glucose-stimulated insulin secretion (GIIS) in pancreatic β-cells has been well established. However, the effects of other digestible monosaccharides on this mechanism remain unclear. This study examined the interaction between intracellular fructose metabolism and GIIS using MIN6-K8 β-cell lines and mouse pancreatic islets. Fructose at millimolar concentrations potentiated insulin secretion in the presence of stimulatory levels (8.8 mM) of glucose. This potentiation was dependent on sweet taste receptor-activated phospholipase Cβ2 (PLCβ2) signaling. Concurrently, metabolic tracing using 13C-labeled fructose and glucose in conjunction with biochemical analyses demonstrated that fructose blunted the glucose-induced increase in the ATP/ADP ratio. Mechanistically, fructose is substantially converted to fructose 1-phosphate (F1P) at the expense of ATP. F1P directly inhibited PKM2 (pyruvate kinase M2), thereby reducing the later glycolytic flux used for ATP production. Remarkably, F1P-mediated PKM2 inhibition was counteracted by TEPP-46, a small-molecule PKM2 activator. TEPP-46 restored glycolytic flux and the ATP/ADP ratio, leading to the enhancement of fructose-potentiated GIIS in MIN6-K8 cells, normal mouse islets, and fructose-unresponsive diabetic mouse islets. These findings reveal an antagonistic interplay between glucose and fructose metabolism in β-cells, highlighting PKM2 as a crucial regulator and broadening our understanding of the relationship between β-cell fuel metabolism and insulin secretion.
Collapse
Affiliation(s)
- Naoya Murao
- Department of Endocrinology, Diabetes and MetabolismFujita Health University, School of MedicineToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Risa Morikawa
- Department of Endocrinology, Diabetes and MetabolismFujita Health University, School of MedicineToyoakeJapan
| | - Yusuke Seino
- Department of Endocrinology, Diabetes and MetabolismFujita Health University, School of MedicineToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Tamio Ohno
- Graduate School of Medicine Center for Research of Laboratory Animals and Medical Research Engineering Division for Research of Laboratory AnimalsGraduate School of Medicine, Nagoya UniversityNagoyaJapan
| | - Norihide Yokoi
- Laboratory of Animal Breeding and Genetics, Graduate School of AgricultureKyoto UniversityKyotoJapan
| | - Yuichiro Yamada
- Department of Endocrinology, Diabetes and MetabolismFujita Health University, School of MedicineToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Atsushi Suzuki
- Department of Endocrinology, Diabetes and MetabolismFujita Health University, School of MedicineToyoakeJapan
| |
Collapse
|
13
|
Velumani K, Rajan PS, Shaik MR, Hussain SA, Shaik B, Guru A, Issac PK. Protective Effect of Artemisinin Against Luperox Induced Oxidative Stress and Insulin Resistance via Pi3k/Akt Pathway in Zebrafish Larvae. Cell Biochem Biophys 2025:10.1007/s12013-025-01747-w. [PMID: 40220071 DOI: 10.1007/s12013-025-01747-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
Oxidative stress plays a critical role in the development of insulin resistance (IR), a key factor in metabolic disorders such as diabetes. Plant active ingredients play a crucial role in protecting organisms from environmental stressors and have shown promising therapeutic potential against various metabolic disorders. Artemisinin (ART), a sesquiterpenoid with a lactone ring obtained from the herb Artemisia annua, exhibits promising therapeutic properties. This study investigates the potential of ART on Luperox (LUP)-induced oxidative stress and the resulting IR in zebrafish larvae, specifically investigating the involvement of the PI3K/AKT signaling pathway. Zebrafish larvae were chosen due to their high sensitivity to oxidative stress, well-characterized glucose metabolism, and genetic similarity to human metabolic pathways. They were exposed to LUP to induce oxidative stress, followed by treatment with ART. The effects were evaluated through biochemical assays, fluorescence staining and gene expression analysis. ART effectively restored key antioxidant enzymes (SOD, CAT, GSH) and mitigated oxidative stress evidenced by reduction in intercellular ROS and lipid peroxidation, as confirmed through DCFDA and DPPP staining assays. Additionally, ART improved glucose uptake and lowered blood glucose levels. Gene expression analysis further indicated increased levels of PI3K/Akt signalling components and antioxidant-related genes (NRF2, HO-1, GPx, and GSR). Our results indicate that artemisinin significantly alleviates oxidative stress by reducing ROS levels and enhancing antioxidant enzyme activity. Furthermore, artemisinin mitigates IR by restoring glucose metabolism and upregulating PI3K/AKT pathway components. These findings highlight the translational potential of plant active ingredients, particularly artemisinin, for the development of therapies targeting IR and oxidative stress-related metabolic disorders.
Collapse
Affiliation(s)
- Kadhirmathiyan Velumani
- Department of Medical Biotechnology, Institute of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - P Sundar Rajan
- Department of Chemical Engineering, Saveetha Engineering College, Chennai, Tamil Nadu, India
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Shaik Althaf Hussain
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Baji Shaik
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Republic of Korea
| | - Ajay Guru
- Department of Cariology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - Praveen Kumar Issac
- Department of Medical Biotechnology, Institute of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India.
| |
Collapse
|
14
|
Pu W, Pan Y, Yang K, Gao J, Tian F, Song J, Huang Y, Li Y. Therapeutic effects and mechanisms of Xinmaitong formula for type 2 diabetes mellitus via GLP-1R signaling. Front Pharmacol 2025; 16:1575450. [PMID: 40271065 PMCID: PMC12014693 DOI: 10.3389/fphar.2025.1575450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Traditional Chinese Medicine (TCM) theory posits that type 2 diabetes mellitus (T2DM) characterized by Qi and Yin deficiency, is associated with elevated blood lipid levels. The Xinmaitong formula (XMT) is a folk remedy believed to lower blood lipid levels. However, the functional components and molecular mechanisms through which XMT exerts its anti-diabetic effects remain to be elucidated. This study aimed to investigate the therapeutic effects and potential mechanisms of XMT in the treatment of T2DM, focusing on the glucagon-like peptide-1 receptor (GLP-1R) signaling pathway. Methods A TCM formula that promotes GLP-1R expression was screened using a GLP-1R promoter-dependent luciferase reporter gene vector (PGL3-GLP-1R-luc). The T2DM mouse model was established using a high-fat diet and streptozotocin (STZ). Blood glucose levels were measured using a glucometer and oral glucose tolerance test (OGTT). Serum biochemical parameters and insulin levels were also assessed. Organ pathology in mice was evaluated using hematoxylin and eosin (H&E) staining. Immunofluorescence (IF) was employed to observe changes in insulin and GLP-1R expression in the pancreas of mice. The effects of medicated serum on Min6 cell growth were examined using a methyl thiazolyl tetrazolium (MTT) assay. A Min6 cell injury model was established to detect cAMP and Ca2+ concentrations. Ultra high-performance liquid chromatography-mass spectrometry (UHPLC-MS) was used to identify blood-absorbed components of XMT. Results Luciferase reporter constructs driven by GLP-1R promoter response elements analysis identified that TCM formula XMT promoted GLP-1R expression. In vivo experiments demonstrated that XMT significantly reduced fasting blood glucose levels in T2DM mice and improved OGTT results. It also exhibited protective effects on islet tissues, notably increasing GLP-1R expression and insulin secretion in the pancreas. Biochemical markers indicated no significant adverse effects on liver or kidney function following XMT administration. After treatment with palmitic acid (PA), GLP-1R expression in Min6 cells was significantly decreased. However, treatment with XMT upregulated GLP-1R expression. Additionally, cyclic adenosine monophosphate (cAMP) and Ca2+ exhibited substantial improvements, and the key pancreatic growth protein PDX1 was activated. Conclusion XMT exerts hypoglycemic effects by upregulating GLP-1R gene expression, enhancing GLP-1R protein synthesis, and subsequently promoting cAMP release. This process activates Ca2+ influx in pancreatic β-cells, triggering insulin exocytosis from islet cells.
Collapse
Affiliation(s)
- Weidong Pu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| | - Yang Pan
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| | - Kang Yang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| | - Jian Gao
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| | - Fen Tian
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| | - Jingrui Song
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yubing Huang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Yanmei Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guizhou Medical University, Guiyang, China
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang, China
- School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guiyang, Guizhou, China
| |
Collapse
|
15
|
Delgadillo-Silva LF, Salazar S, Lopez Noriega L, Provencher-Girard A, Larouche S, Prat A, Rutter GA. Exploration of individual beta cell function over time in vivo: effects of hyperglycemia and glucagon-like peptide-1 receptor (GLP1R) agonism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.31.646461. [PMID: 40236128 PMCID: PMC11996457 DOI: 10.1101/2025.03.31.646461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The coordinated function of beta cells within the pancreatic islet is required for the normal regulation of insulin secretion and is partly controlled by specialized "leader" and highly connected "hub" beta-cell subpopulations. Whether cells within these subpopulations are functionally stable in vivo remains unclear. Here, we establish an approach to monitor Ca 2+ dynamics within individual beta cells over time, after engraftment into the anterior eye chamber, where continuous blood perfusion and near normal innervation pertain. Under normoglycemic conditions, islet network dynamics, and the behavior of individual leaders and hubs, remain stable for at least seven days. Hyperglycemia, resulting from high-fat diet feeding or the loss of a host Gck allele, caused engrafted islets to display incomplete and abortive Ca 2+ waves and overall connectivity was diminished. Whereas hub cell numbers were lowered profoundly in both disease models, leaders largely persisted. Treatment with the GLP1R agonist Exendin-4 led to a recovery of islet-wide Ca 2+ dynamics and the re-emergence of hub cells within minutes, with the effects of the incretin mimetic being more marked than those observed after analogous treatments in vitro . Similar observations were made using 3-dimensional imaging across the whole islet. Our findings thus suggest that incretins may act both directly and indirectly on beta cells in vivo. The approach described may provide broad applicability to the exploration of individual cell function over time in the living animal.
