1
|
Greig J, Bates GT, Yin DI, Briant K, Simonetti B, Cullen PJ, Brodsky FM. CHC22 clathrin recruitment to the early secretory pathway requires two-site interaction with SNX5 and p115. EMBO J 2024; 43:4298-4323. [PMID: 39160272 PMCID: PMC11445476 DOI: 10.1038/s44318-024-00198-y] [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: 10/04/2023] [Revised: 07/15/2024] [Accepted: 07/17/2024] [Indexed: 08/21/2024] Open
Abstract
The two clathrin isoforms, CHC17 and CHC22, mediate separate intracellular transport routes. CHC17 performs endocytosis and housekeeping membrane traffic in all cells. CHC22, expressed most highly in skeletal muscle, shuttles the glucose transporter GLUT4 from the ERGIC (endoplasmic-reticulum-to-Golgi intermediate compartment) directly to an intracellular GLUT4 storage compartment (GSC), from where GLUT4 can be mobilized to the plasma membrane by insulin. Here, molecular determinants distinguishing CHC22 from CHC17 trafficking are defined. We show that the C-terminal trimerization domain of CHC22 interacts with SNX5, which also binds the ERGIC tether p115. SNX5, and the functionally redundant SNX6, are required for CHC22 localization independently of their participation in the endosomal ESCPE-1 complex. In tandem, an isoform-specific patch in the CHC22 N-terminal domain separately mediates binding to p115. This dual mode of clathrin recruitment, involving interactions at both N- and C-termini of the heavy chain, is required for CHC22 targeting to ERGIC membranes to mediate the Golgi-bypass route for GLUT4 trafficking. Interference with either interaction inhibits GLUT4 targeting to the GSC, defining a bipartite mechanism regulating a key pathway in human glucose metabolism.
Collapse
Affiliation(s)
- Joshua Greig
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - George T Bates
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Daowen I Yin
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Kit Briant
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK
| | - Boris Simonetti
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Peter J Cullen
- School of Biochemistry, Faculty of Life Sciences, University of Bristol, Bristol, UK
| | - Frances M Brodsky
- Structural and Molecular Biology, Division of Biosciences, University College London, London, WC1E 6BT, UK.
- Institute of Structural and Molecular Biology, Birkbeck and University College London, London, WC1E 7HX, UK.
| |
Collapse
|
2
|
Kondo M, Sawada K, Matsuda Y, Abe M, Sanechika N, Takanashi Y, Mori Y, Kimura M, Toyoda M. Study of the Effects of Deuterium-Depleted Water on the Expression of GLUT4 and Insulin Resistance in the Muscle Cell Line C2C12. Biomedicines 2024; 12:1771. [PMID: 39200235 PMCID: PMC11351524 DOI: 10.3390/biomedicines12081771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 09/02/2024] Open
Abstract
Deuterium-depleted water (DDW) is used in the treatment of many diseases, including cancer and diabetes. To detect the effect of DDW on gene expression and activation of the insulin-responsive transporter GLUT4 as a mechanism for improving the pathology of diabetes, we investigated the GLUT4 expression and glucose uptake at various concentrations of DDW using the myoblast cell line C2C12 differentiated into myotubes. GLUT4 gene expression significantly increased under deuterium depletion, reaching a maximum value at a deuterium concentration of approximately 50 ppm, which was approximately nine times that of natural water with a deuterium concentration of 150 ppm. GLUT4 protein also showed an increase at similar DDW concentrations. The membrane translocation of GLUT4 by insulin stimulation reached a maximum value at a deuterium concentration of approximately 50-75 ppm, which was approximately 2.2 times that in natural water. Accordingly, glucose uptake also increased by up to 2.2 times at a deuterium concentration of approximately 50 ppm. Drug-induced insulin resistance was attenuated, and the glucose uptake was four times higher in the presence of 10 ng/mL TNF-α and three times higher in the presence of 1 μg/mL resistin at a deuterium concentration of approximately 50 ppm relative to natural water. These results suggest that DDW promotes GLUT4 expression and insulin-stimulated activation in muscle cells and reduces insulin resistance, making it an effective treatment for diabetes.
Collapse
Affiliation(s)
- Masumi Kondo
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University Hachioji Hospital, 1838 Ishikawacho, Hachioji 192-0032, Japan; (M.K.); (M.A.)
| | - Kaichiro Sawada
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yosuke Matsuda
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Makiko Abe
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University Hachioji Hospital, 1838 Ishikawacho, Hachioji 192-0032, Japan; (M.K.); (M.A.)
| | - Noriyuki Sanechika
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yumi Takanashi
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Yoshitaka Mori
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Moritsugu Kimura
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| | - Masao Toyoda
- Division of Nephrology, Endocrinology and Metabolism, Department of Medicine, Tokai University School of Medicine, 143 Shimokasuya, Isehara 259-1193, Japan; (K.S.); (Y.M.); (N.S.); (Y.T.); (Y.M.); (M.K.)
| |
Collapse
|
3
|
Zhao Q, Zheng Y, Li Y, Shi L, Zhang J, Ma D, You M. An Orphan Gene Enhances Male Reproductive Success in Plutella xylostella. Mol Biol Evol 2024; 41:msae142. [PMID: 38990889 PMCID: PMC11290247 DOI: 10.1093/molbev/msae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/28/2024] [Accepted: 07/05/2024] [Indexed: 07/13/2024] Open
Abstract
Plutella xylostella exhibits exceptional reproduction ability, yet the genetic basis underlying the high reproductive capacity remains unknown. Here, we demonstrate that an orphan gene, lushu, which encodes a sperm protein, plays a crucial role in male reproductive success. Lushu is located on the Z chromosome and is prevalent across different P. xylostella populations worldwide. We subsequently generated lushu mutants using transgenic CRISPR/Cas9 system. Knockout of Lushu results in reduced male mating efficiency and accelerated death in adult males. Furthermore, our findings highlight that the deficiency of lushu reduced the transfer of sperms from males to females, potentially resulting in hindered sperm competition. Additionally, the knockout of Lushu results in disrupted gene expression in energy-related pathways and elevated insulin levels in adult males. Our findings reveal that male reproductive performance has evolved through the birth of a newly evolved, lineage-specific gene with enormous potentiality in fecundity success. These insights hold valuable implications for identifying the target for genetic control, particularly in relation to species-specific traits that are pivotal in determining high levels of fecundity.
