1
|
Ernst IVS, Lehtonen L, Nilsson SM, Nielsen FL, Marcher AB, Mandrup S, Madsen JGS. Single Nucleus Multiome Analysis Reveals Early Inflammatory Response to High-Fat Diet in Mouse Pancreatic Islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646568. [PMID: 40236154 PMCID: PMC11996447 DOI: 10.1101/2025.04.01.646568] [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
In periods of sustained hyper-nutrition, pancreatic β-cells undergo functional compensation through transcriptional upregulation of gene programs driving insulin secretion. This adaptation is essential for maintaining systemic glucose homeostasis and metabolic health. Using single nuclei multiomics, we have mapped the early transcriptional compensation mechanisms in murine islets of Langerhans exposed to high-fat diet (HFD) for one and three weeks. We show that β-cells exhibit the largest transcriptional response to HFD, characterized by early activation of proinflammatory eRegulons and downregulation of β-cell identity genes, particularly in a distinct subset of β-cells. Our observations translate to humans, as we observe an increase in the inflammatory gene signatures in human β-cells in pre-diabetes and diabetes. Collectively, these observations point to cellular cross-talk through proinflammatory signaling as a central and early driver of β-cell dysfunction that limits the compensatory capacity of β-cells, which is closely linked to the development of diabetes.
Collapse
|
2
|
York NW, Yan Z, Osipovich AB, Tate A, Patel S, Piston DW, Magnuson MA, Remedi MS, Nichols CG. Loss of β-Cell KATP Reduces Ca2+ Sensitivity of Insulin Secretion and Trpm5 Expression. Diabetes 2025; 74:376-383. [PMID: 39666394 PMCID: PMC11842610 DOI: 10.2337/db24-0650] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
Loss-of-function mutations in ATP-sensitive potassium (KATP) channels cause hyperexcitability and insulin hypersecretion, resulting in congenital hyperinsulinism (CHI). Paradoxically, despite the initial insulin hypersecretion, many CHI cases, as well as KATP knockout (KO) animals, eventually "crossover" to undersecretion and even diabetes. Here, we confirm that Sur1 KO islets exhibit higher intracellular concentration of calcium ion ([Ca2+]i) at all concentrations of glucose but show decreased glucose-stimulated insulin secretion. However, when [Ca2+]i is artificially elevated by increasing extracellular [Ca2+], insulin secretion from Sur1 KO islets increases to the same levels as in wild-type (WT) islets. This indicates that a right-shift in [Ca2+]i dependence of insulin secretion, rather than loss of insulin content or intrinsic secretability, is the primary cause for the crossover. Chronic pharmacological inhibition of KATP channel activity by slow release of glibenclamide in pellet-implanted mice causes a very similar crossover to glucose intolerance and impaired insulin secretion seen in Sur1 KO animals. Whole-islet and single-cell transcriptomic analysis reveal markedly reduced Trpm5 in both conditions. Glibenclamide pellet-implanted Trpm5 KO mice also exhibited significant glucose intolerance. However, this was not as severe as in WT animals, which suggests decreased expression of Trpm5 may play a small role in the disruption of insulin secretion with KATP loss. ARTICLE HIGHLIGHTS Congenital hyperinsulinism caused by loss of ATP-sensitive potassium (KATP) channels crosses over to unexplained undersecretion. Why does loss of β-cell KATP channel activity result in undersecretion of insulin and glucose tolerance, despite elevated intracellular concentration of calcium ion ([Ca2+]i) levels? Superelevation of [Ca2+]i in supraphysiological extracellular [Ca2+] boosted secretion from Sur1 knockout (KO) islets to the same levels as WT, indicating a right-shift in [Ca2+]i dependence of secretion. Transcriptomic analysis revealed markedly reduced β-cell Trpm5 in the absence of KATP. KATP inhibition in Trpm5 KO mice still caused significant glucose intolerance, but slightly less severe than in WT animals. Right-shifted [Ca2+]i dependence of secretion explains crossover. Downregulation of Trpm5 may be involved.