Collapse
|
16
|
Łaszczych D, Czernicka A, Łaszczych K. Targeting GABA signaling in type 1 diabetes and its complications- an update on the state of the art. Pharmacol Rep 2025; 77:409-424. [PMID: 39833509 DOI: 10.1007/s43440-025-00697-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease that leads to the progressive destruction of insulin-producing β cells, resulting in lifelong insulin dependence and a range of severe complications. Beyond conventional glycemic control, innovative therapeutic strategies are needed to address the underlying disease mechanisms. Recent research has highlighted gamma-aminobutyric acid (GABA) as a promising therapeutic target for T1D due to its dual role in modulating both β cell survival and immune response within pancreatic islets. GABA signaling supports β cell regeneration, inhibits α cell hyperactivity, and promotes α-to-β cell transdifferentiation, contributing to improved islet function. Moreover, GABA's influence extends to mitigating T1D complications, including nephropathy, neuropathy, and retinopathy, as well as regulating central nervous system pathways involved in glucose metabolism. This review consolidates the latest advances in GABA-related T1D therapies, covering animal preclinical and human clinical studies and examining the therapeutic potential of GABA receptor modulation, combination therapies, and dietary interventions. Emphasis is placed on the translational potential of GABA-based approaches to enhance β cell viability and counteract autoimmune processes in T1D. Our findings underscore the therapeutic promise of GABA signaling modulation as a novel approach for T1D treatment and encourage further investigation into this pathway's role in comprehensive diabetes management.
Collapse
Affiliation(s)
- Dariusz Łaszczych
- Faculty of Medicine, Collegium Medicum, Nicolaus Copernicus University in Torun, Jagiellońska 13, 85-067, Bydgoszcz, Poland.
| | | | - Katarzyna Łaszczych
- Faculty of Pharmaceutical Sciences, Medical University of Silesia in Katowice, Jedności 8, Sosnowiec, 41-200, Poland
- Ziko Pharmacy, Plebiscytowa 39, Katowice, Poland
| |
Collapse
|
17
|
Ježek P. Physiological Fatty Acid-Stimulated Insulin Secretion and Redox Signaling Versus Lipotoxicity. Antioxid Redox Signal 2025; 42:566-622. [PMID: 39834189 DOI: 10.1089/ars.2024.0799] [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] [Indexed: 01/22/2025]
Abstract
Significance: Type 2 diabetes as a world-wide epidemic is characterized by the insulin resistance concomitant to a gradual impairment of β-cell mass and function (prominently declining insulin secretion) with dysregulated fatty acids (FAs) and lipids, all involved in multiple pathological development. Recent Advances: Recently, redox signaling was recognized to be essential for insulin secretion stimulated with glucose (GSIS), branched-chain keto-acids, and FAs. FA-stimulated insulin secretion (FASIS) is a normal physiological event upon postprandial incoming chylomicrons. This contrasts with the frequent lipotoxicity observed in rodents. Critical Issues: Overfeeding causes FASIS to overlap with GSIS providing repeating hyperinsulinemia, initiates prediabetic states by lipotoxic effects and low-grade inflammation. In contrast the protective effects of lipid droplets in human β-cells counteract excessive lipids. Insulin by FASIS allows FATP1 recruitment into adipocyte plasma membranes when postprandial chylomicrons come late at already low glycemia. Future Directions: Impaired states of pancreatic β-cells and peripheral organs at prediabetes and type 2 diabetes should be revealed, including the inter-organ crosstalk by extracellular vesicles. Details of FA/lipid molecular physiology are yet to be uncovered, such as complex phenomena of FA uptake into cells, postabsorptive inactivity of G-protein-coupled receptor 40, carnitine carrier substrate specificity, the role of carnitine-O-acetyltransferase in β-cells, and lipid droplet interactions with mitochondria. Antioxid. Redox Signal. 42, 566-622.
Collapse
Affiliation(s)
- Petr Ježek
- Department of Mitochondrial Physiology, No.75, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
18
|
Murao N, Morikawa R, Seino Y, Shimomura K, Maejima Y, Yamada Y, Suzuki A. β-Adrenergic Blockers Increase cAMP and Stimulate Insulin Secretion Through a PKA/RYR2/TRPM5 Pathway in Pancreatic β-Cells In Vitro. Pharmacol Res Perspect 2025; 13:e70092. [PMID: 40222952 PMCID: PMC11994265 DOI: 10.1002/prp2.70092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
β-adrenergic blockers (β-blockers) are extensively used to inhibit β-adrenoceptor activation and subsequent cAMP production in many cell types. In this study, we characterized the effects of β-blockers on mouse pancreatic β-cells. Unexpectedly, high concentrations (100 μM) of β-blockers (propranolol and bisoprolol) paradoxically increased cAMP levels 5-10 fold, enhanced Ca2+ influx, and stimulated a 2-4 fold increase in glucose- and glimepiride-induced insulin secretion in MIN6-K8 clonal β-cells and isolated mouse pancreatic islets. These effects were observed despite minimal expression of β-adrenoceptors in these cells. Mechanistically, the cAMP increase led to ryanodine receptor 2 (RYR2) phosphorylation via protein kinase A (PKA), triggering Ca2+-induced Ca2+ release (CICR). CICR then activates transient receptor potential cation channel subfamily M member 5 (TRPM5), resulting in increased Ca2+ influx via voltage-dependent Ca2+ channels. These effects contradict the conventional understanding of the pharmacology of β-blockers, highlighting the variability in β-blocker actions depending on the experimental context.
Collapse
Affiliation(s)
- Naoya Murao
- Department of Endocrinology, School of Medicine, Diabetes and MetabolismFujita Health UniversityToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Risa Morikawa
- Department of Endocrinology, School of Medicine, Diabetes and MetabolismFujita Health UniversityToyoakeJapan
| | - Yusuke Seino
- Department of Endocrinology, School of Medicine, Diabetes and MetabolismFujita Health UniversityToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Kenju Shimomura
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Yuko Maejima
- Department of Bioregulation and Pharmacological MedicineFukushima Medical University School of MedicineFukushimaJapan
| | - Yuichiro Yamada
- Department of Endocrinology, School of Medicine, Diabetes and MetabolismFujita Health UniversityToyoakeJapan
- Yutaka Seino Distinguished Center for Diabetes ResearchKansai Electric Power Medical Research InstituteKyotoJapan
| | - Atsushi Suzuki
- Department of Endocrinology, School of Medicine, Diabetes and MetabolismFujita Health UniversityToyoakeJapan
| |
Collapse
|
19
|
Raoux M, Chapeau D, Lang J. Slow rather than fast calcium events encode physiological inputs and propagate within islets: Lessons from ultrafast imaging on acute pancreatic tissue slices. Acta Physiol (Oxf) 2025; 241:e70028. [PMID: 40083225 DOI: 10.1111/apha.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Matthieu Raoux
- Institute of Chemistry & Biology of Membranes & Nano-Objects, Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France
| | - Dorian Chapeau
- Institute of Chemistry & Biology of Membranes & Nano-Objects, Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France
| | - Jochen Lang
- Institute of Chemistry & Biology of Membranes & Nano-Objects, Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, Pessac, France
| |
Collapse
|
20
|
Lallouet M, Olçomendy L, Gaitan J, Montiège K, Monchablon M, Pirog A, Chapeau D, Puginier E, Renaud S, Raoux M, Lang J. A microfluidic twin islets-on-chip device for on-line electrophysiological monitoring. LAB ON A CHIP 2025; 25:1831-1841. [PMID: 40042033 DOI: 10.1039/d4lc00967c] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
Pancreatic islets play a major role in glucose homeostasis as well as in diabetes, and islets-on-chip devices have been mainly developed using optical means for on-line monitoring. In contrast, no well-characterized electrophysiological platform for on-line analysis with unrivalled temporal resolution has been reported. Extracellular electrophysiology monitors two crucial parameters, islet β-cell activity and β-to-β-cell coupling, does not require chemical or genetic probes with inherent potential bias, is non-invasive and permits repetitive long-term monitoring. We have now developed and characterized a microfluidic islets-on-chip for combined electrophysiology (on-line) and hormone monitoring (off-line) with two chambers for concomitant monitoring. Fabrication of the device, based on commercial or easily manufacturable components, is within the reach of non-specialized laboratories. The chip permits convenient loading as well as long-term culture with comparable glucose kinetics and low shear stress in both chambers. An optimized flow rate did not alter islet β-cell electrical activity or coupling in response to glucose. Culturing for up to 8 days did not change islet survival as well as glucose-induced electrical or secretory kinetics of islet β-cells. The addition of a physiological amino acid mix, in the presence of elevated glucose, made a considerable change in the functional organisation of islet β-cell activity in terms of frequency and coupling, which explains the ensuing strong increase in insulin secretion. This device thus allows reliable long-term multiparametric on-line monitoring in two islet populations. The ease of fabrication, assembly and handling should permit widespread long-term on-line monitoring of islet activity in native micro-organs (e.g. controls/mutants), pseudo-islets or stem-cell-derived islet-like organoids.
Collapse
Affiliation(s)
- Marie Lallouet
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Loic Olçomendy
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Julien Gaitan
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Killian Montiège
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Marie Monchablon
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Antoine Pirog
- Junia, Electronics-Physics-Acoustics Department, F-59000 Lille, France
| | - Dorian Chapeau
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Emilie Puginier
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Sylvie Renaud
- Univ. Bordeaux, CNRS, Bordeaux INP, Integration from Material to System, IMS, UMR 5218, F-33400 Talence, France
| | - Matthieu Raoux
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| | - Jochen Lang
- Univ. Bordeaux, CNRS, Bordeaux INP, Institute of Chemistry and Biology of Membranes, CBMN, UMR 5248, Pessac, France.
| |
Collapse
|
21
|
Gresch A, Osthues J, Hüwel JD, Briggs JK, Berger T, Koch R, Deickert T, Beecks C, Benninger RK, Düfer M. Resolving Spatiotemporal Electrical Signaling Within the Islet via CMOS Microelectrode Arrays. Diabetes 2025; 74:343-354. [PMID: 39585952 PMCID: PMC11842603 DOI: 10.2337/db23-0870] [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: 10/30/2023] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Glucose-stimulated β-cells exhibit synchronized calcium dynamics across the islet that recruit β-cells to enhance insulin secretion. Compared with calcium dynamics, the formation and cell-to-cell propagation of electrical signals within the islet are poorly characterized. To determine factors that influence the propagation of electrical activity across the islet underlying calcium oscillations and β-cell synchronization, we used high-resolution complementary metal-oxide-semiconductor multielectrode arrays (CMOS-MEA) to measure voltage changes associated with the membrane potential of individual cells within intact C57BL6 mouse islets. We measured fast (milliseconds, spikes) and slow (seconds, waves) voltage dynamics. Single spike activity and wave signal velocity were both glucose-dependent, but only spike activity was influenced by N-methyl-d-aspartate receptor activation or inhibition. A repeated glucose stimulus revealed a highly responsive subset of cells in spike activity. When islets were pretreated for 72 h with glucolipotoxic medium, the wave velocity was significantly reduced. Network analysis confirmed that in response to glucolipotoxicity the synchrony of islet cells was affected due to slower propagating electrical waves and not due to altered spike activity. In summary, this approach provided novel insight regarding the propagation of electrical activity and the disruption of cell-to-cell communication due to excessive stimulation. ARTICLE HIGHLIGHTS The high-resolution complementary metal-oxide-semiconductor multielectrode array is suited to track the spatiotemporal propagation of electrical activity through the islet on a cellular scale. A highly responsive subpopulation of islet cells was identified by action potential-like spike activity and proved to be robust to glucolipotoxicity. Electrical waves revealed synchronized electrical activity and their propagation through the islet was slowed down by glucolipotoxicity. The N-methyl-d-aspartate receptor did not influence islet synchronization since modulation of the receptor only affected electrical spikes. The technique is a useful tool for exploring the pancreatic islet network in health and disease.