Collapse
Affiliation(s)
- Qian Zhao
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
| | - Yahong Zheng
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yiying Li
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Lingping Shi
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Jing Zhang
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
| | - Dongna Ma
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Minsheng You
- State Key Laboratory for Ecological Pest Control of Fujian/Taiwan Crops and College of Life Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Ministerial and Provincial Joint Innovation Centre for Safety Production of Cross-Strait Crops, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Joint International Research Laboratory of Ecological Pest Control, Ministry of Education, Fuzhou 350002, China
| |
Collapse
|
4
|
Greeny A, Nair A, Sadanandan P, Satarker S, Famurewa AC, Nampoothiri M. Epigenetic Alterations in Alzheimer's Disease: Impact on Insulin Signaling and Advanced Drug Delivery Systems. BIOLOGY 2024; 13:157. [PMID: 38534427 DOI: 10.3390/biology13030157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/25/2024] [Accepted: 02/26/2024] [Indexed: 03/28/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that predominantly affects the hippocampus and the entorhinal complex, leading to memory lapse and cognitive impairment. This can have a negative impact on an individual's behavior, speech, and ability to navigate their surroundings. AD is one of the principal causes of dementia. One of the most accepted theories in AD, the amyloid β (Aβ) hypothesis, assumes that the buildup of the peptide Aβ is the root cause of AD. Impaired insulin signaling in the periphery and central nervous system has been considered to have an effect on the pathophysiology of AD. Further, researchers have shifted their focus to epigenetic mechanisms that are responsible for dysregulating major biochemical pathways and intracellular signaling processes responsible for directly or indirectly causing AD. The prime epigenetic mechanisms encompass DNA methylation, histone modifications, and non-coding RNA, and are majorly responsible for impairing insulin signaling both centrally and peripherally, thus leading to AD. In this review, we provide insights into the major epigenetic mechanisms involved in causing AD, such as DNA methylation and histone deacetylation. We decipher how the mechanisms alter peripheral insulin signaling and brain insulin signaling, leading to AD pathophysiology. In addition, this review also discusses the need for newer drug delivery systems for the targeted delivery of epigenetic drugs and explores targeted drug delivery systems such as nanoparticles, vesicular systems, networks, and other nano formulations in AD. Further, this review also sheds light on the future approaches used for epigenetic drug delivery.
Collapse
Affiliation(s)
- Alosh Greeny
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ayushi Nair
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Health Science Campus, Kochi 682041, India
| | - Sairaj Satarker
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University, Ndufu-Alike, Ikwo 482123, Nigeria
| | - Madhavan Nampoothiri
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| |
Collapse
|
5
|
Khandayataray P, Samal D, Murthy MK. Arsenic and adipose tissue: an unexplored pathway for toxicity and metabolic dysfunction. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:8291-8311. [PMID: 38165541 DOI: 10.1007/s11356-023-31683-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
Arsenic-contaminated drinking water can induce various disorders by disrupting lipid and glucose metabolism in adipose tissue, leading to insulin resistance. It inhibits adipocyte development and exacerbates insulin resistance, though the precise impact on lipid synthesis and lipolysis remains unclear. This review aims to explore the processes and pathways involved in adipogenesis and lipolysis within adipose tissue concerning arsenic-induced diabetes. Although arsenic exposure is linked to type 2 diabetes, the specific role of adipose tissue in its pathogenesis remains uncertain. The review delves into arsenic's effects on adipose tissue and related signaling pathways, such as SIRT3-FOXO3a, Ras-MAP-AP-1, PI(3)-K-Akt, endoplasmic reticulum stress proteins, CHOP10, and GPCR pathways, emphasizing the role of adipokines. This analysis relies on existing literature, striving to offer a comprehensive understanding of different adipokine categories contributing to arsenic-induced diabetes. The findings reveal that arsenic detrimentally impacts white adipose tissue (WAT) by reducing adipogenesis and promoting lipolysis. Epidemiological studies have hinted at a potential link between arsenic exposure and obesity development, with limited research suggesting a connection to lipodystrophy. Further investigations are needed to elucidate the mechanistic association between arsenic exposure and impaired adipose tissue function, ultimately leading to insulin resistance.
Collapse
Affiliation(s)
- Pratima Khandayataray
- Department of Biotechnology, Academy of Management and Information Technology, Utkal University, Bhubaneswar, Odisha, 752057, India
| | - Dibyaranjan Samal
- Department of Biotechnology, Sri Satya Sai University of Technical and Medical Sciences, Sehore, Madhya Pradesh, 466001, India
| | - Meesala Krishna Murthy
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Punjab, 140401, India.
| |
Collapse
|
6
|
Zaarur N, Meriin AB, Singh M, Goel RK, Zaia J, Kandror KV. Akt may associate with insulin-responsive vesicles via interaction with sortilin. FEBS Lett 2024; 598:390-399. [PMID: 38105115 PMCID: PMC10922807 DOI: 10.1002/1873-3468.14790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/04/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
Insulin-responsive vesicles (IRVs) deliver the glucose transporter Glut4 to the plasma membrane in response to activation of the insulin signaling cascade: insulin receptor-IRS-PI3 kinase-Akt-TBC1D4-Rab10. Previous studies have shown that Akt, TBC1D4, and Rab10 are compartmentalized on the IRVs. Although functionally significant, the mechanism of Akt association with the IRVs remains unknown. Using pull-down assays, immunofluorescence microscopy, and cross-linking, we have found that Akt may be recruited to the IRVs via the interaction with the juxtamembrane domain of the cytoplasmic C terminus of sortilin, a major IRV protein. Overexpression of full-length sortilin increases insulin-stimulated phosphorylation of TBC1D4 and glucose uptake in adipocytes, while overexpression of the cytoplasmic tail of sortilin has the opposite effect. Our findings demonstrate that the IRVs represent both a scaffold and a target of insulin signaling.
Collapse
Affiliation(s)
- Nava Zaarur
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| | - Anatoli B. Meriin
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| | - Maneet Singh
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| | - Raghuveera K. Goel
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
- Center for Network Systems Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| | - Joseph Zaia
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
- Center for Network Systems Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| | - Konstantin V. Kandror
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, MA 02118
| |
Collapse
|
7
|
Geiser A, Foylan S, Tinning PW, Bryant NJ, Gould GW. GLUT4 dispersal at the plasma membrane of adipocytes: a super-resolved journey. Biosci Rep 2023; 43:BSR20230946. [PMID: 37791639 PMCID: PMC10600063 DOI: 10.1042/bsr20230946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/05/2023] Open
Abstract
In adipose tissue, insulin stimulates glucose uptake by mediating the translocation of GLUT4 from intracellular vesicles to the plasma membrane. In 2010, insulin was revealed to also have a fundamental impact on the spatial distribution of GLUT4 within the plasma membrane, with the existence of two GLUT4 populations at the plasma membrane being defined: (1) as stationary clusters and (2) as diffusible monomers. In this model, in the absence of insulin, plasma membrane-fused GLUT4 are found to behave as clusters. These clusters are thought to arise from exocytic events that retain GLUT4 at their fusion sites; this has been proposed to function as an intermediate hub between GLUT4 exocytosis and re-internalisation. By contrast, insulin stimulation induces the dispersal of GLUT4 clusters into monomers and favours a distinct type of GLUT4-vesicle fusion event, known as fusion-with-release exocytosis. Here, we review how super-resolution microscopy approaches have allowed investigation of the characteristics of plasma membrane-fused GLUT4 and further discuss regulatory step(s) involved in the GLUT4 dispersal machinery, introducing the scaffold protein EFR3 which facilitates localisation of phosphatidylinositol 4-kinase type IIIα (PI4KIIIα) to the cell surface. We consider how dispersal may be linked to the control of transporter activity, consider whether macro-organisation may be a widely used phenomenon to control proteins within the plasma membrane, and speculate on the origin of different forms of GLUT4-vesicle exocytosis.
