1
|
Agoncillo ML, Gao Z, De Kraker HC, McHardy SF, Messing RO, Small L, Schmitz-Peiffer C. Effects of a protein kinase C epsilon inhibitor on insulin signalling in lipid-treated HepG2 hepatocytes and glucose tolerance in fat-fed mice. Eur J Pharmacol 2025; 997:177465. [PMID: 40054721 DOI: 10.1016/j.ejphar.2025.177465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 05/02/2025]
Abstract
AIMS Protein kinase C epsilon (PKCε) plays a causative role in the development of glucose intolerance, and is a potential target for the treatment of type 2 diabetes. Here, we examined the effects of the PKCε inhibitor CIDD-0150612 (CP612) on insulin action in palmitate-treated HepG2 hepatocytes in vitro and on glucose homeostasis in fat-fed mice in vivo. METHODS HepG2 cells were treated with palmitate and CP612 and stimulated with insulin. Insulin signalling was examined by immunoblotting and glucose incorporation into glycogen was measured using glucose tracer. Mice were fed a high-fat diet and treated with CP612 prior to glucose tolerance tests and tissue harvest. Proteomic analysis of liver was carried out by mass spectrometry. RESULTS CP612 promoted Akt phosphorylation in a highly insulin-dependent manner and reversed the inhibition of insulin-stimulated Akt phosphorylation and glucose incorporation into glycogen by palmitate. Fat-fed mice treated with CP612 had reduced fat mass, but not lean mass, compared with vehicle-treated littermates. Mice treated acutely with CP612 exhibited elevated fasting blood glucose. However, mice studied 24h after the last dose had lower fasting glucose and improved glucose tolerance with a lower insulin excursion. Proteomic analysis of liver from CP612-treated fat-fed mice indicated a reduction in gluconeogenic gene expression and decreased phosphorylation of the transcription factor Foxk1. CONCLUSIONS The PKCε inhibitor CP612 had beneficial effects on insulin action in hepatocytes and on fat mass and glucose homeostasis in mice. Because certain effects were not previously observed in genetically PKCε-deficient mice, off-target effects may be partly responsible.
Collapse
Affiliation(s)
- Miguel L Agoncillo
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
| | - Zhongmin Gao
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
| | - Harmannus C De Kraker
- Department of Chemistry, Center for Innovative Drug Discovery, University of Texas San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| | - Stanton F McHardy
- Department of Chemistry, Center for Innovative Drug Discovery, University of Texas San Antonio, One UTSA Circle, San Antonio, TX, 78249, USA.
| | - Robert O Messing
- Department of Neuroscience, University of Texas at Austin, E 24th Street, Austin, TX, 78712, USA.
| | - Lewin Small
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia.
| | - Carsten Schmitz-Peiffer
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia; Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia; St Vincent's Clinical School, University of New South Wales, 390 Victoria St, Darlinghurst, NSW, NSW 2010, Australia.
| |
Collapse
|
2
|
Lundsgaard AM, Del Giudice R, Kanta JM, Larance M, Armour SL, London A, Richter MM, Andersen NR, Nicolaisen TS, Carl CS, Sjøberg KA, Bojsen-Møller KN, Knudsen JG, Lagerstedt JO, Fritzen AM, Kiens B. Apolipoprotein A-IV is induced by high-fat diets and mediates positive effects on glucose and lipid metabolism. Mol Metab 2025; 95:102119. [PMID: 40032158 PMCID: PMC11938269 DOI: 10.1016/j.molmet.2025.102119] [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: 12/23/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025] Open
Abstract
OBJECTIVE Low-carbohydrate, high-fat diets under eucaloric conditions are associated with several health-beneficial metabolic effects in humans, particularly in the liver. We recently observed that apolipoprotein A-IV (apoA-IV), a highly abundant apolipoprotein, was among the most upregulated proteins in circulation after six weeks of consuming a high-fat diet in humans. However, the impact of dietary changes in regulating apoA-IV, and the potential effects of apoA-IV on regulation of glucose- and lipid metabolism remain to be fully established. METHODS We investigated the regulation of circulating fasting concentrations of apoA-IV in humans in response to diets enriched in either fat or carbohydrates. Moreover, to study the whole-body and tissue-specific glucose and lipid metabolic effects of apoA-IV, we administrered apoA-IV recombinant protein to mice and isolated pancreatic islets. RESULTS We demonstrate that in healthy human individuals high-fat intake increased fasting plasma apoA-IV concentrations by up to 54%, while high-carbohydrate intake suppressed plasma apoA-IV concentrations. In mice, administration of apoA-IV acutely lowered blood glucose levels both in lean and obese mice. Interestingly, this was related to a dual mechanism, involving both inhibition of hepatic glucose production and increased glucose uptake into white and brown adipose tissues. In addition to an effect on hepatic glucose production, the apoA-IV-induced liver proteome revealed increased capacity for lipoprotein clearance. The effects of apoA-IV in the liver and adipose tissues were concomitant with increased whole-body fatty acid oxidation. Upon glucose stimulation, an improvement in glucose tolerance by apoA-IV administration was related to potentiation of glucose-induced insulin secretion, while apoA-IV inhibited glucagon secretion ex vivo in islets. CONCLUSIONS We find that apoA-IV is potently increased by intake of fat in humans, and that several beneficial metabolic effects, previously associated with high fat intake in humans, are mimicked by administration of apoA-IV protein to mice.