Collapse
Affiliation(s)
- Anne Gresch
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster Pharma Campus, Münster, Germany
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jana Osthues
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster Pharma Campus, Münster, Germany
| | - Jan D. Hüwel
- Department of Mathematics and Computer Science, Chair of Data Science, University of Hagen, Hagen, Germany
| | - Jennifer K. Briggs
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Tim Berger
- Department of Mathematics and Computer Science, Chair of Data Science, University of Hagen, Hagen, Germany
| | - Ruben Koch
- Department of Mathematics and Computer Science, Chair of Data Science, University of Hagen, Hagen, Germany
| | - Thomas Deickert
- Department of Mathematics and Computer Science, Chair of Data Science, University of Hagen, Hagen, Germany
| | - Christian Beecks
- Department of Mathematics and Computer Science, Chair of Data Science, University of Hagen, Hagen, Germany
| | - Richard K.P. Benninger
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Martina Düfer
- Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, University of Münster Pharma Campus, Münster, Germany
| |
Collapse
|
22
|
Kosheleva L, Koshelev D, Lagunas-Rangel FA, Levit S, Rabinovitch A, Schiöth HB. Disease-modifying pharmacological treatments of type 1 diabetes: Molecular mechanisms, target checkpoints, and possible combinatorial treatments. Pharmacol Rev 2025; 77:100044. [PMID: 40014914 PMCID: PMC11964952 DOI: 10.1016/j.pharmr.2025.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/01/2025] Open
Abstract
After a century of extensive scientific investigations, there is still no curative or disease-modifying treatment available that can provide long-lasting remission for patients diagnosed with type 1 diabetes (T1D). Although T1D has historically been regarded as a classic autoimmune disorder targeting and destroying pancreatic islet β-cells, significant research has recently demonstrated that β-cells themselves also play a substantial role in the disease's progression, which could explain some of the unfavorable clinical outcomes. We offer a thorough review of scientific and clinical insights pertaining to molecular mechanisms behind pathogenesis and the different therapeutic interventions in T1D covering over 20 possible pharmaceutical intervention treatments. The interventions are categorized as immune therapies, treatments targeting islet endocrine dysfunctions, medications with dual modes of action in immune and islet endocrine cells, and combination treatments with a broader spectrum of activity. We suggest that these collective findings can provide a valuable platform to discover new combinatorial synergies in search of the curative disease-modifying intervention for T1D. SIGNIFICANCE STATEMENT: This research delves into the underlying causes of T1D and identifies critical mechanisms governing β-cell function in both healthy and diseased states. Thus, we identify specific pathways that could be manipulated by existing or new pharmacological interventions. These interventions fall into several categories: (1) immunomodifying therapies individually targeting immune cell processes, (2) interventions targeting β-cells, (3) compounds that act simultaneously on both immune cell and β-cell pathways, and (4) combinations of compounds simultaneously targeting immune and β-cell pathways.
Collapse
Affiliation(s)
- Liudmila Kosheleva
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Daniil Koshelev
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden
| | - Francisco Alejandro Lagunas-Rangel
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Shmuel Levit
- Diabetes and Metabolism Institute, Assuta Medical Centers, Tel Aviv, Israel
| | | | - Helgi B Schiöth
- Department of Surgical Sciences, Functional Pharmacology and Neuroscience, Uppsala University, Uppsala, Sweden; Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia.
| |
Collapse
|
23
|
Meng X, Zhang H, Zhao Z, Li S, Zhang X, Guo R, Liu H, Yuan Y, Li W, Song Q, Liu J. Type 3 diabetes and metabolic reprogramming of brain neurons: causes and therapeutic strategies. Mol Med 2025; 31:61. [PMID: 39966707 PMCID: PMC11834690 DOI: 10.1186/s10020-025-01101-z] [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: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Abnormal glucose metabolism inevitably disrupts normal neuronal function, a phenomenon widely observed in Alzheimer's disease (AD). Investigating the mechanisms of metabolic adaptation during disease progression has become a central focus of research. Considering that impaired glucose metabolism is closely related to decreased insulin signaling and insulin resistance, a new concept "type 3 diabetes mellitus (T3DM)" has been coined. T3DM specifically refers to the brain's neurons becoming unresponsive to insulin, underscoring the strong link between diabetes and AD. Recent studies reveal that during brain insulin resistance, neurons exhibit mitochondrial dysfunction, reduced glucose metabolism, and elevated lactate levels. These findings suggest that impaired insulin signaling caused by T3DM may lead to a compensatory metabolic shift in neurons toward glycolysis. Consequently, this review aims to explore the underlying causes of T3DM and elucidate how insulin resistance drives metabolic reprogramming in neurons during AD progression. Additionally, it highlights therapeutic strategies targeting insulin sensitivity and mitochondrial function as promising avenues for the successful development of AD treatments.
Collapse
Affiliation(s)
- Xiangyuan Meng
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Hui Zhang
- Institute of Agricultural Quality Standard and Testing Technology, Jilin Academy of Agricultural Sciences, Changchun, 130021, China
| | - Zhenhu Zhao
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Siyao Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Xin Zhang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Ruihan Guo
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Huimin Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Yiling Yuan
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Wanrui Li
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Qi Song
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China
| | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, 130021, China.
| |
Collapse
|
24
|
Shao B, Wang Z, Luo P, Du P, Zhang X, Zhang H, Si X, Ma S, Chen W, Huang Y. Identifying insulin-responsive circRNAs in chicken pectoralis. BMC Genomics 2025; 26:148. [PMID: 39955508 PMCID: PMC11830218 DOI: 10.1186/s12864-025-11347-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/10/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) are stable, covalently closed non-coding RNAs formed by reverse splicing of precursor mRNA. They play critical roles in various biological processes, including insulin secretion and metabolism. However, their function in avian skeletal muscle's response to insulin remains poorly understood. This study aimed to comprehensively identify insulin-responsive circRNAs and explore their temporal and breed-specific regulation in poultry. RESULTS Using strand-specific RNA sequencing (ssRNA-Seq) on the pectoralis muscles of both Arbor Acres (AA) broilers and Silky fowls following insulin administration (5 IU/kg.BW, PBS as control). We identified 2,027 muscle circRNAs. Insulin-responsive circRNAs were detected in Silky fowls (29) and broilers (45) at 120 min, and in broilers (20) at 15 min post-injection. These circRNAs are enriched in processes such as exocrine pancreas development, response to exogenous stimuli, and regulation of intracellular signal transduction, likely mediated through a circRNA-miRNA network. Fewer insulin-responsive circRNAs were shared between time points in broilers (1) or between breeds (3) at 120 min. We further characterized a conserved insulin-responsive circRNA (circINSR), formed by exon 2 of the Insulin Receptor (INSR). The circINSR showed a similar spatiotemporal expression pattern to INSR, but exhibited distinct changes post-insulin administration. In broilers, INSR expression was dynamically modulated, while circINSR was downregulated only at 15 min (P < 0.01). Conversely, glucose did not change muscle circINSR but increased INSR at 10 min (P < 0.01). Energy restriction significantly downregulated circINSR (P < 0.01) without affecting INSR levels, and pyruvate had no effect on either circINSR or INSR levels. CONCLUSION This study reveals the dynamic and breed-specific roles of circRNAs, particularly circINSR, in avian skeletal muscle's response to insulin. The distinct regulation of circINSR and INSR under various metabolic conditions suggests a complex regulatory mechanism. These findings provide novel insights into the molecular basis of insulin signaling in avian species and highlight the potential of circRNAs as biomarkers for metabolic regulation.
Collapse
Affiliation(s)
- Binghao Shao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Ziyang Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pengna Luo
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Pengfei Du
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xiangli Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Huaiyong Zhang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Xuemeng Si
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Sen Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China
| | - Wen Chen
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| | - Yanqun Huang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450002, China.
| |
Collapse
|
25
|
Thieu T, Holmes WR. Impact of the Microtubule Cytoskeleton on Insulin Transport in Beta Cells: A 3D Computational Study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637971. [PMID: 39990414 PMCID: PMC11844541 DOI: 10.1101/2025.02.12.637971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Glucose-stimulated insulin secretion (GSIS) in pancreatic β cells is vital to metabolic homeostasis. Recent evidence has highlighted the critical role of the cells' microtubule (MT) cytoskeleton in regulating transport and availability of insulin containing vesicles. How these vesicles move within the cell and how that mobility is influenced by the MT network is however not well understood. The MT network in these cells is dense and randomly oriented. Further insulin vesicles are relatively large compared to the spaces in this dense meshwork. Here we develop a 3D computational model that simulates vesicle motions in the dense MT network of the β cell. The structure of this MT network, along with the dynamics of vesicle motions, are calibrated to microscopy data from β cells to ensure physiological relevance. Our results reveal a number of key observations. 1) The MT network in β cells likely impairs motion of larger vesicles (200 - 300 nm in diameter). 2) This is in part a consequence of their "caging" by the MT network. 3) This results in a substantial reduction in the likelihood of vesicles transiting from the cells interior to the plasma membrane, a pre-cursor to GSIS. 4) Dynamic remodeling of the MT network reduces the strength of these effects. 5) That same remodeling however introduces anomalous (sub-diffusion) motion characteristics. Taken together, these results indicate that the dense MT network of the β cell substantially inhibits mobility and availability (for GSIS) of insulin. It further sheds light on how the complex filament network in cells leads to statistically anomalous motions. Finally, this modeling further provides a test-bed for determining how potential manipulations of the structure and dynamics of this network would tune GSIS. SIGNIFICANCE Insulin release from pancreatic β cells is crucial for blood sugar regulation, and recent research suggests the microtubule network inside these cells plays a key role in how insulin is transported and released. This study developed a 3D computational model to explore how insulin vesicles move through this dense network. Results show that the microtubules can "cage" larger vesicles, making it difficult for them to reach the cell surface for insulin release. Dynamic remodeling of the network can however increase insulin mobility and availability. These findings highlight the impact of the microtubule network on insulin transport and secretion and provide insight into potential ways to tune this process.