Collapse
Affiliation(s)
- Angéline Geiser
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Shannan Foylan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Peter W Tinning
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| | - Nia J Bryant
- Department of Biology, University of York, Heslington, York, U.K
| | - Gwyn W Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, U.K
| |
Collapse
|
8
|
Tatti P, Singh P. Insulin Resistance: An Unresolved Riddle. J Clin Med 2023; 12:6394. [PMID: 37835038 PMCID: PMC10573251 DOI: 10.3390/jcm12196394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 10/15/2023] Open
Abstract
Insulin resistance (IR) is a rather common condition that is often diagnosed on the basis of an arbitrary "increased insulin value" or the presence of symptoms indicative of the Metabolic Syndrome [...].
Collapse
|
9
|
Chen H, Weinberg ZY, Kumar GA, Puthenveedu MA. Vesicle-associated membrane protein 2 is a cargo-selective v-SNARE for a subset of GPCRs. J Cell Biol 2023; 222:e202207070. [PMID: 37022307 PMCID: PMC10082327 DOI: 10.1083/jcb.202207070] [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: 07/15/2022] [Revised: 01/26/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023] Open
Abstract
Vesicle fusion at the plasma membrane is critical for releasing hormones and neurotransmitters and for delivering the cognate G protein-coupled receptors (GPCRs) to the cell surface. The SNARE fusion machinery that releases neurotransmitters has been well characterized. In contrast, the fusion machinery that delivers GPCRs is still unknown. Here, using high-speed multichannel imaging to simultaneously visualize receptors and v-SNAREs in real time in individual fusion events, we identify VAMP2 as a selective v-SNARE for GPCR delivery. VAMP2 was preferentially enriched in vesicles that mediate the surface delivery of μ opioid receptor (MOR), but not other cargos, and was required selectively for MOR recycling. Interestingly, VAMP2 did not show preferential localization on MOR-containing endosomes, suggesting that v-SNAREs are copackaged with specific cargo into separate vesicles from the same endosomes. Together, our results identify VAMP2 as a cargo-selective v-SNARE and suggest that surface delivery of specific GPCRs is mediated by distinct fusion events driven by distinct SNARE complexes.
Collapse
Affiliation(s)
- Hao Chen
- Department of Pharmacology, University of MichiganMedical School, Ann Arbor, MI, USA
| | - Zara Y. Weinberg
- Department of Pharmacology, University of MichiganMedical School, Ann Arbor, MI, USA
| | - G. Aditya Kumar
- Department of Pharmacology, University of MichiganMedical School, Ann Arbor, MI, USA
| | | |
Collapse
|
10
|
Park JS, Perl A. Endosome Traffic Modulates Pro-Inflammatory Signal Transduction in CD4 + T Cells-Implications for the Pathogenesis of Systemic Lupus Erythematosus. Int J Mol Sci 2023; 24:10749. [PMID: 37445926 DOI: 10.3390/ijms241310749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/10/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Endocytic recycling regulates the cell surface receptor composition of the plasma membrane. The surface expression levels of the T cell receptor (TCR), in concert with signal transducing co-receptors, regulate T cell responses, such as proliferation, differentiation, and cytokine production. Altered TCR expression contributes to pro-inflammatory skewing, which is a hallmark of autoimmune diseases, such as systemic lupus erythematosus (SLE), defined by a reduced function of regulatory T cells (Tregs) and the expansion of CD4+ helper T (Th) cells. The ensuing secretion of inflammatory cytokines, such as interferon-γ and interleukin (IL)-4, IL-17, IL-21, and IL-23, trigger autoantibody production and tissue infiltration by cells of the adaptive and innate immune system that induce organ damage. Endocytic recycling influences immunological synapse formation by CD4+ T lymphocytes, signal transduction from crosslinked surface receptors through recruitment of adaptor molecules, intracellular traffic of organelles, and the generation of metabolites to support growth, cytokine production, and epigenetic control of DNA replication and gene expression in the cell nucleus. This review will delineate checkpoints of endosome traffic that can be targeted for therapeutic interventions in autoimmune and other disease conditions.
Collapse
Affiliation(s)
- Joy S Park
- Department of Medicine, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Andras Perl
- Department of Medicine, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Biochemistry and Molecular Biology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
- Department of Microbiology and Immunology, Norton College of Medicine, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| |
Collapse
|
11
|
Pánico P, Velasco M, Salazar AM, Ostrosky-Wegman P, Hiriart M. The effects of sucrose and arsenic on muscular insulin signaling pathways differ between the gastrocnemius and quadriceps muscles. Front Endocrinol (Lausanne) 2023; 14:1165415. [PMID: 37229459 PMCID: PMC10205014 DOI: 10.3389/fendo.2023.1165415] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Introduction Insulin resistance in muscle can originate from a sedentary lifestyle, hypercaloric diets, or exposure to endocrine-disrupting pollutants such as arsenic. In skeletal muscle, insulin stimulates glucose uptake by translocating GLUT4 to the sarcolemma. This study aimed to evaluate the alterations induced by sucrose and arsenic exposure in vivo on the pathways involved in insulinstimulated GLUT4 translocation in the quadriceps and gastrocnemius muscles. Methods Male Wistar rats were treated with 20% sucrose (S), 50 ppm sodium arsenite (A), or both (A+S) in drinking water for 8 weeks. We conducted an intraperitoneal insulin tolerance (ITT) test on the seventh week of treatment. The quadriceps and gastrocnemius muscles were obtained after overnight fasting or 30 min after intraperitoneal insulin injection. We assessed changes in GLUT4 translocation to the sarcolemma by cell fractionation and abundance of the proteins involved in GLUT4 translocation by Western blot. Results Male rats consuming S and A+S gained more weight than control and Atreated animals. Rats consuming S, A, and A+S developed insulin resistance assessed through ITT. Neither treatments nor insulin stimulation in the quadriceps produced changes in GLUT4 levels in the sarcolemma and Akt phosphorylation. Conversely, A and A+S decreased protein expression of Tether containing UBX domain for GLUT4 (TUG), and A alone increased calpain-10 expression. All treatments reduced this muscle's protein levels of VAMP2. Conversely, S and A treatment increased basal GLUT4 levels in the sarcolemma of the gastrocnemius, while all treatments inhibited insulin-induced GLUT4 translocation. These effects correlated with lower basal levels of TUG and impaired insulin-stimulated TUG proteolysis. Moreover, animals treated with S had reduced calpain-10 protein levels in this muscle, while A and A+S inhibited insulin-induced Akt phosphorylation. Conclusion Arsenic and sucrose induce systemic insulin resistance due to defects in GLUT4 translocation induced by insulin. These defects depend on which muscle is being analyzed, in the quadriceps there were defects in GLUT4 retention and docking while in the gastrocnemius the Akt pathway was impacted by arsenic and the proteolytic pathway was impaired by arsenic and sucrose.