Collapse
Affiliation(s)
- Nathaniel W. York
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Zihan Yan
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Anna B. Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Abbie Tate
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Sumit Patel
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - David W. Piston
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Mark A. Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Maria S. Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Medicine, Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO
| | - Colin G. Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
3
|
Magnuson MA, Osipovich AB. Ca 2+ signaling and metabolic stress-induced pancreatic β-cell failure. Front Endocrinol (Lausanne) 2024; 15:1412411. [PMID: 39015185 PMCID: PMC11250477 DOI: 10.3389/fendo.2024.1412411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Early in the development of Type 2 diabetes (T2D), metabolic stress brought on by insulin resistance and nutrient overload causes β-cell hyperstimulation. Herein we summarize recent studies that have explored the premise that an increase in the intracellular Ca2+ concentration ([Ca2+]i), brought on by persistent metabolic stimulation of β-cells, causes β-cell dysfunction and failure by adversely affecting β-cell function, structure, and identity. This mini-review builds on several recent reviews that also describe how excess [Ca2+]i impairs β-cell function.
Collapse
Affiliation(s)
- Mark A. Magnuson
- Department of Molecular Physiology and Biophysics and Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, United States
| | | |
Collapse
|
4
|
Turbitt J, Moffett RC, Brennan L, Johnson PRV, Flatt PR, McClenaghan NH, Tarasov AI. Molecular determinants and intracellular targets of taurine signalling in pancreatic islet β-cells. Acta Physiol (Oxf) 2024; 240:e14101. [PMID: 38243723 DOI: 10.1111/apha.14101] [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: 06/26/2023] [Revised: 12/26/2023] [Accepted: 01/08/2024] [Indexed: 01/21/2024]
Abstract
AIM Despite its abundance in pancreatic islets of Langerhans and proven antihyperglycemic effects, the impact of the essential amino acid, taurine, on islet β-cell biology has not yet received due consideration, which prompted the current studies exploring the molecular selectivity of taurine import into β-cells and its acute and chronic intracellular interactions. METHODS The molecular aspects of taurine transport were probed by exposing the clonal pancreatic BRIN BD11 β-cells and primary mouse and human islets to a range of the homologs of the amino acid (assayed at 2-20 mM), using the hormone release and imaging of intracellular signals as surrogate read-outs. Known secretagogues were employed to profile the interaction of taurine with acute and chronic intracellular signals. RESULTS Taurine transporter TauT was expressed in the islet β-cells, with the transport of taurine and homologs having a weak sulfonate specificity but significant sensitivity to the molecular weight of the transporter. Taurine, hypotaurine, homotaurine, and β-alanine enhanced insulin secretion in a glucose-dependent manner, an action potentiated by cytosolic Ca2+ and cAMP. Acute and chronic β-cell insulinotropic effects of taurine were highly sensitive to co-agonism with GLP-1, forskolin, tolbutamide, and membrane depolarization, with an unanticipated indifference to the activation of PKC and CCK8 receptors. Pre-culturing with GLP-1 or KATP channel inhibitors sensitized or, respectively, desensitized β-cells to the acute taurine stimulus. CONCLUSION Together, these data demonstrate the pathways whereby taurine exhibits a range of beneficial effects on insulin secretion and β-cell function, consistent with the antidiabetic potential of its dietary low-dose supplementation.