Collapse
|
26
|
Li AH, Tsai WS, Tsai WH, Yang SB. Systemic Glucose Homeostasis Requires Pancreatic but Not Neuronal ATP-sensitive Potassium Channels. FUNCTION 2025; 6:zqaf002. [PMID: 39809576 PMCID: PMC11815579 DOI: 10.1093/function/zqaf002] [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: 11/07/2024] [Revised: 12/31/2024] [Accepted: 01/09/2025] [Indexed: 01/16/2025] Open
Abstract
The adenosine triphosphate (ATP)-sensitive potassium (KATP) channels, composed of Kir6.2 and sulfonylurea receptor 1 (SUR1) subunits, are essential for glucose homeostasis. While the role of pancreatic KATP channels in regulating insulin secretion is well-documented, the specific contributions of neuronal KATP channels remain unclear due to challenges in precisely targeting neuronal subpopulations. In this study, we utilized a Kir6.2 conditional knockout mouse model to distinguish the roles of KATP channels in different cell types. Our findings demonstrate that deletion of neuronal KATP channels does not impair glucose homeostasis, as glucose-sensing neurons retained their responsiveness despite the absence of functional KATP channels. In contrast, the deletion of KATP channels in pancreatic β cells led to significant hyperglycemia and glucose intolerance, indicating unstable blood glucose levels under varying physiological conditions. Importantly, we showed that restoring KATP channel function exclusively in pancreatic β cells within a global Kir6.2 knockout background effectively reversed glucose regulation defects. This underscores the critical role of pancreatic KATP channels in maintaining systemic glucose homeostasis. Our results challenge the previous hypothesis that neuronal KATP channels are essential for glucose regulation, suggesting that their primary function may be neuroprotective rather than homeostatic. These findings highlight pancreatic KATP channels as key regulators of glucose balance and potential therapeutic targets for correcting glucose dysregulation.
Collapse
Affiliation(s)
- Athena H Li
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 115, Taiwan
| | - Wen-Sheng Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Wen-Hao Tsai
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
| | - Shi-Bing Yang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
27
|
Oguh AU, Haemmerle MW, Sen S, Rozo AV, Shrestha S, Cartailler JP, Fazelinia H, Ding H, Preza S, Yang J, Yang X, Sussel L, Alvarez-Dominguez JR, Doliba N, Spruce LA, Arrojo E Drigo R, Stoffers DA. E3 ligase substrate adaptor SPOP fine-tunes the UPR of pancreatic β cells. Genes Dev 2025; 39:261-279. [PMID: 39797759 PMCID: PMC11789638 DOI: 10.1101/gad.352010.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/07/2024] [Indexed: 01/13/2025]
Abstract
The Cullin-3 E3 ligase adaptor protein SPOP targets proteins for ubiquitination and proteasomal degradation. We previously established the β-cell transcription factor (TF) and human diabetes gene PDX1 as an SPOP substrate, suggesting a functional role for SPOP in the β cell. Here, we generated a β-cell-specific Spop deletion mouse strain (Spop βKO) and found that Spop is necessary to prevent aberrant basal insulin secretion and for maintaining glucose-stimulated insulin secretion through impacts on glycolysis and glucose-stimulated calcium flux. Integration of proteomic, TF-regulatory gene network, and biochemical analyses identified XBP1 as a functionally important SPOP substrate in pancreatic β cells. Furthermore, loss of SPOP strengthened the IRE1α-XBP1 axis of unfolded protein response (UPR) signaling. ER stress promoted proteasomal degradation of SPOP, supporting a model whereby SPOP fine-tunes XBP1 activation during the UPR. These results position SPOP as a regulator of β-cell function and proper UPR activation.
Collapse
Affiliation(s)
- Alexis U Oguh
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Matthew W Haemmerle
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Sabyasachi Sen
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Andrea V Rozo
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Shristi Shrestha
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Jean-Philippe Cartailler
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Hossein Fazelinia
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Hua Ding
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Sam Preza
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Juxiang Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Xiaodun Yang
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lori Sussel
- Department of Pediatrics and Cell and Developmental Biology, Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Juan R Alvarez-Dominguez
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Nicolai Doliba
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA
| | - Lynn A Spruce
- Proteomics Core Facility, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19146, USA
| | - Rafael Arrojo E Drigo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37232, USA
| | - Doris A Stoffers
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19146, USA;
| |
Collapse
|
28
|
Dolenšek J, Pohorec V, Skelin Klemen M, Gosak M, Stožer A. Ultrafast multicellular calcium imaging of calcium spikes in mouse beta cells in tissue slices. Acta Physiol (Oxf) 2025; 241:e14261. [PMID: 39803792 PMCID: PMC11726428 DOI: 10.1111/apha.14261] [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: 01/16/2024] [Revised: 11/29/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The crucial steps in beta cell stimulus-secretion coupling upon stimulation with glucose are oscillatory changes in metabolism, membrane potential, intracellular calcium concentration, and exocytosis. The changes in membrane potential consist of bursts of spikes, with silent phases between them being dominated by membrane repolarization and absence of spikes. Assessing intra- and intercellular coupling at the multicellular level is possible with ever-increasing detail, but our current ability to simultaneously resolve spikes from many beta cells remains limited to double-impalement electrophysiological recordings. METHODS Since multicellular calcium imaging of spikes would enable a better understanding of coupling between changes in membrane potential and calcium concentration in beta cell collectives, we set out to design an appropriate methodological approach. RESULTS Combining the acute tissue slice method with ultrafast calcium imaging, we were able to resolve and quantify individual spikes within bursts at a temporal resolution of >150 Hz over prolonged periods, as well as describe their glucose-dependent properties. In addition, by simultaneous patch-clamp recordings we were able to show that calcium spikes closely follow membrane potential changes. Both bursts and spikes coordinate across islets in the form of intercellular waves, with bursts typically displaying global and spikes more local patterns. CONCLUSIONS This method and the associated findings provide additional insight into the complex signaling within beta cell networks. Once extended to tissue from diabetic animals and human donors, this approach could help us better understand the mechanistic basis of diabetes and find new molecular targets.
Collapse
Affiliation(s)
- Jurij Dolenšek
- Faculty of MedicineUniversity of MariborMariborSlovenia
- Faculty of Natural Sciences and MathematicsUniversity of MariborMariborSlovenia
| | | | | | - Marko Gosak
- Faculty of MedicineUniversity of MariborMariborSlovenia
- Faculty of Natural Sciences and MathematicsUniversity of MariborMariborSlovenia
- Alma Mater Europaea UniversityMariborSlovenia
| | - Andraž Stožer
- Faculty of MedicineUniversity of MariborMariborSlovenia
| |
Collapse
|
29
|
Chen Cardenas SM, Baker TA, Shimoda LA, Bernal-Mizrachi E, Punjabi NM. L-type calcium channel blockade worsens glucose tolerance and β-cell function in C57BL6/J mice exposed to intermittent hypoxia. Am J Physiol Endocrinol Metab 2025; 328:E161-E172. [PMID: 39763275 DOI: 10.1152/ajpendo.00423.2023] [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: 12/14/2023] [Revised: 11/08/2024] [Accepted: 12/02/2024] [Indexed: 01/24/2025]
Abstract
Intermittent hypoxemia (IH), a pathophysiologic consequence of obstructive sleep apnea (OSA), adversely affects insulin sensitivity, insulin secretion, and glucose tolerance. Nifedipine, an L-type calcium channel blocker frequently used for the treatment of hypertension, can also impair insulin sensitivity and secretion. However, the cumulative and interactive repercussions of IH and nifedipine on glucose homeostasis have not been previously investigated. Adult male C57BL6/J mice were exposed to either nifedipine or vehicle concurrently with IH or intermittent air (IA) over 5 days. IH exposure entailed cycling fractional-inspired oxygen levels between 0.21 and 0.055 at a rate of 60 events/h. Nifedipine (20 mg/kg/day) or vehicle was administered via subcutaneous osmotic pumps resulting in four groups of mice: IA-vehicle (control), IA-nifedipine, IH-vehicle, and IH-nifedipine. Compared with IA (control), IH increased fasting glucose (mean Δ: 33.0 mg/dL; P < 0.001) and insulin (mean Δ: 0.53 ng/mL; P < 0.001) with nifedipine having no independent effect. Furthermore, glucose tolerance was worse with nifedipine alone, and IH further exacerbated the impairment in glucose disposal (P = 0.013 for interaction). Nifedipine also decreased glucose-stimulated insulin secretion and the insulinogenic index, with addition of IH attenuating those measures further. There were no discernible alterations in insulin biosynthesis/processing, insulin content, or islet morphology. These findings underscore the detrimental impact of IH on insulin sensitivity and glucose tolerance while highlighting that nifedipine exacerbates these disturbances through impaired β-cell function. Consequently, cautious use of L-type calcium channel blockers is warranted in patients with OSA, particularly in those at risk for type 2 diabetes.NEW & NOTEWORTHY The results of this study demonstrate the interaction between intermittent hypoxemia (IH) and nifedipine in a murine model. IH raises fasting glucose and insulin levels, with nifedipine exacerbating these disturbances. Glucose tolerance worsens when nifedipine is administered alone, and IH magnifies the impairment in glucose disposal. These findings raise the possibility of potential deleterious effects of L-type calcium channel blockers in patients with obstructive sleep apnea (OSA).