Collapse
Affiliation(s)
- Pablo Pánico
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Myrian Velasco
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ana María Salazar
- Department of Genomic Medicine and Environmental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Patricia Ostrosky-Wegman
- Department of Genomic Medicine and Environmental Toxicology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcia Hiriart
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
12
|
Sun Z, Li Y, Tan X, Liu W, He X, Pan D, Li E, Xu L, Long L. Friend or Foe: Regulation, Downstream Effectors of RRAD in Cancer. Biomolecules 2023; 13:biom13030477. [PMID: 36979412 PMCID: PMC10046484 DOI: 10.3390/biom13030477] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
Ras-related associated with diabetes (RRAD), a member of the Ras-related GTPase superfamily, is primarily a cytosolic protein that actives in the plasma membrane. RRAD is highly expressed in type 2 diabetes patients and as a biomarker of congestive heart failure. Mounting evidence showed that RRAD is important for the progression and metastasis of tumor cells, which play opposite roles as an oncogene or tumor suppressor gene depending on cancer and cell type. These findings are of great significance, especially given that relevant molecular mechanisms are being discovered. Being regulated in various pathways, RRAD plays wide spectrum cellular activity including tumor cell division, motility, apoptosis, and energy metabolism by modulating tumor-related gene expression and interacting with multiple downstream effectors. Additionally, RRAD in senescence may contribute to its role in cancer. Despite the twofold characters of RRAD, targeted therapies are becoming a potential therapeutic strategy to combat cancers. This review will discuss the dual identity of RRAD in specific cancer type, provides an overview of the regulation and downstream effectors of RRAD to offer valuable insights for readers, explore the intracellular role of RRAD in cancer, and give a reference for future mechanistic studies.
Collapse
Affiliation(s)
- Zhangyue Sun
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Yongkang Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xiaolu Tan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Wanyi Liu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Xinglin He
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
| | - Deyuan Pan
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Enmin Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Liyan Xu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
- Cancer Research Center, Institute of Basic Medical Science, Shantou University Medical College, Shantou 515041, China
- Guangdong Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou 515041, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
- Correspondence: ; Tel.: +86-754-88900460; Fax: +86-754-88900847
| |
Collapse
|
13
|
Weimershaus M, Carvalho C, Rignault R, Waeckel-Enee E, Dussiot M, van Endert P, Maciel TT, Hermine O. Mast cell-mediated inflammation relies on insulin-regulated aminopeptidase controlling cytokine export from the Golgi. J Allergy Clin Immunol 2023:S0091-6749(23)00090-8. [PMID: 36708814 DOI: 10.1016/j.jaci.2023.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 12/31/2022] [Accepted: 01/11/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND On activation, mast cells rapidly release preformed inflammatory mediators from large cytoplasmic granules via regulated exocytosis. This acute degranulation is followed by a late activation phase involving synthesis and secretion of cytokines, growth factors, and other inflammatory molecules via the constitutive pathway that remains ill defined. OBJECTIVE We investigated the role for an insulin-responsive vesicle-like endosomal compartment, marked by insulin-regulated aminopeptidase (IRAP), in the secretion of TNF-α and IL-6 in mast cells and macrophages. METHODS Murine knockout (KO) mouse models (IRAP-KO and kit-Wsh/sh) were used to study inflammatory disease models and to measure and mechanistically investigate cytokine secretion and degranulation in bone marrow-derived mast cells in vitro. RESULTS IRAP-KO mice are protected from TNF-α-dependent kidney injury and inflammatory arthritis. In the absence of IRAP, TNF-α and IL-6 but not IL-10 fail to be efficiently secreted. Moreover, chemical targeting of IRAP endosomes reduced proinflammatory cytokine secretion. Mechanistically, impaired TNF-α export from the Golgi and reduced colocalization of vesicle-associated membrane protein (VAMP) 3-positive TNF-α transport vesicles with syntaxin 4 (aka Stx4) was observed in IRAP-KO mast cells, while VAMP8-dependent exocytosis of secretory granules was facilitated. CONCLUSION IRAP plays a novel role in mast cell-mediated inflammation through the regulation of exocytic trafficking of cytokines.
Collapse
Affiliation(s)
- Mirjana Weimershaus
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France.
| | - Caroline Carvalho
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France
| | - Rachel Rignault
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France; Université de Paris Cité, Paris, France
| | | | - Michael Dussiot
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France; Université de Paris Cité, Paris, France; Laboratory of Excellence GR-Ex, Paris, France
| | - Peter van Endert
- INSERM UMR 1151, CNRS UMR 8253, Paris, France; Université de Paris Cité, Paris, France
| | - Thiago Trovati Maciel
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France; Laboratory of Excellence GR-Ex, Paris, France
| | - Olivier Hermine
- Imagine Institute, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM U1163, F-75015, Paris, France; Université de Paris Cité, Paris, France; Hôpital Necker Enfants Malades, Paris, France; Laboratory of Excellence GR-Ex, Paris, France
| |
Collapse
|
14
|
Abstract
The global prevalences of obesity and type 2 diabetes mellitus have reached epidemic status, presenting a heavy burden on society. It is therefore essential to find novel mechanisms and targets that could be utilized in potential treatment strategies and, as such, intracellular membrane trafficking has re-emerged as a regulatory tool for controlling metabolic homeostasis. Membrane trafficking is an essential physiological process that is responsible for the sorting and distribution of signalling receptors, membrane transporters and hormones or other ligands between different intracellular compartments and the plasma membrane. Dysregulation of intracellular transport is associated with many human diseases, including cancer, neurodegeneration, immune deficiencies and metabolic diseases, such as type 2 diabetes mellitus and its associated complications. This Review focuses on the latest advances on the role of endosomal membrane trafficking in metabolic physiology and pathology in vivo, highlighting the importance of this research field in targeting metabolic diseases.
Collapse
Affiliation(s)
- Jerome Gilleron
- Université Côte d'Azur, Institut National de la Santé et de la Recherche Médicale (Inserm), UMR1065 C3M, Team Cellular and Molecular Pathophysiology of Obesity, Nice, France.
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
15
|
Bremner SK, Al Shammari WS, Milligan RS, Hudson BD, Sutherland C, Bryant NJ, Gould GW. Pleiotropic effects of Syntaxin16 identified by gene editing in cultured adipocytes. Front Cell Dev Biol 2022; 10:1033501. [PMID: 36467416 PMCID: PMC9716095 DOI: 10.3389/fcell.2022.1033501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 11/02/2022] [Indexed: 08/01/2023] Open
Abstract
Adipocytes play multiple roles in the regulation of glucose metabolism which rely on the regulation of membrane traffic. These include secretion of adipokines and serving as an energy store. Central to their energy storing function is the ability to increase glucose uptake in response to insulin, mediated through translocation of the facilitative glucose transporter GLUT4 to the cell surface. The trans-Golgi reticulum localized SNARE protein syntaxin 16 (Sx16) has been identified as a key component of the secretory pathway required for insulin-regulated trafficking of GLUT4. We used CRISPR/Cas9 technology to generate 3T3-L1 adipocytes lacking Sx16 to understand the role of the secretory pathway on adipocyte function. GLUT4 mRNA and protein levels were reduced in Sx16 knockout adipocytes and insulin stimulated GLUT4 translocation to the cell surface was reduced. Strikingly, neither basal nor insulin-stimulated glucose transport were affected. By contrast, GLUT1 levels were upregulated in Sx16 knockout cells. Levels of sortilin and insulin regulated aminopeptidase were also increased in Sx16 knockout adipocytes which may indicate an upregulation of an alternative GLUT4 sorting pathway as a compensatory mechanism for the loss of Sx16. In response to chronic insulin stimulation, Sx16 knockout adipocytes exhibit elevated insulin-independent glucose transport and significant alterations in lactate metabolism. We further show that the adipokine secretory pathways are impaired in Sx16 knockout cells. Together this demonstrates a role for Sx16 in the control of glucose transport, the response to elevated insulin, cellular metabolic profiles and adipocytokine secretion.