Collapse
Affiliation(s)
- Julie Turbitt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | | | - Lorraine Brennan
- UCD Institute of Food and Health, UCD School of Agriculture and Food Science, University College Dublin, Dublin 4, Republic of Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Republic of Ireland
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK
- Oxford Biomedical Research Centre (OxBRC), Oxford, UK
| | - Peter R Flatt
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - Neville H McClenaghan
- School of Biomedical Sciences, Ulster University, Coleraine, UK
- Department of Life Sciences, Atlantic Technological University, Sligo, Republic of Ireland
| | | |
Collapse
|
5
|
Sue N, Thai LM, Saito A, Boyer CK, Fordham AM, Yan C, Davenport A, Tao J, Bensellam M, Cantley J, Shi YC, Stephens SB, Imaizumi K, Biden TJ. Independent activation of CREB3L2 by glucose fills a regulatory gap in mouse β-cells by co-ordinating insulin biosynthesis with secretory granule formation. Mol Metab 2024; 79:101845. [PMID: 38013154 PMCID: PMC10755490 DOI: 10.1016/j.molmet.2023.101845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
OBJECTIVE Although individual steps have been characterized, there is little understanding of the overall process whereby glucose co-ordinates the biosynthesis of insulin with its export out of the endoplasmic reticulum (ER) and incorporation into insulin secretory granules (ISGs). Here we investigate a role for the transcription factor CREB3L2 in this context. METHODS MIN6 cells and mouse islets were analysed by immunoblotting after treatment with glucose, fatty acids, thapsigargin and various inhibitors. Knockdown of CREB3L2 was achieved using si or sh constructs by transfection, or viral delivery. In vivo metabolic phenotyping was conducted after deletion of CREB3L2 in β-cells of adult mice using Ins1-CreER+. Islets were isolated for RNAseq and assays of glucose-stimulated insulin secretion (GSIS). Trafficking was monitored in islet monolayers using a GFP-tagged proinsulin construct that allows for synchronised release from the ER. RESULTS With a Km ≈3.5 mM, glucose rapidly (T1/2 0.9 h) increased full length (FL) CREB3L2 followed by a slower rise (T1/2 2.5 h) in its transcriptionally-active cleavage product, P60 CREB3L2. Glucose stimulation repressed the ER stress marker, CHOP, and this was partially reverted by knockdown of CREB3L2. Activation of CREB3L2 by glucose was not due to ER stress, however, but a combination of O-GlcNAcylation, which impaired proteasomal degradation of FL-CREB3L2, and mTORC1 stimulation, which enhanced its conversion to P60. cAMP generation also activated CREB3L2, but independently of glucose. Deletion of CREB3L2 inhibited GSIS ex vivo and, following a high-fat diet (HFD), impaired glucose tolerance and insulin secretion in vivo. RNAseq revealed that CREB3L2 regulated genes controlling trafficking to-and-from the Golgi, as well as a broader cohort associated with β-cell compensation during a HFD. Although post-Golgi trafficking appeared intact, knockdown of CREB3L2 impaired the generation of both nascent ISGs and proinsulin condensates in the Golgi, implying a defect in ER export of proinsulin and/or its processing in the Golgi. CONCLUSION The stimulation of CREB3L2 by glucose defines a novel, rapid and direct mechanism for co-ordinating the synthesis, packaging and storage of insulin, thereby minimizing ER overload and optimizing β-cell function under conditions of high secretory demand. Upregulation of CREB3L2 also potentially contributes to the benefits of GLP1 agonism and might in itself constitute a novel means of treating β-cell failure.
Collapse
Affiliation(s)
- Nancy Sue
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Le May Thai
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Atsushi Saito
- Department of Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center, Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Ashleigh M Fordham
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Chenxu Yan
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Aimee Davenport
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Jiang Tao
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Mohammed Bensellam
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - James Cantley
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia
| | - Yan-Chuan Shi
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, Department of Internal Medicine, University of Iowa, Iowa City, IA 52246, USA
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Trevor J Biden
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine, The University of New South Wales, Sydney, Australia.
| |
Collapse
|
6
|
Osipovich AB, Zhou FY, Chong JJ, Trinh LT, Cottam MA, Shrestha S, Cartailler JP, Magnuson MA. Deletion of Ascl1 in pancreatic β-cells improves insulin secretion, promotes parasympathetic innervation, and attenuates dedifferentiation during metabolic stress. Mol Metab 2023; 78:101811. [PMID: 37769990 PMCID: PMC10570713 DOI: 10.1016/j.molmet.2023.101811] [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: 08/02/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVE ASCL1, a pioneer transcription factor, is essential for neural cell differentiation and function. Previous studies have shown that Ascl1 expression is increased in pancreatic β-cells lacking functional KATP channels or after feeding of a high fat diet (HFD) suggesting that it may contribute to the metabolic stress response of β-cells. METHODS We generated β-cell-specific Ascl1 knockout mice (Ascl1βKO) and assessed their glucose homeostasis, islet morphology and gene expression after feeding either a normal diet or HFD for 12 weeks, or in combination with a genetic disruption of Abcc8, an essential KATP channel component. RESULTS Ascl1 expression is increased in response to both a HFD and membrane depolarization and requires CREB-dependent Ca2+ signaling. No differences in glucose homeostasis or islet morphology were observed in Ascl1βKO mice fed a normal diet or in the absence of KATP channels. However, male Ascl1βKO mice fed a HFD exhibited decreased blood glucose levels, improved glucose tolerance, and increased β-cell proliferation. Bulk RNA-seq analysis of islets from Ascl1βKO mice from three studied conditions showed alterations in genes associated with the secretory function. HFD-fed Ascl1βKO mice showed the most extensive changes with increased expression of genes necessary for glucose sensing, insulin secretion and β-cell proliferation, and a decrease in genes associated with β-cell dysfunction, inflammation and dedifferentiation. HFD-fed Ascl1βKO mice also displayed increased expression of parasympathetic neural markers and cholinergic receptors that was accompanied by increased insulin secretion in response to acetylcholine and an increase in islet innervation. CONCLUSIONS Ascl1 expression is induced by stimuli that cause Ca2+-signaling to the nucleus and contributes in a multifactorial manner to the loss of β-cell function by promoting the expression of genes associated with cellular dedifferentiation, attenuating β-cells proliferation, suppressing acetylcholine sensitivity, and repressing parasympathetic innervation of islets. Thus, the removal of Ascl1 from β-cells improves their function in response to metabolic stress.