Collapse
Affiliation(s)
- Anne-Marie Lundsgaard
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk A/S, Søborg, Denmark
| | - Rita Del Giudice
- Department of Experimental Medical Science, Lund University, Lund, Sweden; Department of Biomedical Science and Biofilms - Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Josephine M Kanta
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Mark Larance
- Charles Perkins Centre and School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Sarah L Armour
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Amalie London
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Michael M Richter
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Clinical Biochemistry, Copenhagen University Hospital Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Nicoline R Andersen
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine S Nicolaisen
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Christian S Carl
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kim A Sjøberg
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Kirstine Nyvold Bojsen-Møller
- Department of Endocrinology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark; Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jakob G Knudsen
- Section for Cell Biology and Physiology, Department of Biology, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Jens O Lagerstedt
- Department of Experimental Medical Science, Lund University, Lund, Sweden; Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
| | - Andreas M Fritzen
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark; Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- The August Krogh Section for Human & Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
3
|
Hao H, Yuan Y, Ito A, Eberand BM, Tjondro H, Cielesh M, Norris N, Moreno CL, Maxwell JWC, Neely GG, Payne RJ, Kebede MA, Urbauer RJB, Passam FH, Larance M, Haltiwanger RS. FUT10 and FUT11 are protein O-fucosyltransferases that modify protein EMI domains. Nat Chem Biol 2025; 21:598-610. [PMID: 39775168 PMCID: PMC11949838 DOI: 10.1038/s41589-024-01815-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: 02/06/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025]
Abstract
O-Fucosylation plays crucial roles in various essential biological events. Alongside the well-established O-fucosylation of epidermal growth factor-like repeats by protein O-fucosyltransferase 1 (POFUT1) and thrombospondin type 1 repeats by POFUT2, we recently identified a type of O-fucosylation on the elastin microfibril interface (EMI) domain of Multimerin-1 (MMRN1). Here, using AlphaFold2 screens, co-immunoprecipitation, enzymatic assays combined with mass spectrometric analysis and CRISPR-Cas9 knockouts, we demonstrate that FUT10 and FUT11, originally annotated in UniProt as α1,3-fucosyltransferases, are actually POFUTs responsible for modifying EMI domains; thus, we renamed them as POFUT3 and POFUT4, respectively. Like POFUT1/2, POFUT3/4 function in the endoplasmic reticulum, require folded domain structures for modification and participate in a non-canonical endoplasmic reticulum quality control pathway for EMI domain-containing protein secretion. This finding expands the O-fucosylation repertoire and provides an entry point for further exploration in this emerging field of O-fucosylation.