Collapse
Affiliation(s)
- Shaun K. Bremner
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Woroud S. Al Shammari
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Roderick S. Milligan
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | - Brian D. Hudson
- The Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, United Kingdom
| | - Calum Sutherland
- Department of Cellular Medicine, Ninewells Hospital, University of Dundee, Glasgow, United Kingdom
| | - Nia J. Bryant
- Department of Biology, University of York, York, United Kingdom
| | - Gwyn W. Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
16
|
Cai K, Deng L, Zheng D, Li L, He Z, Yu C. MICAL1 facilitates pancreatic cancer proliferation, migration, and invasion by activating WNT/β-catenin pathway. J Transl Med 2022; 20:528. [DOI: 10.1186/s12967-022-03749-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/02/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Background
MICAL1 is involved in the malignant processes of several types of cancer; however, the role of MICAL1 in pancreatic cancer (PC) has not been well-characterized. This study aimed to investigate the expression and function of MICAL1 in PC.
Methods
RT-qPCR and immunohistochemistry were used to detect MICAL1 expression in PC and adjacent nontumor tissues. Cell Counting Kit-8, EdU, clone formation, wound healing, and Transwell assays as well as animal models were used to investigate the effects of overexpression or inhibition of MICAL1 expression on the proliferation, invasion, and metastasis of PC cells. RNA-seq was used to explore the main pathway underlying the functions of MICAL1. Proteomics, mass spectrometry, and co-immunoprecipitation assays were used to investigate the interaction of proteins with MICAL1. Rescue experiments were conducted to validate these findings.
Results
Both MICAL1 mRNA and protein levels were upregulated in PC tissues compared with matched adjacent nontumor tissues. The expression level of MICAL1 was associated with the proliferative and metastatic status of PC. Repression of MICAL1 significantly inhibited PC cell growth, migration, and invasion in vitro and in vivo. RNA sequencing analysis indicated that MICAL1 was closely correlated with the WNT pathway. Overexpression of MICAL1 (1) promoted the phosphorylation of TBC1D1 at the Ser660 site, (2) facilitated the distribution of FZD7 on the cytomembrane, (3) inhibited the degradation of FZD7 in the lysosome, and (4) activated the WNT pathway.
Conclusions
MICAL1 was upregulated in PC and involved in stimulating the progression of PC cells by activating the WNT/β-catenin signaling pathway. Therefore, MICAL1 is a potential therapeutic target for PC.
Collapse
|
17
|
Inhibitors of RNA and protein synthesis cause Glut4 translocation and increase glucose uptake in adipocytes. Sci Rep 2022; 12:15640. [PMID: 36123369 PMCID: PMC9485115 DOI: 10.1038/s41598-022-19534-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 08/30/2022] [Indexed: 12/29/2022] Open
Abstract
Insulin stimulates glucose uptake in adipocytes by triggering translocation of glucose transporter 4-containg vesicles to the plasma membrane. Under basal conditions, these vesicles (IRVs for insulin-responsive vesicles) are retained inside the cell via a “static” or “dynamic” mechanism. We have found that inhibitors of RNA and protein synthesis, actinomycin D and emetine, stimulate Glut4 translocation and glucose uptake in adipocytes without engaging conventional signaling proteins, such as Akt, TBC1D4, or TUG. Actinomycin D does not significantly affect endocytosis of Glut4 or recycling of transferrin, suggesting that it specifically increases exocytosis of the IRVs. Thus, the intracellular retention of the IRVs in adipocytes requires continuous RNA and protein biosynthesis de novo. These results point out to the existence of a short-lived inhibitor of IRV translocation thus supporting the “static” model.
Collapse
|
18
|
Mendonca A, Thandapani P, Nagarajan P, Venkatesh S, Sundaresan S. Role of microRNAs in regulation of insulin secretion and insulin signaling involved in type 2 diabetes mellitus. J Biosci 2022. [DOI: 10.1007/s12038-022-00295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
19
|
Vorotnikov AV, Popov DV, Makhnovskii PA. Signaling and Gene Expression in Skeletal Muscles in Type 2 Diabetes: Current Results and OMICS Perspectives. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1021-1034. [PMID: 36180992 DOI: 10.1134/s0006297922090139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscles mainly contribute to the emergence of insulin resistance, impaired glucose tolerance and the development of type 2 diabetes. Molecular mechanisms that regulate glucose uptake are diverse, including the insulin-dependent as most important, and others as also significant. They involve a wide range of proteins that control intracellular traffic and exposure of glucose transporters on the cell surface to create an extensive regulatory network. Here, we highlight advantages of the omics approaches to explore the insulin-regulated proteins and genes in human skeletal muscle with varying degrees of metabolic disorders. We discuss methodological aspects of the assessment of metabolic dysregulation and molecular responses of human skeletal muscle to insulin. The known molecular mechanisms of glucose uptake regulation and the first results of phosphoproteomic and transcriptomic studies are reviewed, which unveiled a large-scale array of insulin targets in muscle cells. They demonstrate that a clear depiction of changes that occur during metabolic dysfunction requires systemic and combined analysis at different levels of regulation, including signaling pathways, transcription factors, and gene expression. Such analysis seems promising to explore yet undescribed regulatory mechanisms of glucose uptake by skeletal muscle and identify the key regulators as potential therapeutic targets.
Collapse
Affiliation(s)
- Alexander V Vorotnikov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia.
- National Medical Research Center of Cardiology, Ministry of Healthcare of the Russian Federation, Moscow, 121552, Russia
| | - Daniil V Popov
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia.