Collapse
Affiliation(s)
- Anna B Osipovich
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Frank Y Zhou
- College of Arts and Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Judy J Chong
- College of Arts and Sciences, Vanderbilt University, Nashville, TN 37232, USA
| | - Linh T Trinh
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Mathew A Cottam
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Shristi Shrestha
- Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA
| | | | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA.
| |
Collapse
|
7
|
Hou X, Chen Y, Zhou B, Tang W, Ding Z, Chen L, Wu Y, Yang H, Du C, Yang D, Ma G, Cao H. Talin-1 inhibits Smurf1-mediated Stat3 degradation to modulate β-cell proliferation and mass in mice. Cell Death Dis 2023; 14:709. [PMID: 37903776 PMCID: PMC10616178 DOI: 10.1038/s41419-023-06235-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2023]
Abstract
Insufficient pancreatic β-cell mass and reduced insulin expression are key events in the pathogenesis of diabetes mellitus (DM). Here we demonstrate the high expression of Talin-1 in β-cells and that deficiency of Talin-1 reduces β-cell proliferation, which leads to reduced β-cell mass and insulin expression, thus causing glucose intolerance without affecting peripheral insulin sensitivity in mice. High-fat diet fed exerbates these phenotypes. Mechanistically, Talin-1 interacts with the E3 ligase smad ubiquitination regulatory factor 1 (Smurf1), which prohibits ubiquitination of the signal transducer and activator of transcription 3 (Stat3) mediated by Smurf1, and ablation of Talin-1 enhances Smurf1-mediated ubiquitination of Stat3, leading to decreased β-cell proliferation and mass. Furthermore, haploinsufficiency of Talin-1 and Stat3 genes, but not that of either gene, in β-cell in mice significantly impairs glucose tolerance and insulin expression, indicating that both factors indeed function in the same genetic pathway. Finally, inducible deletion Talin-1 in β-cell causes glucose intolerance in adult mice. Collectively, our findings reveal that Talin-1 functions as a crucial regulator of β-cell mass, and highlight its potential as a therapeutic target for DM patients.