Collapse
Affiliation(s)
- Huilin Hao
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Youxi Yuan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
| | - Atsuko Ito
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA, USA
- Regional Fish Institute, Ltd., Kyoto, Japan
| | - Benjamin M Eberand
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Harry Tjondro
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Michelle Cielesh
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Nicholas Norris
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Cesar L Moreno
- Charles Perkins Centre, School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Joshua W C Maxwell
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - G Gregory Neely
- Charles Perkins Centre, School of Life and Environmental Sciences, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard J Payne
- School of Chemistry, Faculty of Science, The University of Sydney, Sydney, New South Wales, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales, Australia
| | - Melkam A Kebede
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Freda H Passam
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Mark Larance
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| | | |
Collapse
|
4
|
Yau B, An Y, Germanos M, Schwarzkopf P, van der Kraan AG, Larance M, Webster H, Burns C, Asensio CS, Kebede MA. VPS41 deletion triggers progressive loss of insulin stores and downregulation of β-cell identity. Am J Physiol Endocrinol Metab 2025; 328:E457-E469. [PMID: 39716868 DOI: 10.1152/ajpendo.00389.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/25/2024]
Abstract
Vacuolar protein sorting-associated protein 41 (VPS41) has been established as a requirement for normal insulin secretory function in pancreatic β cells. Genetic deletion of VPS41 in mouse pancreatic β cells results in diabetes, although the mechanisms are not understood. Presently, we show that VPS41 deletion results in rapid mature insulin degradation and downregulation of β-cell identity. This phenotype is observed in vivo, with VPS41KO mice displaying progressive loss of insulin content and β-cell function with age. In acute VPS41 depletion in vitro, the loss of insulin is associated with increased degradative pathway activity, increased Adapter Protein 3 complex colocalization with lysosomes, increased nuclear localization of transcription factor E3, and downregulation of PDX1 and INS mRNA expression. Inhibition of lysosomal degradation rescues the rapidly depleted insulin content. These data evidence a VPS41-dependent mechanism for both insulin content degradation and loss of β-cell identity in β cells.NEW & NOTEWORTHY In this study, we show that acute VPS41 deletion results in rapid degradation of insulin, whereas chronic VPS41 deletion results in downregulation of β-cell identity. In acute VPS41 depletion in vitro, the loss of insulin is associated with increased degradative pathway activity, increased Adapter Protein 3 complex colocalization with lysosomes, increased nuclear localization of transcription factor E3, and downregulation of PDX1 and INS mRNA expression. Inhibition of lysosomal degradation rescues the rapidly depleted insulin content.
Collapse
Affiliation(s)
- Belinda Yau
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Yousun An
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Mark Germanos
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Patricia Schwarzkopf
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - A Gabrielle van der Kraan
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Mark Larance
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Hayley Webster
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Christian Burns
- Department of Biological Sciences, University of Denver, Denver, Colorado, United States
| | - Cedric S Asensio
- Department of Biological Sciences, University of Denver, Denver, Colorado, United States
| | - Melkam A Kebede
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| |
Collapse
|
5
|
Matta L, Weber P, Erener S, Walth-Hummel A, Hass D, Bühler LK, Klepac K, Szendroedi J, Guerra J, Rohm M, Sterr M, Lickert H, Bartelt A, Herzig S. Chronic intermittent fasting impairs β cell maturation and function in adolescent mice. Cell Rep 2025; 44:115225. [PMID: 39827461 DOI: 10.1016/j.celrep.2024.115225] [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: 06/19/2024] [Revised: 12/01/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025] Open
Abstract
Intermittent fasting (IF) is a nutritional lifestyle intervention with broad metabolic benefits, but whether the impact of IF depends on the individual's age is unclear. Here, we investigated the effects of IF on systemic metabolism and β cell function in old, middle-aged, and young mice. Short-term IF improves glucose homeostasis across all age groups without altering islet function and morphology. In contrast, while chronic IF is beneficial for adult mice, it results in impaired β cell function in the young. Using single-cell RNA sequencing (scRNA-seq), we delineate that the β cell maturation and function scores are reduced in young mice. In human islets, a similar pattern is observed in type 1 (T1D), but not type 2 (T2D), diabetes, suggesting that the impact of chronic IF in adolescence is linked to the development of β cell dysfunction. Our study suggests considering the duration of IF in younger persons, as it may worsen rather than reduce diabetes outcomes.
Collapse
Affiliation(s)
- Leonardo Matta
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peter Weber
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Suheda Erener
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Alina Walth-Hummel
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg 69120, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Daniela Hass
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lea K Bühler
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg 69120, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Katarina Klepac
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Julia Szendroedi
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg 69120, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Joel Guerra
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg 69120, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany
| | - Michael Sterr
- German Center for Diabetes Research, 85764 Neuherberg, Germany; Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany
| | - Heiko Lickert
- German Center for Diabetes Research, 85764 Neuherberg, Germany; Institute of Diabetes and Regeneration Research, Helmholtz Munich, Neuherberg, Germany; School of Medicine, Technical University of Munich (TUM), Munich, Germany
| | - Alexander Bartelt
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Technische Universität München, Munich, Germany; Chair of Translational Nutritional Medicine, TUM School of Life Sciences, Research Department of Molecular Life Sciences, Technical University of Munich, Freising, Germany; Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Munich, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany; Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg 69120, Germany; German Center for Diabetes Research, 85764 Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Technische Universität München, Munich, Germany; Chair Molecular Metabolic Control, Technical University Munich, Munich 80333, Germany.