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Pavel A Makhnovskii
- Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, 123007, Russia
| |
Collapse
|
20
|
Cornejo PJ, Vergoni B, Ohanna M, Angot B, Gonzalez T, Jager J, Tanti JF, Cormont M. The Stress-Responsive microRNA-34a Alters Insulin Signaling and Actions in Adipocytes through Induction of the Tyrosine Phosphatase PTP1B. Cells 2022; 11:cells11162581. [PMID: 36010657 PMCID: PMC9406349 DOI: 10.3390/cells11162581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022] Open
Abstract
Metabolic stresses alter the signaling and actions of insulin in adipocytes during obesity, but the molecular links remain incompletely understood. Members of the microRNA-34 (miR-34 family play a pivotal role in stress response, and previous studies showed an upregulation of miR-34a in adipose tissue during obesity. Here, we identified miR-34a as a new mediator of adipocyte insulin resistance. We confirmed the upregulation of miR-34a in adipose tissues of obese mice, which was observed in the adipocyte fraction exclusively. Overexpression of miR-34a in 3T3-L1 adipocytes or in fat pads of lean mice markedly reduced Akt activation by insulin and the insulin-induced glucose transport. This was accompanied by a decreased expression of VAMP2, a target of miR-34a, and an increased expression of the tyrosine phosphatase PTP1B. Importantly, PTP1B silencing prevented the inhibitory effect of miR-34a on insulin signaling. Mechanistically, miR-34a decreased the NAD+ level through inhibition of Naprt and Nampt, resulting in an inhibition of Sirtuin-1, which promoted an upregulation of PTP1B. Furthermore, the mRNA expression of Nampt and Naprt was decreased in adipose tissue of obese mice. Collectively, our results identify miR-34a as a new inhibitor of insulin signaling in adipocytes, providing a potential pathway to target to fight insulin resistance.
Collapse
Affiliation(s)
- Pierre-Jean Cornejo
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Bastien Vergoni
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Mickaël Ohanna
- Université Côte d’Azur, Inserm, C3M, “Team Microenvironnement, Signalisation et Cancer”, 06204 Nice, France
| | - Brice Angot
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Teresa Gonzalez
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
- Aix Marseille Université, Inserm, INRAE, C2VN, 13385 Marseille, France
| | - Jennifer Jager
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Jean-François Tanti
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Mireille Cormont
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
- Correspondence: ; Tel.: +33-4-89-15-38-31
| |
Collapse
|
21
|
Endosomal v-ATPase as a Sensor Determining Myocardial Substrate Preference. Metabolites 2022; 12:metabo12070579. [PMID: 35888703 PMCID: PMC9316095 DOI: 10.3390/metabo12070579] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The heart is a metabolically flexible omnivore that can utilize a variety of substrates for energy provision. To fulfill cardiac energy requirements, the healthy adult heart mainly uses long-chain fatty acids and glucose in a balanced manner, but when exposed to physiological or pathological stimuli, it can switch its substrate preference to alternative substrates such as amino acids (AAs) and ketone bodies. Using the failing heart as an example, upon stress, the fatty acid/glucose substrate balance is upset, resulting in an over-reliance on either fatty acids or glucose. A chronic fuel shift towards a single type of substrate is linked with cardiac dysfunction. Re-balancing myocardial substrate preference is suggested as an effective strategy to rescue the failing heart. In the last decade, we revealed that vacuolar-type H+-ATPase (v-ATPase) functions as a key regulator of myocardial substrate preference and, therefore, as a novel potential treatment approach for the failing heart. Fatty acids, glucose, and AAs selectively influence the assembly state of v-ATPase resulting in modulation of its proton-pumping activity. In this review, we summarize these novel insights on v-ATPase as an integrator of nutritional information. We also describe its exploitation as a therapeutic target with focus on supplementation of AA as a nutraceutical approach to fight lipid-induced insulin resistance and contractile dysfunction of the heart.
Collapse
|
22
|
Yuan Y, Kong F, Xu H, Zhu A, Yan N, Yan C. Cryo-EM structure of human glucose transporter GLUT4. Nat Commun 2022; 13:2671. [PMID: 35562357 PMCID: PMC9106701 DOI: 10.1038/s41467-022-30235-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 04/22/2022] [Indexed: 12/28/2022] Open
Abstract
GLUT4 is the primary glucose transporter in adipose and skeletal muscle tissues. Its cellular trafficking is regulated by insulin signaling. Failed or reduced plasma membrane localization of GLUT4 is associated with diabetes. Here, we report the cryo-EM structures of human GLUT4 bound to a small molecule inhibitor cytochalasin B (CCB) at resolutions of 3.3 Å in both detergent micelles and lipid nanodiscs. CCB-bound GLUT4 exhibits an inward-open conformation. Despite the nearly identical conformation of the transmembrane domain to GLUT1, the cryo-EM structure reveals an extracellular glycosylation site and an intracellular helix that is invisible in the crystal structure of GLUT1. The structural study presented here lays the foundation for further mechanistic investigation of the modulation of GLUT4 trafficking. Our methods for cryo-EM analysis of GLUT4 will also facilitate structural determination of many other small size solute carriers. Small solute carriers remain difficult to study by single particle cryo-EM. Here, the authors report the cryo-EM structure of human insulin-responsive glucose transporter GLUT4 (55 kDa) without rigid soluble domains or binders.
Collapse
Affiliation(s)
- Yafei Yuan
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fang Kong
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Hanwen Xu
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Angqi Zhu
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Nieng Yan
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China. .,Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| | - Chuangye Yan
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
23
|
Lee KD, Ilavenil S, Karnan M, Yang CJ, Kim D, Choi KC. Novel Bacillus ginsengihumi CMRO6 Inhibits Adipogenesis via p38MAPK/Erk44/42 and Stimulates Glucose Uptake in 3T3-L1 Pre-Adipocytes through Akt/AS160 Signaling. Int J Mol Sci 2022; 23:4727. [PMID: 35563118 PMCID: PMC9104516 DOI: 10.3390/ijms23094727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/22/2022] Open
Abstract
The health benefits of probiotics have been known for decades, but there has only been limited use of probiotics in the treatment of obesity. In this study, we describe, for the first time, the role of cell-free metabolites (CM) from Bacillus ginsengihumi-RO6 (CMRO6) in adipogenesis and lipogenesis in 3T3-L1 pre-adipocytes. The experimental results show that CMRO6 treatment effectively reduced lipid droplet accumulation and the expression of CCAAT/enhancer-binding protein α and β (C/EBPα and C/EBPβ), peroxisome proliferator-activated receptor γ (PPAR-γ), serum regulatory binding protein 1c (SREBP-1c), fatty acid-binding protein 4 (FABP4), fatty acid synthase (FAS), acetyl CoA carboxylase (ACC), phosphorylated p38MAPK, and Erk44/42. Additionally, CMRO6 treatment significantly increased glucose uptake and phosphorylated Akt (S473), AS160, and TBC1D1 protein expressions. Considering the results of this study, B. ginsengihumi may be a novel probiotic used for the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Kyung Dong Lee
- Department of Companion Animals, Dongsin University, Naju 58245, Korea;
| | - Soundharrajan Ilavenil
- Grassland and Forages Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea; (S.I.); (M.K.)
| | - Muthusamy Karnan
- Grassland and Forages Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea; (S.I.); (M.K.)
| | - Chul-Ju Yang
- Department of Animal Science and Technology, Sunchon National University, Suncheon 57922, Korea;
| | - Dahye Kim
- Animal Genomics and Bioinformatics Division, National Institute of Animal Science, Wanju 55365, Korea;
| | - Ki Choon Choi
- Grassland and Forages Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Korea; (S.I.); (M.K.)