Collapse
Affiliation(s)
- Stanley M Chen Cardenas
- Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Tess A Baker
- Division of Endocrinology, Diabetes, and Metabolism, Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Larissa A Shimoda
- Division of Pulmonary, Critical Care, and Sleep Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes, and Metabolism, Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, United States
| | - Naresh M Punjabi
- Division of Pulmonary, Critical Care, and Sleep Medicine, Miller School of Medicine, University of Miami, Miami, Florida, United States
| |
Collapse
|
30
|
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
|
31
|
Bandesh K, Motakis E, Nargund S, Kursawe R, Selvam V, Bhuiyan RM, Eryilmaz GN, Krishnan SN, Spracklen CN, Ucar D, Stitzel ML. Single-cell decoding of human islet cell type-specific alterations in type 2 diabetes reveals converging genetic- and state-driven β -cell gene expression defects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633590. [PMID: 39896672 PMCID: PMC11785113 DOI: 10.1101/2025.01.17.633590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Pancreatic islets maintain glucose homeostasis through coordinated action of their constituent endocrine and affiliate cell types and are central to type 2 diabetes (T2D) genetics and pathophysiology. Our understanding of robust human islet cell type-specific alterations in T2D remains limited. Here, we report comprehensive single cell transcriptome profiling of 245,878 human islet cells from a 48-donor cohort spanning non-diabetic (ND), pre-diabetic (PD), and T2D states, identifying 14 distinct cell types detected in every donor from each glycemic state. Cohort analysis reveals ~25-30% loss of functional beta cell mass in T2D vs. ND or PD donors resulting from (1) reduced total beta cell numbers/proportions and (2) reciprocal loss of 'high function' and gain of senescent β -cell subpopulations. We identify in T2D β -cells 511 differentially expressed genes (DEGs), including new (66.5%) and validated genes (e.g., FXYD2, SLC2A2, SYT1), and significant neuronal transmission and vitamin A metabolism pathway alterations. Importantly, we demonstrate newly identified DEG roles in human β -cell viability and/or insulin secretion and link 47 DEGs to diabetes-relevant phenotypes in knockout mice, implicating them as potential causal islet dysfunction genes. Additionally, we nominate as candidate T2D causal genes and therapeutic targets 27 DEGs for which T2D genetic risk variants (GWAS SNPs) and pathophysiology (T2D vs. ND) exert concordant expression effects. We provide this freely accessible atlas for data exploration, analysis, and hypothesis testing. Together, this study provides new genomic resources for and insights into T2D pathophysiology and human islet dysfunction.
Collapse
Affiliation(s)
- Khushdeep Bandesh
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Efthymios Motakis
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Siddhi Nargund
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Giray Naim Eryilmaz
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
| | - Sai Nivedita Krishnan
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
| | - Cassandra N. Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts Amherst, Amherst, MA, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| | - Michael L. Stitzel
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032 USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06032 USA
- Institute for Systems Genomics, UConn, Farmington, CT 06032 USA
| |
Collapse
|
32
|
Perez-Serna AA, Guzman-Llorens D, Dos Santos RS, Marroqui L. Bcl-2 and Bcl-xL in Diabetes: Contributions to Endocrine Pancreas Viability and Function. Biomedicines 2025; 13:223. [PMID: 39857806 PMCID: PMC11760435 DOI: 10.3390/biomedicines13010223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Diabetes is a chronic metabolic disorder whose prevalence increases every year, affecting more than 530 million adults worldwide. Type 1 (T1D) and type 2 diabetes (T2D), the most common forms of diabetes, are characterized by the loss of functional pancreatic β-cells, mostly due to apoptosis. B-cell leukemia/lymphoma 2 (Bcl-2) and B-cell lymphoma-extra large (Bcl-xL), two anti-apoptotic proteins belonging to the Bcl-2 family, are crucial for regulating the intrinsic pathway of apoptosis. However, over the years, they have been implicated in many other cellular processes, including intracellular Ca2+ homeostasis and the regulation of mitochondrial metabolism. Thus, understanding the biological processes in which these proteins are involved may be crucial to designing new therapeutic targets. This review summarizes the roles of Bcl-2 and Bcl-xL in apoptosis and metabolic homeostasis. It focuses on how the dysregulation of Bcl-2 and Bcl-xL affects pancreatic β-cell function and survival, and the consequences for diabetes development.
Collapse
Affiliation(s)
- Atenea A. Perez-Serna
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Guzman-Llorens
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
| | - Reinaldo S. Dos Santos
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
- Unidad de Investigación, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana (FISABIO), Hospital General Universitario de Elche, Camí de l’Almazara 11, 03203 Elche, Alicante, Spain
| | - Laura Marroqui
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universidad Miguel Hernández de Elche, 03202 Elche, Alicante, Spain; (A.A.P.-S.); (D.G.-L.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
33
|
Hu M, Liu T, Huang H, Ogi D, Tan Y, Ye K, Jin S. Extracellular matrix proteins refine microenvironments for pancreatic organogenesis from induced pluripotent stem cell differentiation. Theranostics 2025; 15:2229-2249. [PMID: 39990212 PMCID: PMC11840725 DOI: 10.7150/thno.104883] [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: 10/09/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025] Open
Abstract
Rationale: The current understanding on manipulating signaling pathways to generate mature human islet organoids with all major hormone-secreting endocrine cell types (i.e., α, β, δ, and γ cells) from induced pluripotent stem cells (iPSCs) is insufficient. However, donor islet shortage necessitates that we produce functional islets in vitro. In this study, we aimed to find decellularized pancreatic extracellular matrix (dpECM) proteins that leverage signaling pathways and promote functional iPSC islet organogenesis. Methods: We performed proteomic analysis to identify key islet promoting factors from porcine and rat dpECM. With this, we identified collagen type II (COL2) as a potential biomaterial cue that endorses islet development from iPSCs. Using global transcriptome profiling, gene set enrichment analysis, immunofluorescence microscopy, flow cytometry, Western blot, and glucose-stimulated hormonal secretion analysis, we examined COL2's role in regulating iPSC pancreatic lineage specification and signaling pathways, critical to islet organogenesis and morphogenesis. Results: We discovered COL2 acts as a functional biomaterial that augments islet development from iPSCs, similar to collagen type V (COL5) as reported in our earlier study. COL2 substantially stimulates the formation of endocrine progenitors and subsequent islet organoids with significantly elevated expressions of pancreatic signature genes and proteins. Furthermore, it enhances islets' glucose sensitivity for hormonal secretion. A cluster of gene expressions associated with various signaling pathways, including but not limited to oxidative phosphorylation, insulin secretion, cell cycle, the canonical WNT, hypoxia, and interferon-γ response, were considerably affected by COL2 and COL5 cues. Conclusion: We demonstrated dpECM's crucial role in refining stem cell differentiation microenvironments for organoid development and maturation. Our findings on biomaterial-stimulated signaling for stem cell specification, organogenesis, and maturation open up a new way to increase the differentiation efficacy of endocrine tissues that can contribute to the production of biologically functional islets.
Collapse
Affiliation(s)
- Ming Hu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Tianzheng Liu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Hui Huang
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Derek Ogi
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Yinfei Tan
- Genomics Facility, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| | - Sha Jin
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, New York 13902, USA
| |
Collapse
|
34
|
Green C, Zaman V, Blumenstock K, Banik NL, Haque A. Dysregulation of Metabolic Peptides in the Gut-Brain Axis Promotes Hyperinsulinemia, Obesity, and Neurodegeneration. Biomedicines 2025; 13:132. [PMID: 39857716 PMCID: PMC11763097 DOI: 10.3390/biomedicines13010132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Metabolic peptides can influence metabolic processes and contribute to both inflammatory and/or anti-inflammatory responses. Studies have shown that there are thousands of metabolic peptides, made up of short chains of amino acids, that the human body produces. These peptides are crucial for regulating many different processes like metabolism and cell signaling, as they bind to receptors on various cells. This review will cover the role of three specific metabolic peptides and their roles in hyperinsulinemia, diabetes, inflammation, and neurodegeneration, as well as their roles in type 3 diabetes and dementia. The metabolic peptides glucagon-like peptide 1 (GLP-1), gastric inhibitor polypeptide (GIP), and pancreatic peptide (PP) will be discussed, as dysregulation within their processes can lead to the development of various inflammatory and neurodegenerative diseases. Research has been able to closely investigate the connections between these metabolic peptides and their links to the gut-brain axis, highlighting changes made in the gut that can lead to dysfunction in processes in the brain, as well as changes made in the brain that can lead to dysregulation in the gut. The role of metabolic peptides in the development and potentially reversal of diseases such as obesity, hyperinsulinemia, and type 2 diabetes will also be discussed. Furthermore, we review the potential links between these conditions and neuroinflammation and the development of neurodegenerative diseases like dementia, specifically Parkinson's disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Camille Green
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (C.G.); (V.Z.); (N.L.B.)
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (C.G.); (V.Z.); (N.L.B.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA;
| | - Kayce Blumenstock
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA;
- Department of Pharmacology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Narendra L. Banik
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (C.G.); (V.Z.); (N.L.B.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA;
- Department of Pharmacology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Azizul Haque
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC 29425, USA; (C.G.); (V.Z.); (N.L.B.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee Street, Charleston, SC 29401, USA;
- Department of Pharmacology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| |
Collapse
|
35
|
Gojani EG, Wang B, Li D, Kovalchuk O, Kovalchuk I. Single and Combined Impact of Semaglutide, Tirzepatide, and Metformin on β-Cell Maintenance and Function Under High-Glucose-High-Lipid Conditions: A Comparative Study. Int J Mol Sci 2025; 26:421. [PMID: 39796271 PMCID: PMC11720205 DOI: 10.3390/ijms26010421] [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: 12/02/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/30/2025] Open
Abstract
Type 2 diabetes (T2D), the most common form, is marked by insulin resistance and β-cell failure. β-cell dysfunction under high-glucose-high-lipid (HG-HL) conditions is a key contributor to the progression of T2D. This study evaluates the comparative effects of 10 nM semaglutide, 10 nM tirzepatide, and 1 mM metformin, both alone and in combination, on INS-1 β-cell maintenance and function under HG-HL conditions. INS-1 cells were pretreated for 2 h with single doses of metformin (1 mM), semaglutide (10 nM), tirzepatide (10 nM), or combinations of 1 mM metformin with either 10 nM semaglutide or 10 nM tirzepatide, followed by 48 h of HG-HL stimulation. The results indicate that combining 1 mM metformin with either 10 nM semaglutide or 10 nM tirzepatide significantly enhances the effects of 10 nM semaglutide and 10 nM tirzepatide on HG-HL-induced apoptosis and dysregulated cell cycle. Specifically, the combination treatments demonstrated superior restoration of glucose-stimulated insulin secretion (GSIS) functionality compared to 1 mM metformin, 10 nM semaglutide, and 10 nM tirzepatide.