| |
Collapse
|
6
|
Germanos M, Yau B, Taper M, Yeoman C, Wilson A, An Y, Cattin-Ortolá J, Masler D, Tong J, Naghiloo S, Needham EJ, van der Kraan AG, Sun K, Loudovaris T, Diaz-Vegas A, Larance M, Thomas H, von Blume J, Thorn P, Ailion M, Asensio C, Kebede MA. Cab45G trafficking through the insulin secretory pathway is altered in human type 2 diabetes. iScience 2025; 28:111719. [PMID: 39898024 PMCID: PMC11787600 DOI: 10.1016/j.isci.2024.111719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/29/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
In type 2 diabetes (T2D), the rate of insulin secretory granule biogenesis can limit insulin secretion from pancreatic β-cells. Using rat insulinoma INS1 β-cells, we show that the soluble Ca2+-binding/trafficking protein, Cab45G, serves as a non-essential chaperone for insulin granule biogenesis. In β-cells, Cab45G is stored within a cis-Golgi reservoir. Cab45G deletion dysregulates Ca2+ homeostasis and leads to secretory abnormality, but insulin granule biogenesis remains intact. Increasing Cab45G biosynthesis leads to anterograde trafficking into insulin granules, stimulating their production. Using human donor islets, we identify increased anterograde Cab45G trafficking in obese humans with and without T2D, consistent with the heightened demand for granule biogenesis. However, humans with T2D demonstrate decreased Golgi Cab45G localization and increased granule Cab45G localization compared to those without T2D. Our study provides the first insight into Cab45G function in specialized secretory cells and opens avenues of investigation into mechanisms associated with β-cell compensation and failure.
Collapse
Affiliation(s)
- Mark Germanos
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Belinda Yau
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Matthew Taper
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Cara Yeoman
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Amy Wilson
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Yousun An
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | | | - Drew Masler
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Jason Tong
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Sheyda Naghiloo
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Elise J Needham
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - A Gabrielle van der Kraan
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Kitty Sun
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Thomas Loudovaris
- Immunology and Diabetes Unit, St Vincent’s Institute, Department of Medicine, St Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Alexis Diaz-Vegas
- School of Life and Environmental Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Mark Larance
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Helen Thomas
- Immunology and Diabetes Unit, St Vincent’s Institute, Department of Medicine, St Vincent’s Hospital, University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Julia von Blume
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Peter Thorn
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Michael Ailion
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cedric Asensio
- Department of Biological Sciences, University of Denver, Denver, CO 80210, USA
| | - Melkam Alamerew Kebede
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
7
|
Aumailley L, Lebel M. Sex and organ specific proteomic responses to vitamin C deficiency in the brain, heart, liver, and spleen of Gulo-/- mice. PLoS One 2024; 19:e0311857. [PMID: 39388511 PMCID: PMC11476689 DOI: 10.1371/journal.pone.0311857] [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: 06/21/2024] [Accepted: 09/25/2024] [Indexed: 10/12/2024] Open
Abstract
Recent advances in mass spectrometry have indicated that the water-soluble antioxidant vitamin C differentially modulates the abundance of various proteins in the hepatic tissue of female and male mice. In this study, we performed LC-MS/MS to identify and quantify proteins that correlate with serum vitamin C concentrations in the whole brain, heart, liver, and spleen tissues in mice deficient for the enzyme L-Gulonolactone oxidase required for vitamin C synthesis in mammals. This work shows for the first time that various biological processes affected by a vitamin C deficiency are not only sex specific dependent but also tissue specific dependent even though many proteins have been identified and quantified in more than three organs. For example, the abundance of several complex III subunits of the mitochondrial electron transport chain correlated positively with the levels of serum vitamin C only in the liver and not in the other tissues examined in this study even though such proteins were identified in all the organs analyzed. Western blot analyses on the Uqcrc1 and Uqcrfs1 complex III subunits validated the mass spectrometry results. Interestingly, the ferritin subunits represented the few quantified protein complexes that correlated positively with serum vitamin C in all the organs examined. Concomitantly, serum ferritin light chain 1 was inversely correlated with vitamin C levels in the serum. Thus, our study provides an initial comprehensive atlas of proteins significantly correlating with vitamin C in four organs in mice that will be a useful resource to the scientific community.