Collapse
Affiliation(s)
- Xiaoting Hou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yangshan Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Bo Zhou
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wanze Tang
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhen Ding
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Litong Chen
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yun Wu
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital; Guangdong Provincial High-level Clinical Key Specialty; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University; The Hong Kong University of Science and Technology Medical Center, Guangdong, China
| | - Hongyu Yang
- Department of Oral and Maxillofacial Surgery, Stomatological Center, Peking University Shenzhen Hospital; Guangdong Provincial High-level Clinical Key Specialty; Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment; The Institute of Stomatology, Peking University Shenzhen Hospital, Shenzhen Peking University; The Hong Kong University of Science and Technology Medical Center, Guangdong, China
| | - Changzheng Du
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dazhi Yang
- The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Guixing Ma
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Huiling Cao
- Department of Biochemistry, School of Medicine, Southern University of Science and Technology, Key University Laboratory of Metabolism and Health of Guangdong, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
8
|
Carroll DT, Elsakr JM, Miller A, Fuhr J, Lindsley SR, Kirigiti M, Takahashi DL, Dean TA, Wesolowski SR, McCurdy CE, Friedman JE, Aagaard KM, Kievit P, Gannon M. Maternal Western-style diet in nonhuman primates leads to offspring islet adaptations including altered gene expression and insulin hypersecretion. Am J Physiol Endocrinol Metab 2023; 324:E577-E588. [PMID: 37134140 PMCID: PMC10259856 DOI: 10.1152/ajpendo.00087.2023] [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: 03/22/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Maternal overnutrition is associated with increased susceptibility to type 2 diabetes in the offspring. Rodent models have shown that maternal overnutrition influences islet function in offspring. To determine whether maternal Western-style diet (WSD) alters prejuvenile islet function in a model that approximates that of human offspring, we utilized a well-characterized Japanese macaque model. We compared islet function from offspring exposed to WSD throughout pregnancy and lactation and weaned to WSD (WSD/WSD) compared with islets from offspring exposed only to postweaning WSD (CD/WSD) at 1 yr of age. WSD/WSD offspring islets showed increased basal insulin secretion and an exaggerated increase in glucose-stimulated insulin secretion, as assessed by dynamic ex vivo perifusion assays, relative to CD/WSD-exposed offspring. We probed potential mechanisms underlying insulin hypersecretion using transmission electron microscopy to evaluate β-cell ultrastructure, qRT-PCR to quantify candidate gene expression, and Seahorse assay to assess mitochondrial function. Insulin granule density, mitochondrial density, and mitochondrial DNA ratio were similar between groups. However, islets from WSD/WSD male and female offspring had increased expression of transcripts known to facilitate stimulus-secretion coupling and changes in the expression of cell stress genes. Seahorse assay revealed increased spare respiratory capacity in islets from WSD/WSD male offspring. Overall, these results show that maternal WSD feeding confers changes to genes governing insulin secretory coupling and results in insulin hypersecretion as early as the postweaning period. The results suggest a maternal diet leads to early adaptation and developmental programming in offspring islet genes that may underlie future β-cell dysfunction.NEW & NOTEWORTHY Programed adaptations in islets in response to maternal WSD exposure may alter β-cell response to metabolic stress in offspring. We show that islets from maternal WSD-exposed offspring hypersecrete insulin, possibly due to increased components of stimulus-secretion coupling. These findings suggest that islet hyperfunction is programed by maternal diet, and changes can be detected as early as the postweaning period in nonhuman primate offspring.
Collapse
Affiliation(s)
- Darian T Carroll
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Joseph M Elsakr
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Allie Miller
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer Fuhr
- Department of Veterans Affairs Tennessee Valley, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sarah Rene Lindsley
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Melissa Kirigiti
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Stephanie R Wesolowski
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma, United States
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Veterans Affairs Tennessee Valley, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
9
|
Nichols CG, York NW, Remedi MS. ATP-Sensitive Potassium Channels in Hyperinsulinism and Type 2 Diabetes: Inconvenient Paradox or New Paradigm? Diabetes 2022; 71:367-375. [PMID: 35196393 PMCID: PMC8893938 DOI: 10.2337/db21-0755] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/28/2021] [Indexed: 11/13/2022]
Abstract
Secretion of insulin from pancreatic β-cells is complex, but physiological glucose-dependent secretion is dominated by electrical activity, in turn controlled by ATP-sensitive potassium (KATP) channel activity. Accordingly, loss-of-function mutations of the KATP channel Kir6.2 (KCNJ11) or SUR1 (ABCC8) subunit increase electrical excitability and secretion, resulting in congenital hyperinsulinism (CHI), whereas gain-of-function mutations cause underexcitability and undersecretion, resulting in neonatal diabetes mellitus (NDM). Thus, diazoxide, which activates KATP channels, and sulfonylureas, which inhibit KATP channels, have dramatically improved therapies for CHI and NDM, respectively. However, key findings do not fit within this simple paradigm: mice with complete absence of β-cell KATP activity are not hyperinsulinemic; instead, they are paradoxically glucose intolerant and prone to diabetes, as are older human CHI patients. Critically, despite these advances, there has been little insight into any role of KATP channel activity changes in the development of type 2 diabetes (T2D). Intriguingly, the CHI progression from hypersecretion to undersecretion actually mirrors the classical response to insulin resistance in the progression of T2D. In seeking to explain the progression of CHI, multiple lines of evidence lead us to propose that underlying mechanisms are also similar and that development of T2D may involve loss of KATP activity.