Collapse
Affiliation(s)
| | | | | | | | - Igor Kovalchuk
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (E.G.G.); (B.W.); (D.L.); (O.K.)
| |
Collapse
|
36
|
Belal M, Mucha M, Monteil A, Winyard PG, Pawlak R, Walker JJ, Tabak J, Belle MDC. The background sodium leak channel NALCN is a major controlling factor in pituitary cell excitability. J Physiol 2025; 603:301-317. [PMID: 39620829 PMCID: PMC11737539 DOI: 10.1113/jp284036] [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: 01/05/2023] [Accepted: 10/29/2024] [Indexed: 01/18/2025] Open
Abstract
The pituitary gland produces and secretes a variety of hormones that are essential to life, such as for the regulation of growth and development, metabolism, reproduction, and the stress response. This is achieved through an intricate signalling interplay between the brain and peripheral feedback signals that shape pituitary cell excitability by regulating the ion channel properties of these cells. In addition, endocrine anterior pituitary cells spontaneously fire action potentials to regulate the intracellular calcium ([Ca2+]i) level, an essential signalling conduit for hormonal secretion. To this end, pituitary cells must regulate their resting membrane potential (RMP) close to the firing threshold, but the molecular identity of the ionic mechanisms responsible for this remains largely unknown. Here, we revealed that the sodium leak channel NALCN, known to modulate neuronal excitability elsewhere in the brain, regulates excitability in the mouse anterior endocrine pituitary cells. Using viral transduction combined with powerful electrophysiology methods and calcium imaging, we show that NALCN forms the major Na+ leak conductance in these cells, appropriately tuning cellular RMP for sustaining spontaneous firing activity. Genetic depletion of NALCN channel activity drastically hyperpolarised these cells, suppressing their firing and [Ca2+]i oscillations. Remarkably, despite this profound function of NALCN conductance in controlling pituitary cell excitability, it represents a very small fraction of the total cell conductance. Because NALCN responds to hypothalamic hormones, our results also provide a plausible mechanism through which hormonal feedback signals from the brain and body could powerfully affect pituitary activity to influence hormonal function. KEY POINTS: Pituitary hormones are essential to life as they regulate important physiological processes, such as growth and development, metabolism, reproduction and the stress response. Pituitary hormonal secretion relies on the spontaneous electrical activity of pituitary cells and co-ordinated inputs from the brain and periphery. This appropriately regulates intracellular calcium signals in pituitary cells to trigger hormonal release. Using viral transduction in combination with electrophysiology and calcium imaging, we show that the activity of the background leak channel NALCN is a major controlling factor in eliciting spontaneous electrical activity and intracellular calcium signalling in pituitary cells. Remarkably, our results revealed that a minute change in NALCN activity could have a major influence on pituitary cell excitability. Our study provides a plausible mechanism through which the brain and body could intricately control pituitary activity to influence hormonal function.
Collapse
Affiliation(s)
- Marziyeh Belal
- University of Exeter Medical School, Hatherly LabsExeterDevonUK
- Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Mariusz Mucha
- University of Exeter Medical School, Hatherly LabsExeterDevonUK
| | - Arnaud Monteil
- IGFUniversity of Montpellier, CNRS, INSERMMontpellierFrance
- Department of Physiology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | | | - Robert Pawlak
- University of Exeter Medical School, Hatherly LabsExeterDevonUK
| | - Jamie J. Walker
- College of Engineering, Mathematics and Physical SciencesUniversity of ExeterExeterUK
- EPSRC Centre for Predictive Modelling in HealthcareUniversity of ExeterExeterUK
- Bristol Medical School, Translational Health SciencesUniversity of BristolBristolUK
| | - Joel Tabak
- University of Exeter Medical School, Hatherly LabsExeterDevonUK
| | - Mino D. C. Belle
- University of Exeter Medical School, Hatherly LabsExeterDevonUK
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
| |
Collapse
|
37
|
Feng X, Zhang H, Yang S, Cui D, Wu Y, Qi X, Su Z. From stem cells to pancreatic β-cells: strategies, applications, and potential treatments for diabetes. Mol Cell Biochem 2025; 480:173-190. [PMID: 38642274 DOI: 10.1007/s11010-024-04999-x] [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: 01/16/2024] [Accepted: 03/21/2024] [Indexed: 04/22/2024]
Abstract
Loss and functional failure of pancreatic β-cells results in disruption of glucose homeostasis and progression of diabetes. Although whole pancreas or pancreatic islet transplantation serves as a promising approach for β-cell replenishment and diabetes therapy, the severe scarcity of donor islets makes it unattainable for most diabetic patients. Stem cells, particularly induced pluripotent stem cells (iPSCs), are promising for the treatment of diabetes owing to their self-renewal capacity and ability to differentiate into functional β-cells. In this review, we first introduce the development of functional β-cells and their heterogeneity and then turn to highlight recent advances in the generation of β-cells from stem cells and their potential applications in disease modeling, drug discovery and clinical therapy. Finally, we have discussed the current challenges in developing stem cell-based therapeutic strategies for improving the treatment of diabetes. Although some significant technical hurdles remain, stem cells offer great hope for patients with diabetes and will certainly transform future clinical practice.
Collapse
Affiliation(s)
- Xingrong Feng
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Hongmei Zhang
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Shanshan Yang
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Daxin Cui
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Yanting Wu
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Xiaocun Qi
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China
| | - Zhiguang Su
- Molecular Medicine Research Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, 1 Keyuan 4th Road, Gaopeng Street, Chengdu, 610041, China.
| |
Collapse
|
38
|
Shukla S, Comerci CJ, Süel GM, Jahed Z. Bioelectronic tools for understanding the universal language of electrical signaling across species and kingdoms. Biosens Bioelectron 2025; 267:116843. [PMID: 39426280 DOI: 10.1016/j.bios.2024.116843] [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: 07/24/2023] [Revised: 09/10/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Modern bioelectronic tools are rapidly advancing to detect electric potentials within networks of electrogenic cells, such as cardiomyocytes, neurons, and pancreatic beta cells. However, it is becoming evident that electrical signaling is not limited to the animal kingdom but may be a universal form of cell-cell communication. In this review, we discuss the existing evidence of, and tools used to collect, subcellular, single-cell and network-level electrical signals across kingdoms, including bacteria, plants, fungi, and even viruses. We discuss how cellular networks employ altered electrical "circuitry" and intercellular mechanisms across kingdoms, and we assess the functionality and scalability of cutting-edge nanobioelectronics to collect electrical signatures regardless of cell size, shape, or function. Researchers today aim to design micro- and nano-topographic structures which harness mechanosensitive membrane and cytoskeletal pathways that enable tight electrical coupling to subcellular compartments within high-throughput recording systems. Finally, we identify gaps in current knowledge of inter-species and inter-kingdom electrical signaling and propose critical milestones needed to create a central theory of electrical signaling across kingdoms. Our discussion demonstrates the need for high resolution, high throughput tools which can probe multiple, diverse cell types at once in their native or experimentally-modeled environments. These advancements will not only reveal the underlying biophysical laws governing the universal language of electrical communication, but can enable bidirectional electrical communication and manipulation of biological systems.
Collapse
Affiliation(s)
- Shivani Shukla
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States
| | - Colin J Comerci
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Gürol M Süel
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, United States
| | - Zeinab Jahed
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, United States; Aiiso Yufeng Li Family Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, United States.
| |
Collapse
|
39
|
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: 0] [Impact Index Per Article: 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
|
40
|
Hatamie A, He X, Ewing A, Rorsman P. From Insulin Measurement to Partial Exocytosis Model: Advances in Single Pancreatic Beta Cell Amperometry over Four Decades. ACS MEASUREMENT SCIENCE AU 2024; 4:629-637. [PMID: 39713028 PMCID: PMC11659994 DOI: 10.1021/acsmeasuresciau.4c00058] [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: 08/13/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 12/24/2024]
Abstract
Single cell Amperometry (SCA) is a powerful, sensitive, high temporal resolution electrochemical technique used to quantify secreted molecular messengers from individual cells and vesicles. This technique has been extensively applied to study the process of exocytosis, and it has also been applied, albeit less frequently, to investigate insulin exocytosis from single pancreatic beta cells. Insufficient insulin release can lead to diabetes, a chronic lifestyle disorder that affects millions of people worldwide. This review aims to summarize and highlight electrochemical measurements of insulin via monitoring its secretion from beta cells by SCA with micro- and nanoelectrodes since the 1990s and to explain how and why serotonin is used as a proxy for monitoring insulin during exocytosis from single beta cells. Finally, we describe how the combination of SCA measurements with the intracellular vesicle impact electrochemical cytometry (IVIEC) technique has led to important findings regarding fractional release types in beta cells. These findings, reported recently, have opened a new window in the study of pore formation, exocytosis from single vesicles, and the mechanisms of insulin secretion. This sensitive cellular electroanalysis approach should help in the development of novel therapeutic strategies targeting diabetes in the future.
Collapse
Affiliation(s)
- Amir Hatamie
- Department
of Physiology, Sahlgrenska Academy, University
of Gothenburg, Medicinaregatan 11−13, 41390 Gothenburg, Sweden
- Department
of Chemistry and Molecular Biology, University
of Gothenburg, Kemivägen 10, 412 96, Gothenburg, Sweden
- Department
of Chemistry, Institute for Advanced Studies
in Basic Sciences (IASBS), Prof. Sobouti Boulevard, PO-Box 45195-1159, Zanjan, 45137-66731, Iran
| | - Xiulan He
- College
of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Andrew Ewing
- Department
of Physiology, Sahlgrenska Academy, University
of Gothenburg, Medicinaregatan 11−13, 41390 Gothenburg, Sweden
| | - Patrik Rorsman
- Department
of Physiology, Sahlgrenska Academy, University
of Gothenburg, Medicinaregatan 11−13, 41390 Gothenburg, Sweden
- Oxford
Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford OX3 7LJ, U.K.
| |
Collapse
|
41
|
Shrivastava A, Kumar A, Aggarwal LM, Pradhan S, Choudhary S, Ashish A, Kashyap K, Mishra S. Evolution of Bioelectric Membrane Potentials: Implications in Cancer Pathogenesis and Therapeutic Strategies. J Membr Biol 2024; 257:281-305. [PMID: 39183198 DOI: 10.1007/s00232-024-00323-2] [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: 05/30/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Electrophysiology typically deals with the electrical properties of excitable cells like neurons and muscles. However, all other cells (non-excitable) also possess bioelectric membrane potentials for intracellular and extracellular communications. These membrane potentials are generated by different ions present in fluids available in and outside the cell, playing a vital role in communication and coordination between the cell and its organelles. Bioelectric membrane potential variations disturb cellular ionic homeostasis and are characteristic of many diseases, including cancers. A rapidly increasing interest has emerged in sorting out the electrophysiology of cancer cells. Compared to healthy cells, the distinct electrical properties exhibited by cancer cells offer a unique way of understanding cancer development, migration, and progression. Decoding the altered bioelectric signals influenced by fluctuating electric fields benefits understanding cancer more closely. While cancer research has predominantly focussed on genetic and molecular traits, the delicate area of electrophysiological characteristics has increasingly gained prominence. This review explores the historical exploration of electrophysiology in the context of cancer cells, shedding light on how alterations in bioelectric membrane potentials, mediated by ion channels and gap junctions, contribute to the pathophysiology of cancer.