| |
Collapse
|
24
|
Yue X, Wang J, Chang CY, Liu J, Yang X, Zhou F, Qiu X, Bhatt V, Guo JY, Su X, Zhang L, Feng Z, Hu W. Leukemia inhibitory factor drives glucose metabolic reprogramming to promote breast tumorigenesis. Cell Death Dis 2022; 13:370. [PMID: 35440095 PMCID: PMC9018736 DOI: 10.1038/s41419-022-04820-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 12/13/2022]
Abstract
LIF, a multifunctional cytokine, is frequently overexpressed in many types of solid tumors, including breast cancer, and plays an important role in promoting tumorigenesis. Currently, how LIF promotes tumorigenesis is not well-understood. Metabolic reprogramming is a hallmark of cancer cells and a key contributor to cancer progression. However, the role of LIF in cancer metabolic reprogramming is unclear. In this study, we found that LIF increases glucose uptake and drives glycolysis, contributing to breast tumorigenesis. Blocking glucose uptake largely abolishes the promoting effect of LIF on breast tumorigenesis. Mechanistically, LIF overexpression enhances glucose uptake via activating the AKT/GLUT1 axis to promote glycolysis. Blocking the AKT signaling by shRNA or its inhibitors greatly inhibits glycolysis driven by LIF and largely abolishes the promoting effect of LIF on breast tumorigenesis. These results demonstrate an important role of LIF overexpression in glucose metabolism reprogramming in breast cancers, which contributes to breast tumorigenesis. This study also reveals an important mechanism underlying metabolic reprogramming of breast cancers, and identifies LIF and its downstream signaling as potential therapeutic targets for breast cancers, especially those with LIF overexpression.
Collapse
Affiliation(s)
- Xuetian Yue
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Jianming Wang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Chun-Yuan Chang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Xue Yang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Fan Zhou
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Xia Qiu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Vrushank Bhatt
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Jessie Yanxiang Guo
- Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
- Department of Chemical Biology, Rutgers Ernest Mario School of Pharmacy, Piscataway, NJ, USA
| | - Xiaoyang Su
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, USA
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA
| | - Lanjing Zhang
- Department of Pathology, Princeton Medical Center, Plainsboro, NJ, USA
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ, USA.
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, USA.
- Department of Pharmacology, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
25
|
Akinnuga AM, Siboto A, Khumalo B, Sibiya NH, Ngubane P, Khathi A. Evaluation of the effects of bredemolic acid on selected markers of glucose homeostasis in diet-induced prediabetic rats. Arch Physiol Biochem 2022; 128:306-312. [PMID: 31686537 DOI: 10.1080/13813455.2019.1680697] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT Pentacyclic triterpenes (such as maslinic acid) are natural anti-diabetic agents that ameliorate glucose metabolism in diet-induced prediabetes. However, the effects of bredemolic acid (BA), maslinic acid isomer, is yet unknown in prediabetic (PD) conditions. OBJECTIVES To investigate the effects of BA on some glucose homeostasis parameters in high-fat high-carbohydrate (HFHC) diet-induced PD rats. METHODS Thirty-six (36) male rats (150-180 g) were divided into two groups, the normal diet (ND) non-prediabetic, NPD (n = 6) and the HFHC diet PD groups (n = 30). The PD animals were further sub-divided into five groups (n = 6) where they were treated with BA for 12 weeks while monitoring changes in blood glucose, caloric intake, and body weight. RESULTS Diet-induced prediabetes resulted in increased body weight, caloric intake, glycated haemoglobin, and glucose tolerance. BA treatment ameliorated glucose tolerance, lowered plasma insulin and increased expression of glucose transporter 4 (GLUT 4) in rats. CONCLUSIONS BA administration restored glucose homeostasis in diet-induced prediabetes regardless of diet intervention.
Collapse
Affiliation(s)
- Akinjide Moses Akinnuga
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Angezwa Siboto
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Bongiwe Khumalo
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville, South Africa
| | | | - Phikelelani Ngubane
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Andile Khathi
- Department of Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Westville, South Africa
| |
Collapse
|
26
|
Cai H, Wang X, Zhang Z, Chen J, Wang F, Wang L, Liu J. Moderate l-lactate administration suppresses adipose tissue macrophage M1 polarization to alleviate obesity-associated insulin resistance. J Biol Chem 2022; 298:101768. [PMID: 35218776 PMCID: PMC8941214 DOI: 10.1016/j.jbc.2022.101768] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/16/2022] [Accepted: 02/17/2022] [Indexed: 12/11/2022] Open
Abstract
As a crucial metabolic intermediate, l-lactate is involved in redox balance, energy balance, and acid-base balance in organisms. Moderate exercise training transiently elevates plasma l-lactate levels and ameliorates obesity-associated type 2 diabetes. However, whether moderate l-lactate administration improves obesity-associated insulin resistance remains unclear. In this study, we defined 800 mg/kg/day as the dose of moderate l-lactate administration. In mice fed with a high-fat diet (HFD), moderate l-lactate administration for 12 weeks was shown to alleviate weight gain, fat accumulation, and insulin resistance. Along with the phenotype alterations, white adipose tissue thermogenesis was also found to be elevated in HFD-fed mice. Meanwhile, moderate l-lactate administration suppressed the infiltration and proinflammatory M1 polarization of adipose tissue macrophages (ATMs) in HFD-fed mice. Furthermore, l-lactate treatment suppressed the lipopolysaccharide-induced M1 polarization of bone marrow-derived macrophages (BMDMs). l-lactate can bind to the surface receptor GPR132, which typically drives the downstream cAMP-PKA signaling. As a nutrient sensor, AMP-activated protein kinase (AMPK) critically controls macrophage inflammatory signaling and phenotype. Thus, utilizing inhibitors of the kinases PKA and AMPK as well as siRNA against GPR132, we demonstrated that GPR132-PKA-AMPKα1 signaling mediated the suppression caused by l-lactate treatment on BMDM M1 polarization. Finally, l-lactate addition remarkably resisted the impairment of lipopolysaccharide-treated BMDM conditional media on adipocyte insulin sensitivity. In summary, moderate l-lactate administration suppresses ATM proinflammatory M1 polarization through activation of the GPR132-PKA-AMPKα1 signaling pathway to improve insulin resistance in HFD-fed mice, suggesting a new therapeutic and interventional approach to obesity-associated type 2 diabetes.