Collapse
Affiliation(s)
- Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Nathaniel W York
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO
| | - Maria S Remedi
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, MO
- Division of Endocrinology Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
10
|
Benderradji H, Kraiem S, Courty E, Eddarkaoui S, Bourouh C, Faivre E, Rolland L, Caron E, Besegher M, Oger F, Boschetti T, Carvalho K, Thiroux B, Gauvrit T, Nicolas E, Gomez-Murcia V, Bogdanova A, Bongiovanni A, Muhr-Tailleux A, Lancel S, Bantubungi K, Sergeant N, Annicotte JS, Buée L, Vieau D, Blum D, Buée-Scherrer V. Impaired Glucose Homeostasis in a Tau Knock-In Mouse Model. Front Mol Neurosci 2022; 15:841892. [PMID: 35250480 PMCID: PMC8889017 DOI: 10.3389/fnmol.2022.841892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/21/2022] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia. While impaired glucose homeostasis has been shown to increase AD risk and pathological loss of tau function, the latter has been suggested to contribute to the emergence of the glucose homeostasis alterations observed in AD patients. However, the links between tau impairments and glucose homeostasis, remain unclear. In this context, the present study aimed at investigating the metabolic phenotype of a new tau knock-in (KI) mouse model, expressing, at a physiological level, a human tau protein bearing the P301L mutation under the control of the endogenous mouse Mapt promoter. Metabolic investigations revealed that, while under chow diet tau KI mice do not exhibit significant metabolic impairments, male but not female tau KI animals under High-Fat Diet (HFD) exhibited higher insulinemia as well as glucose intolerance as compared to control littermates. Using immunofluorescence, tau protein was found colocalized with insulin in the β cells of pancreatic islets in both mouse (WT, KI) and human pancreas. Isolated islets from tau KI and tau knock-out mice exhibited impaired glucose-stimulated insulin secretion (GSIS), an effect recapitulated in the mouse pancreatic β-cell line (MIN6) following tau knock-down. Altogether, our data indicate that loss of tau function in tau KI mice and, particularly, dysfunction of pancreatic β cells might promote glucose homeostasis impairments and contribute to metabolic changes observed in AD.
Collapse
Affiliation(s)
- Hamza Benderradji
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Sarra Kraiem
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Courty
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Sabiha Eddarkaoui
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Cyril Bourouh
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Emilie Faivre
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Laure Rolland
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Emilie Caron
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Development and Plasticity of the Neuroendocrine Brain, Lille, France
| | - Mélanie Besegher
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, Animal Facility, Lille, France
| | - Frederik Oger
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Theo Boschetti
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Kévin Carvalho
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Bryan Thiroux
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Thibaut Gauvrit
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Emilie Nicolas
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Victoria Gomez-Murcia
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Anna Bogdanova
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Antonino Bongiovanni
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41—UMS 2014—PLBS, BioImaging Center Lille, Lille, France
| | - Anne Muhr-Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Steve Lancel
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167—RID-AGE—Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France
| | - Kadiombo Bantubungi
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Nicolas Sergeant
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Jean-Sebastien Annicotte
- Univ. Lille, INSERM, CNRS, CHU Lille, Institut Pasteur de Lille, Inserm U1283-UMR8199—EGID, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Didier Vieau
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - David Blum
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
- *Correspondence: David Blum
| | - Valérie Buée-Scherrer
- Univ. Lille, Inserm, CHU Lille, U1172 LilNCog—Lille Neuroscience & Cognition, Lille, France
- Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|
11
|
Screening of probiotics with efficient α-glucosidase inhibitory ability and study on the structure and function of its extracellular polysaccharide. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2021.101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
12
|
Tong X, Stein R. Lipid Droplets Protect Human β-Cells From Lipotoxicity-Induced Stress and Cell Identity Changes. Diabetes 2021; 70:2595-2607. [PMID: 34433630 PMCID: PMC8564404 DOI: 10.2337/db21-0261] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 08/19/2021] [Indexed: 12/12/2022]
Abstract
Free fatty acids (FFAs) are often stored in lipid droplet (LD) depots for eventual metabolic and/or synthetic use in many cell types, such a muscle, liver, and fat. In pancreatic islets, overt LD accumulation was detected in humans but not mice. LD buildup in islets was principally observed after roughly 11 years of age, increasing throughout adulthood under physiologic conditions, and also enriched in type 2 diabetes. To obtain insight into the role of LDs in human islet β-cell function, the levels of a key LD scaffold protein, perilipin 2 (PLIN2), were manipulated by lentiviral-mediated knockdown (KD) or overexpression (OE) in EndoCβH2-Cre cells, a human cell line with adult islet β-like properties. Glucose-stimulated insulin secretion was blunted in PLIN2KD cells and improved in PLIN2OE cells. An unbiased transcriptomic analysis revealed that limiting LD formation induced effectors of endoplasmic reticulum (ER) stress that compromised the expression of critical β-cell function and identity genes. These changes were essentially reversed by PLIN2OE or using the ER stress inhibitor, tauroursodeoxycholic acid. These results strongly suggest that LDs are essential for adult human islet β-cell activity by preserving FFA homeostasis.