Collapse
Affiliation(s)
- Anju Shrivastava
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India.
| | - Amit Kumar
- Department of Anatomy, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Lalit Mohan Aggarwal
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyajit Pradhan
- Radiation Oncology, Mahamana Pandit Madhan Mohan Malaviya Cancer Centre, Varanasi, India
| | - Sunil Choudhary
- Radiotherapy and Radiation Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Ashish Ashish
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Keshav Kashyap
- Department of Physiology, Chhattisgarh Institute of Medical Sciences, Bilaspur, India
| | - Shivani Mishra
- Department of Anatomy, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
42
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
43
|
Bisht S, Singh MF. The triggering pathway, the metabolic amplifying pathway, and cellular transduction in regulation of glucose-dependent biphasic insulin secretion. Arch Physiol Biochem 2024; 130:854-865. [PMID: 38196246 DOI: 10.1080/13813455.2023.2299920] [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: 08/07/2023] [Revised: 12/08/2023] [Accepted: 12/16/2023] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Insulin secretion is a highly regulated process critical for maintaining glucose homeostasis. This abstract explores the intricate interplay between three essential pathways: The Triggering Pathway, The Metabolic Amplifying Pathway, and Cellular Transduction, in orchestrating glucose-dependent biphasic insulin secretion. MECHANISM During the triggering pathway, glucose metabolism in pancreatic beta-cells leads to ATP production, closing ATP-sensitive potassium channels and initiating insulin exocytosis. The metabolic amplifying pathway enhances insulin secretion via key metabolites like NADH and glutamate, enhancing calcium influx and insulin granule exocytosis. Additionally, the cellular transduction pathway involves G-protein coupled receptors and cyclic AMP, modulating insulin secretion. RESULT AND CONCLUSION These interconnected pathways ensure a dynamic insulin response to fluctuating glucose levels, with the initial rapid phase and the subsequent sustained phase. Understanding these pathways' complexities provides crucial insights into insulin dysregulation in diabetes and highlights potential therapeutic targets to restore glucose-dependent insulin secretion.
Collapse
Affiliation(s)
- Shradha Bisht
- Amity Institute of Pharmacy, Amity University, Lucknow, Uttar Pradesh, India
| | - Mamta F Singh
- School of Pharmaceutical Sciences, SBS University, Balawala, Uttarakhand, India
| |
Collapse
|
44
|
Liu S, Zou P. Recent Development of Chemigenetic Hybrid Voltage Indicators Enabled by Bioconjugation Chemistry. Bioconjug Chem 2024; 35:1711-1715. [PMID: 39474692 DOI: 10.1021/acs.bioconjchem.4c00383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Fluorescent voltage indicators enable the optical recording of electrophysiology across large cell populations with subcellular resolution; however, their application is often constrained by a limited photon budget. To address this limitation, advanced bioconjugation methods have been employed to site-specifically attach bright and photostable organic dyes to cell-specific protein scaffolds in live cells. The resulting chemigenetic hybrid voltage indicators enable sustained monitoring of voltage fluctuations with an exceptional signal-to-noise ratio, both in vitro and in vivo. This Viewpoint discusses recent advancements in the development of these indicators through bioconjugation chemistry.
Collapse
Affiliation(s)
- Shuzhang Liu
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
| | - Peng Zou
- College of Chemistry and Molecular Engineering, Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, Peking University, Beijing 100871, China
- PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research (CIBR), Beijing 102206, China
| |
Collapse
|
45
|
Sevastianov VI, Ponomareva AS, Baranova NV, Belova AD, Kirsanova LA, Nikolskaya AO, Kuznetsova EG, Chuykova EO, Skaletskiy NN, Skaletskaya GN, Nemets EA, Basok YB, Gautier SV. A Tissue-Engineered Construct Based on a Decellularized Scaffold and the Islets of Langerhans: A Streptozotocin-Induced Diabetic Model. Life (Basel) 2024; 14:1505. [PMID: 39598303 PMCID: PMC11595861 DOI: 10.3390/life14111505] [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: 10/04/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Producing a tissue-engineered pancreas based on a tissue-specific scaffold from a decellularized pancreas, imitating the natural pancreatic tissue microenvironment and the islets of Langerhans, is one of the approaches to treating patients with type 1 diabetes mellitus (T1DM). The aim of this work was to investigate the ability of a fine-dispersed tissue-specific scaffold (DP scaffold) from decellularized human pancreas fragments to support the islets' survival and insulin-producing function when injected in a streptozotocin-induced diabetic rat model. The developed decellularization protocol allows us to obtain a scaffold with a low DNA content (33 [26; 38] ng/mg of tissue, p < 0.05) and with the preservation of GAGs (0.92 [0.84; 1.16] µg/mg, p < 0.05) and fibrillar collagen (273.7 [241.2; 303.0] µg/mg, p < 0.05). Rat islets of Langerhans were seeded in the obtained scaffolds. The rats with stable T1DM were treated by intraperitoneal injections of rat islets alone and islets seeded on the DP scaffold. The blood glucose level was determined for 10 weeks with a histological examination of experimental animals' pancreas. A more pronounced decrease in the recipient rats' glycemia was detected after comparing the islets seeded on the DP scaffold with the control injection (by 71.4% and 51.2%, respectively). It has been shown that the DP scaffold facilitates a longer survival and the efficient function of pancreatic islets in vivo and can be used to engineer a pancreas.
Collapse
Affiliation(s)
- Victor I. Sevastianov
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Institute of Biomedical Research and Technology (IBRT), Autonomous Non-Profit Organization, 123557 Moscow, Russia
| | - Anna S. Ponomareva
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Natalia V. Baranova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Aleksandra D. Belova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Lyudmila A. Kirsanova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Alla O. Nikolskaya
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Eugenia G. Kuznetsova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Elizaveta O. Chuykova
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Nikolay N. Skaletskiy
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Galina N. Skaletskaya
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Evgeniy A. Nemets
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Yulia B. Basok
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
| | - Sergey V. Gautier
- The Shumakov National Medical Research Center of Transplantology and Artificial Organs, 123182 Moscow, Russia; (V.I.S.); (N.V.B.); (A.D.B.); (A.O.N.); (E.G.K.); (N.N.S.); (G.N.S.); (E.A.N.); (Y.B.B.); (S.V.G.)
- The Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| |
Collapse
|
46
|
Bellman J, Sjöros T, Hägg D, Atencio Herre E, Hieta J, Eskola O, Laitinen K, Nuutila P, Jansson JO, Jansson PA, Kalliokoski K, Roivainen A, Ohlsson C. Loading Enhances Glucose Uptake in Muscles, Bones, and Bone Marrow of Lower Extremities in Humans. J Clin Endocrinol Metab 2024; 109:3126-3136. [PMID: 38753869 PMCID: PMC11570666 DOI: 10.1210/clinem/dgae344] [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: 03/08/2024] [Revised: 04/29/2024] [Accepted: 05/15/2024] [Indexed: 05/18/2024]
Abstract
CONTEXT Increased standing time has been associated with improved health, but the underlying mechanism is unclear. OBJECTIVES We herein investigate if increased weight loading increases energy demand and thereby glucose uptake (GU) locally in bone and/or muscle in the lower extremities. METHODS In this single-center clinical trial with a randomized crossover design (ClinicalTrials.gov ID, NCT05443620), we enrolled 10 men with body mass index between 30 and 35 kg/m2. Participants were treated with both high load (standing with weight vest weighing 11% of body weight) and no load (sitting) on the lower extremities. GU was measured using whole-body quantitative positron emission tomography/computed tomography imaging. The primary endpoint was the change in GU ratio between loaded bones (ie, femur and tibia) and nonloaded bones (ie, humerus). RESULTS High load increased the GU ratio between lower and upper extremities in cortical diaphyseal bone (eg, femur/humerus ratio increased by 19%, P = .029), muscles (eg, m. quadriceps femoris/m. triceps brachii ratio increased by 28%, P = .014), and certain bone marrow regions (femur/humerus diaphyseal bone marrow region ratio increased by 17%, P = .041). Unexpectedly, we observed the highest GU in the bone marrow region of vertebral bodies, but its GU was not affected by high load. CONCLUSION Increased weight-bearing loading enhances GU in muscles, cortical bone, and bone marrow of the exposed lower extremities. This could be interpreted as increased local energy demand in bone and muscle caused by increased loading. The physiological importance of the increased local GU by static loading remains to be determined.