Collapse
Affiliation(s)
- Lucie Aumailley
- Centre de Recherche du CHU de Québec, Faculty of Medicine, Université Laval, Québec City Québec, Canada
| | - Michel Lebel
- Centre de Recherche du CHU de Québec, Faculty of Medicine, Université Laval, Québec City Québec, Canada
| |
Collapse
|
8
|
Marko DM, Conn MO, Schertzer JD. Intermittent fasting influences immunity and metabolism. Trends Endocrinol Metab 2024; 35:821-833. [PMID: 38719726 DOI: 10.1016/j.tem.2024.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 09/12/2024]
Abstract
Intermittent fasting (IF) modifies cell- and tissue-specific immunometabolic responses that dictate metabolic flexibility and inflammation during obesity and type 2 diabetes (T2D). Fasting forces periods of metabolic flexibility and necessitates increased use of different substrates. IF can lower metabolic inflammation and improve glucose metabolism without lowering obesity and can influence time-dependent, compartmentalized changes in immunity. Liver, adipose tissue, skeletal muscle, and immune cells communicate to relay metabolic and immune signals during fasting. Here we review the connections between metabolic and immune cells to explain the divergent effects of IF compared with classic caloric restriction (CR) strategies. We also explore how the immunometabolism of metabolic diseases dictates certain IF outcomes, where the gut microbiota triggers changes in immunity and metabolism during fasting.
Collapse
Affiliation(s)
- Daniel M Marko
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Meghan O Conn
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada; Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Centre for Metabolism, Obesity, and Diabetes Research, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
9
|
Diaz-Vegas A, Cooke KC, Cutler HB, Yau B, Masson SWC, Harney D, Fuller OK, Potter M, Madsen S, Craw NR, Zhang Y, Moreno CL, Kebede MA, Neely GG, Stöckli J, Burchfield JG, James DE. Deletion of miPEP in adipocytes protects against obesity and insulin resistance by boosting muscle metabolism. Mol Metab 2024; 86:101983. [PMID: 38960128 PMCID: PMC11292358 DOI: 10.1016/j.molmet.2024.101983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/05/2024] Open
Abstract
Mitochondria facilitate thousands of biochemical reactions, covering a broad spectrum of anabolic and catabolic processes. Here we demonstrate that the adipocyte mitochondrial proteome is markedly altered across multiple models of insulin resistance and reveal a consistent decrease in the level of the mitochondrial processing peptidase miPEP. OBJECTIVE To determine the role of miPEP in insulin resistance. METHODS To experimentally test this observation, we generated adipocyte-specific miPEP knockout mice to interrogate its role in the aetiology of insulin resistance. RESULTS We observed a strong phenotype characterised by enhanced insulin sensitivity and reduced adiposity, despite normal food intake and physical activity. Strikingly, these phenotypes vanished when mice were housed at thermoneutrality, suggesting that metabolic protection conferred by miPEP deletion hinges upon a thermoregulatory process. Tissue specific analysis of miPEP deficient mice revealed an increment in muscle metabolism, and upregulation of the protein FBP2 that is involved in ATP hydrolysis in the gluconeogenic pathway. CONCLUSION These findings suggest that miPEP deletion initiates a compensatory increase in skeletal muscle metabolism acting as a protective mechanism against diet-induced obesity and insulin resistance.
Collapse
Affiliation(s)
- Alexis Diaz-Vegas
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia.
| | - Kristen C Cooke
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Harry B Cutler
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Belinda Yau
- School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - Stewart W C Masson
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Dylan Harney
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Oliver K Fuller
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Meg Potter
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Søren Madsen
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Niamh R Craw
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Yiju Zhang
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Cesar L Moreno
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Melkam A Kebede
- School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia
| | - G Gregory Neely
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Jacqueline Stöckli
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - James G Burchfield
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - David E James
- School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia; Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia; School of Medical Sciences, University of Sydney, Camperdown, New South Wales, Australia.
| |
Collapse
|