Collapse
Affiliation(s)
- Xin Tong
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Roland Stein
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| |
Collapse
|
13
|
Nakamura A, Omori K, Terauchi Y. Glucokinase activation or inactivation: Which will lead to the treatment of type 2 diabetes? Diabetes Obes Metab 2021; 23:2199-2206. [PMID: 34105236 DOI: 10.1111/dom.14459] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/24/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022]
Abstract
Glucokinase, which phosphorylates glucose to form glucose-6-phosphate, plays a critical role in regulating blood glucose levels. On the basis of data of glucokinase-knockout and transgenic mice and humans with glucokinase mutations, glucokinase was targeted for drug development aiming to augment its activity, and thereby reduce hyperglycaemia in patients with diabetes. In fact, various small molecule compounds have been developed and clinically tested as glucokinase activators. However, some have been discontinued because of efficacy and safety issues. One of these issues is loss of the drug's efficacy over time. This unsustained glycaemic efficacy may be associated with the excess glycolysis by glucokinase activation in pancreatic beta cells, resulting in beta-cell failure. Recently, we have shown that glucokinase haploinsufficiency ameliorated glucose intolerance by increasing beta-cell function and mass in a mouse model of diabetes. Given that a similar phenotype has been observed in glucokinase-activated beta cells and diabetic beta cells, glucokinase inactivation may be a new therapeutic target for type 2 diabetes.
Collapse
Affiliation(s)
- Akinobu Nakamura
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuno Omori
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| |
Collapse
|
14
|
Brown ML, Lopez A, Meyer N, Richter A, Thompson TB. FSTL3-Neutralizing Antibodies Enhance Glucose-Responsive Insulin Secretion in Dysfunctional Male Mouse and Human Islets. Endocrinology 2021; 162:6128796. [PMID: 33539535 PMCID: PMC8384134 DOI: 10.1210/endocr/bqab019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/23/2022]
Abstract
Diabetes is caused by insufficient insulin production from pancreatic beta cells or insufficient insulin action, leading to an inability to control blood glucose. While a wide range of treatments exist to alleviate the symptoms of diabetes, therapies addressing the root cause of diabetes through replacing lost beta cells with functional cells remain an object of active pursuit. We previously demonstrated that genetic deletion of Fstl3, a critical regulator of activin activity, enhanced beta cell number and glucose-responsive insulin production. These observations suggested the hypothesis that FSTL3 neutralization could be used to therapeutically enhance beta cell number and function in humans. To pursue this possibility, we developed an FSTL3-neutralizing antibody, FP-101, and characterized its ability to prevent or disrupt FSTL3 from complexing with activin or related ligands. This antibody was selective for FSTL3 relative to the closely related follistatin, thereby reducing the chance for off-target effects. In vitro assays with FP-101 and activin revealed that FP-101-mediated neutralization of FSTL3 can enhance both insulin secretion and glucose responsiveness to nonfunctional mouse and human islets under conditions that model diabetes. Thus, FSTL3 neutralization may provide a novel therapeutic strategy for treating diabetes through repairing dysfunctional beta cells.