Collapse
Affiliation(s)
- Jakob Bellman
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Tanja Sjöros
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Daniel Hägg
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Erika Atencio Herre
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Janina Hieta
- Nutrition and Food Research Center and Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, Faculty of Medicine, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Olli Eskola
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Kirsi Laitinen
- Nutrition and Food Research Center and Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, Faculty of Medicine, University of Turku, FI-20014 Turun yliopisto, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
- Department of Endocrinology, Turku University Hospital, FI-20520 Turku, Finland
| | - John-Olov Jansson
- Department of Physiology, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, SE-41390 Gothenburg, Sweden
| | - Per-Anders Jansson
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
- Gothia Forum, Region Västra Götaland, Sahlgrenska University Hospital, SE-41346 Gothenburg, Sweden
| | - Kari Kalliokoski
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, FI-20014 Turun yliopisto, Finland
- Turku PET Centre, Turku University Hospital, FI-20520 Turku, Finland
- InFLAMES Research Flagship, University of Turku, FI-20014 Turku, Finland
| | - Claes Ohlsson
- Sahlgrenska Osteoporosis Centre, Center for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, SE-41345 Gothenburg, Sweden
- Department of Drug Treatment, Region Västra Götaland, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden
| |
Collapse
|
47
|
Jia R, Liang L, Yin Y, Niu C, Zhao X, Shuwen X, Zhang M, Yan X. Vitamin D supplementation could enhance the effectiveness of glibenclamide in treating type 2 diabetes by improving the function of pancreatic β-cells through the NF-κB pathway. Biochem Biophys Res Commun 2024; 733:150596. [PMID: 39197196 DOI: 10.1016/j.bbrc.2024.150596] [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: 08/14/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/01/2024]
Abstract
PURPOSE The high morbidity and mortality associated with type 2 diabetes mellitus (T2DM) pose a significant global health challenge, necessitating the development of more efficient anti-diabetic drugs with fewer side effects. This study investigated the intervention of vitamin D3 combined with glibenclamide in rats with T2DM to elucidate its effects on pancreatic β-cells through the NF-κB pathway. METHODS Twenty-four healthy male Sprague-Dawley (SD) rats were randomly assigned to four groups: the control group (CG), the model group (MG), the glibenclamide group (GG), and the glibenclamide + vitamin D3 group (GDG). After inducing the T2DM model using high-fat and high-sugar diet and intraperitoneal injection of streptozotocin, the rats in the GG group were administered glibenclamide orally (0.6 mg/kg/day), while those in the GDG group received both glibenclamide (0.6 mg/kg/day) and vitamin D3 (500 IU/kg/day) in corn oil for a duration of 8 weeks. Biochemical indices were measured, and histopathological changes in pancreatic tissue and islet β cells were observed using hematoxylin and eosin staining. The expression of pancreatic nuclear factor κB (NF-κB), islet β-cells, and inflammatory cytokines were assessed using the TUNEL method and PCR. RESULTS According to the data from this current study, the GDG group showed significant positive differences in plasma biochemical indices, as well as in the expression of β cells, NF-κB p65, TNF-α, IL-1β, INF-γ, and Fas, compared to the GG and CG groups (P < 0.05). CONCLUSION The results suggest that vitamin D has beneficial effects on T2DM by improving the functions of islet β cells through inhibition of the NF-κB signaling pathway. Therefore, it is suggested that vitamin D supplementation, when used alongside antidiabetic drugs, may more effectively prevent and treat T2DM.
Collapse
Affiliation(s)
- Ruimin Jia
- School of Nursing, Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Liting Liang
- Graduate School of Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Yongkai Yin
- Graduate School of Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Changmiao Niu
- Graduate School of Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Xingmei Zhao
- School of Nursing, Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Xue Shuwen
- School of Nursing, Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Mingzhi Zhang
- School of Nursing, Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China
| | - Xiaoning Yan
- Graduate School of Shanxi University of Chinese Medicine, No. 121, Daxue Street, Higher Education Park, Jinzhong City, Shanxi Province, 030619, China.
| |
Collapse
|
48
|
Salazar S, Delgadillo-Silva LF, Carapeto P, Dakessian K, Melhem R, Provencher-Girard A, Ostinelli G, Turgeon J, Kaci I, Migneault F, Huising MO, Hébert MJ, Rutter GA. Sex-dependent additive effects of dorzagliatin and incretin on insulin secretion in a novel mouse model of GCK-MODY. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.09.622781. [PMID: 39605321 PMCID: PMC11601264 DOI: 10.1101/2024.11.09.622781] [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: 11/29/2024]
Abstract
Glucokinase (GK) catalyses the key regulatory step in glucose-stimulated insulin secretion. Correspondingly, hetero- and homozygous mutations in human GCK cause maturity-onset diabetes of the young (GCK-MODY) and permanent neonatal diabetes (PNDM), respectively. To explore the possible utility of glucokinase activators (GKA) and of glucagon-like receptor-1 (GLP-1) agonists in these diseases, we have developed a novel hypomorphic Gck allele in mice encoding an aberrantly spliced mRNA deleted for exons 2 and 3. In islets from homozygous knock-in (GckKI/KI) mice, GK immunoreactivity was reduced by >85%, and glucose-stimulated insulin secretion eliminated. Homozygous GckKI/KI mice were smaller than wildtype littermates and displayed frank diabetes (fasting blood glucose >18 mmol/L; HbA1c ~12%), ketosis and nephropathy. Heterozygous GckKI/+ mice were glucose intolerant (HbA1c ~5.5%). Abnormal glucose-stimulated Ca2+ dynamics and beta cell-beta cell connectivity in GckKI/+ islets were completely reversed by the recently-developed GKA, dorzagliatin, which was largely inactive in homozygous GckKI/KI mouse islets. The GLP-1 receptor agonist exendin-4 improved glucose tolerance in male GckKI/+ mice, an action potentiated by dorzagliatin, in male but not female mice. Sex-dependent additive effects of these agents were also observed on insulin secretion in vitro. Combined treatment with GKA and incretin may thus be useful in GCK-MODY or GCK-PNDM.
Collapse
Affiliation(s)
| | | | | | | | - Rana Melhem
- CR-CHUM and University of Montreal, QC, Canada
| | | | | | | | - Imane Kaci
- CR-CHUM and University of Montreal, QC, Canada
| | | | - Mark O Huising
- Department of Neurobiology, Physiology and Behavior, College of Biological Sciences, University of California, Davis, Davis, CA
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA
| | - Marie-Josée Hébert
- CR-CHUM and University of Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université de Montréal, Montréal, QC, Canada
- Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Guy A Rutter
- CR-CHUM and University of Montreal, QC, Canada
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, Hammersmith Hospital, W12 ONN London U.K
- Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Lee Kong Chian School of Medicine, Nanyang Technological College, Singapore
| |
Collapse
|
49
|
Wevers A, San Roman-Mata S, Navarro-Ledesma S, Pruimboom L. The Role of Insulin Within the Socio-Psycho-Biological Framework in Type 2 Diabetes-A Perspective from Psychoneuroimmunology. Biomedicines 2024; 12:2539. [PMID: 39595105 PMCID: PMC11591609 DOI: 10.3390/biomedicines12112539] [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: 06/30/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
The interplay between socio-psychological factors and biological systems is pivotal in defining human health and disease, particularly in chronic non-communicable diseases. Recent advancements in psychoneuroimmunology and mitochondrial psychobiology have emphasized the significance of psychological factors as critical determinants of disease onset, progression, recurrence, and severity. These insights align with evolutionary biology, psychology, and psychiatry, highlighting the inherent social nature of humans. This study proposes a theory that expands insulin's role beyond traditional metabolic functions, incorporating it into the Mitochondrial Information Processing System (MIPS) and exploring it from an evolutionary medicine perspective to explore its function in processing psychological and social factors into biological responses. This narrative review comprises data from preclinical animal studies, longitudinal cohort studies, cross-sectional studies, machine learning analyses, and randomized controlled trials, and investigates the role of insulin in health and disease. The result is a proposal for a theoretical framework of insulin as a social substance within the socio-psycho-biological framework, emphasizing its extensive roles in health and disease. Type 2 Diabetes Mellitus (T2DM) with musculoskeletal disorders and neurodegeneration exemplifies this narrative. We suggest further research towards a comprehensive treatment protocol meeting evolutionary expectations, where incorporating psychosocial interventions plays an essential role. By supporting the concept of 'insulin resilience' and suggesting the use of heart rate variability to assess insulin resilience, we aim to provide an integrative approach to managing insulin levels and monitoring the effectiveness of interventions. This integrative strategy addresses broader socio-psychological factors, ultimately improving health outcomes for individuals with T2DM and musculoskeletal complications and neurodegeneration while providing new insights into the interplay between socio-psychological factors and biological systems in chronic diseases.
Collapse
Affiliation(s)
- Anne Wevers
- Clinical Medicine and Public Health PhD Program, Faculty of Health Sciences, University of Granada, 18071 Granada, Spain;
| | - Silvia San Roman-Mata
- Department of Nursing, Faculty of Health Sciences, Campus of Melilla, University of Granada, 52004 Melilla, Spain;
| | - Santiago Navarro-Ledesma
- Department of Physical Therapy, Faculty of Health Sciences, Campus of Melilla, University of Granada, 52004 Melilla, Spain
- University Chair in Clinical Psychoneuroimmunology, Campus of Melilla, University of Granada and PNI Europe, 52004 Melilla, Spain;
| | - Leo Pruimboom
- University Chair in Clinical Psychoneuroimmunology, Campus of Melilla, University of Granada and PNI Europe, 52004 Melilla, Spain;
| |
Collapse
|
50
|
Sokolowski EK, Kursawe R, Selvam V, Bhuiyan RM, Thibodeau A, Zhao C, Spracklen CN, Ucar D, Stitzel ML. Multi-omic human pancreatic islet endoplasmic reticulum and cytokine stress response mapping provides type 2 diabetes genetic insights. Cell Metab 2024; 36:2468-2488.e7. [PMID: 39383866 PMCID: PMC11798411 DOI: 10.1016/j.cmet.2024.09.006] [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: 12/22/2023] [Revised: 06/14/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Endoplasmic reticulum (ER) and inflammatory stress responses contribute to islet dysfunction in type 2 diabetes (T2D). Comprehensive genomic understanding of these human islet stress responses and whether T2D-associated genetic variants modulate them is lacking. Here, comparative transcriptome and epigenome analyses of human islets exposed ex vivo to these stressors revealed 30% of expressed genes and 14% of islet cis-regulatory elements (CREs) as stress responsive, modulated largely in an ER- or cytokine-specific fashion. T2D variants overlapped 86 stress-responsive CREs, including 21 induced by ER stress. We linked the rs6917676-T T2D risk allele to increased islet ER-stress-responsive CRE accessibility and allele-specific β cell nuclear factor binding. MAP3K5, the ER-stress-responsive putative rs6917676 T2D effector gene, promoted stress-induced β cell apoptosis. Supporting its pro-diabetogenic role, MAP3K5 expression correlated inversely with human islet β cell abundance and was elevated in T2D β cells. This study provides genome-wide insights into human islet stress responses and context-specific T2D variant effects.
Collapse
Affiliation(s)
- Eishani K Sokolowski
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Romy Kursawe
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Vijay Selvam
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Redwan M Bhuiyan
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Asa Thibodeau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Chi Zhao
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Cassandra N Spracklen
- Department of Biostatistics and Epidemiology, University of Massachusetts, Amherst, Amherst, MA 01003, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| | - Michael L Stitzel
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA; Institute of Systems Genomics, University of Connecticut, Farmington, CT 06032, USA.
| |
Collapse
|