Collapse
Affiliation(s)
- Melissa L Brown
- Department of Nutrition and Public Health, University of Saint Joseph, West Hartford, CT 06117, USA
- Correspondence: Melissa Brown, PhD, RD, CSSD, LD, University of Saint Joseph, 1678 Asylum Ave, West Hartford, CT 06117, USA. E-mail:
| | - Alexa Lopez
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Nolan Meyer
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Alden Richter
- Fairbanks Pharmaceuticals, Inc., Concord, MA 01742, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
15
|
Satin LS, Soleimanpour SA, Walker EM. New Aspects of Diabetes Research and Therapeutic Development. Pharmacol Rev 2021; 73:1001-1015. [PMID: 34193595 PMCID: PMC8274312 DOI: 10.1124/pharmrev.120.000160] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Both type 1 and type 2 diabetes mellitus are advancing at exponential rates, placing significant burdens on health care networks worldwide. Although traditional pharmacologic therapies such as insulin and oral antidiabetic stalwarts like metformin and the sulfonylureas continue to be used, newer drugs are now on the market targeting novel blood glucose-lowering pathways. Furthermore, exciting new developments in the understanding of beta cell and islet biology are driving the potential for treatments targeting incretin action, islet transplantation with new methods for immunologic protection, and the generation of functional beta cells from stem cells. Here we discuss the mechanistic details underlying past, present, and future diabetes therapies and evaluate their potential to treat and possibly reverse type 1 and 2 diabetes in humans. SIGNIFICANCE STATEMENT: Diabetes mellitus has reached epidemic proportions in the developed and developing world alike. As the last several years have seen many new developments in the field, a new and up to date review of these advances and their careful evaluation will help both clinical and research diabetologists to better understand where the field is currently heading.
Collapse
Affiliation(s)
- Leslie S Satin
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.) ; ;
| | - Scott A Soleimanpour
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.)
| | - Emily M Walker
- Department of Pharmacology (L.S.S.), Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine (L.S.S., S.A.S., E.M.W.), and Brehm Diabetes Center (L.S.S., S.A.S., E.M.W.), University of Michigan Medical School, Ann Arbor, Michigan; and VA Ann Arbor Healthcare System, Ann Arbor, Michigan (S.A.S.) ; ;
| |
Collapse
|
16
|
Omori K, Nakamura A, Miyoshi H, Yamauchi Y, Kawata S, Takahashi K, Kitao N, Nomoto H, Kameda H, Cho KY, Terauchi Y, Atsumi T. Glucokinase Inactivation Paradoxically Ameliorates Glucose Intolerance by Increasing β-Cell Mass in db/db Mice. Diabetes 2021; 70:917-931. [PMID: 33608422 DOI: 10.2337/db20-0881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/22/2021] [Indexed: 11/13/2022]
Abstract
Efficacy of glucokinase activation on glycemic control is limited to a short-term period. One reason might be related to excess glucose signaling by glucokinase activation toward β-cells. In this study, we investigated the effect of glucokinase haploinsufficiency on glucose tolerance as well as β-cell function and mass using a mouse model of type 2 diabetes. Our results showed that in db/db mice with glucokinase haploinsufficiency, glucose tolerance was ameliorated by augmented insulin secretion associated with the increase in β-cell mass when compared with db/db mice. Gene expression profiling and immunohistochemical and metabolomic analyses revealed that glucokinase haploinsufficiency in the islets of db/db mice was associated with lower expression of stress-related genes, greater expression of transcription factors involved in the maintenance and maturation of β-cell function, less mitochondrial damage, and a superior metabolic pattern. These effects of glucokinase haploinsufficiency could preserve β-cell mass under diabetic conditions. These findings verified our hypothesis that optimizing excess glucose signaling in β-cells by inhibiting glucokinase could prevent β-cell insufficiency, leading to improving glucose tolerance in diabetes status by preserving β-cell mass. Therefore, glucokinase inactivation in β-cells, paradoxically, could be a potential strategy for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Kazuno Omori
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akinobu Nakamura
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hideaki Miyoshi
- Division of Diabetes and Obesity, Faculty of Medicine and Graduate School of Medicine Hokkaido University, Sapporo, Japan
| | - Yuki Yamauchi
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinichiro Kawata
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kiyohiko Takahashi
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Naoyuki Kitao
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroshi Nomoto
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hiraku Kameda
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kyu Yong Cho
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Tatsuya Atsumi
- Department of Rheumatology, Endocrinology, and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|