Collapse
Affiliation(s)
- Hao Cai
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Xin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Zhixin Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Juan Chen
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Fangbin Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Jian Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China; Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, Anhui, China.
| |
Collapse
|
27
|
Di Vincenzo M, Martino M, Lariccia V, Giancola G, Licini C, Di Benedetto G, Arnaldi G, Orciani M. Mesenchymal Stem Cells Exposed to Persistently High Glucocorticoid Levels Develop Insulin-Resistance and Altered Lipolysis: A Promising In Vitro Model to Study Cushing's Syndrome. Front Endocrinol (Lausanne) 2022; 13:816229. [PMID: 35282448 PMCID: PMC8907420 DOI: 10.3389/fendo.2022.816229] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/20/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND In Cushing's syndrome (CS), chronic glucocorticoid excess (GC) and disrupted circadian rhythm lead to insulin resistance (IR), diabetes mellitus, dyslipidaemia and cardiovascular comorbidities. As undifferentiated, self-renewing progenitors of adipocytes, mesenchymal stem cells (MSCs) may display the detrimental effects of excess GC, thus revealing a promising model to study the molecular mechanisms underlying the metabolic complications of CS. METHODS MSCs isolated from the abdominal skin of healthy subjects were treated thrice daily with GCs according to two different regimens: lower, circadian-decreasing (Lower, Decreasing Exposure, LDE) versus persistently higher doses (Higher, Constant Exposure, HCE), aimed at mimicking either the physiological condition or CS, respectively. Subsequently, MSCs were stimulated with insulin and glucose thrice daily, resembling food uptake and both glucose uptake/GLUT-4 translocation and the expression of LIPE, ATGL, IL-6 and TNF-α genes were analyzed at predefined timepoints over three days. RESULTS LDE to GCs did not impair glucose uptake by MSCs, whereas HCE significantly decreased glucose uptake by MSCs only when prolonged. Persistent signs of IR occurred after 30 hours of HCE to GCs. Compared to LDE, MSCs experiencing HCE to GCs showed a downregulation of lipolysis-related genes in the acute period, followed by overexpression once IR was established. CONCLUSIONS Preserving circadian GC rhythmicity is crucial to prevent the occurrence of metabolic alterations. Similar to mature adipocytes, MSCs suffer from IR and impaired lipolysis due to chronic GC excess: MSCs could represent a reliable model to track the mechanisms involved in GC-induced IR throughout cellular differentiation.
Collapse
Affiliation(s)
- Mariangela Di Vincenzo
- Histology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Marianna Martino
- Division of Endocrinology and Metabolic Diseases, Department of Clinical and Molecular Sciences, Umberto I Hospital, Università Politecnica delle Marche, Ancona, Italy
| | - Vincenzo Lariccia
- Pharmacology, Department of Biomedical Sciences and Public Health, Università Politecnica delle Marche, Ancona, Italy
| | - Giulia Giancola
- Division of Endocrinology and Metabolic Diseases, Department of Clinical and Molecular Sciences, Umberto I Hospital, Università Politecnica delle Marche, Ancona, Italy
| | - Caterina Licini
- Histology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Giovanni Di Benedetto
- Clinic of Plastic and Reconstructive Surgery, Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, Ancona, Italy
| | - Giorgio Arnaldi
- Division of Endocrinology and Metabolic Diseases, Department of Clinical and Molecular Sciences, Umberto I Hospital, Università Politecnica delle Marche, Ancona, Italy
- *Correspondence: Giorgio Arnaldi,
| | - Monia Orciani
- Histology, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
28
|
Bogan JS. Ubiquitin-like processing of TUG proteins as a mechanism to regulate glucose uptake and energy metabolism in fat and muscle. Front Endocrinol (Lausanne) 2022; 13:1019405. [PMID: 36246906 PMCID: PMC9556833 DOI: 10.3389/fendo.2022.1019405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
In response to insulin stimulation, fat and muscle cells mobilize GLUT4 glucose transporters to the cell surface to enhance glucose uptake. Ubiquitin-like processing of TUG (Aspscr1, UBXD9) proteins is a central mechanism to regulate this process. Here, recent advances in this area are reviewed. The data support a model in which intact TUG traps insulin-responsive "GLUT4 storage vesicles" at the Golgi matrix by binding vesicle cargoes with its N-terminus and matrix proteins with its C-terminus. Insulin stimulation liberates these vesicles by triggering endoproteolytic cleavage of TUG, mediated by the Usp25m protease. Cleavage occurs in fat and muscle cells, but not in fibroblasts or other cell types. Proteolytic processing of intact TUG generates TUGUL, a ubiquitin-like protein modifier, as the N-terminal cleavage product. In adipocytes, TUGUL modifies a single protein, the KIF5B kinesin motor, which carries GLUT4 and other vesicle cargoes to the cell surface. In muscle, this or another motor may be modified. After cleavage of intact TUG, the TUG C-terminal product is extracted from the Golgi matrix by the p97 (VCP) ATPase. In both muscle and fat, this cleavage product enters the nucleus, binds PPARγ and PGC-1α, and regulates gene expression to promote fatty acid oxidation and thermogenesis. The stability of the TUG C-terminal product is regulated by an Ate1 arginyltransferase-dependent N-degron pathway, which may create a feedback mechanism to control oxidative metabolism. Although it is now clear that TUG processing coordinates glucose uptake with other aspects of physiology and metabolism, many questions remain about how this pathway is regulated and how it is altered in metabolic disease in humans.
Collapse
Affiliation(s)
- Jonathan S. Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, United States
- Yale Center for Molecular and Systems Metabolism, Yale School of Medicine, New Haven, CT, United States
- *Correspondence: Jonathan S. Bogan,
| |
Collapse
|
29
|
Xu Y, Jin L, Toomre D. Imaging Single-Vesicle Exocytosis with Total Internal Reflection Fluorescence Microscopy (TIRFM). Methods Mol Biol 2022; 2473:157-164. [PMID: 35819765 DOI: 10.1007/978-1-0716-2209-4_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Total internal reflection fluorescence microscopy (TIRFM) provides extremely thin optical sectioning with excellent signal-to-noise ratios, which allows for visualization of membrane dynamics at the cell surface with superb spatiotemporal resolution. In this chapter, TIRFM is used to record and analyze exocytosis of single glucose transporter-4 (GLUT4) containing vesicles in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Yingke Xu
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China.
| | - Luhong Jin
- Department of Biomedical Engineering, Key Laboratory of Biomedical Engineering of Ministry of Education, State Key Laboratory of Modern Optical Instrumentation, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China
| | - Derek Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Pánico P, Velasco M, Salazar AM, Picones A, Ortiz-Huidobro RI, Guerrero-Palomo G, Salgado-Bernabé ME, Ostrosky-Wegman P, Hiriart M. Is Arsenic Exposure a Risk Factor for Metabolic Syndrome? A Review of the Potential Mechanisms. Front Endocrinol (Lausanne) 2022; 13:878280. [PMID: 35651975 PMCID: PMC9150370 DOI: 10.3389/fendo.2022.878280] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/11/2022] [Indexed: 12/14/2022] Open
Abstract
Exposure to arsenic in drinking water is a worldwide health problem. This pollutant is associated with increased risk of developing chronic diseases, including metabolic diseases. Metabolic syndrome (MS) is a complex pathology that results from the interaction between environmental and genetic factors. This condition increases the risk of developing type 2 diabetes, cardiovascular diseases, and cancer. The MS includes at least three of the following signs, central obesity, impaired fasting glucose, insulin resistance, dyslipidemias, and hypertension. Here, we summarize the existing evidence of the multiple mechanisms triggered by arsenic to developing the cardinal signs of MS, showing that this pollutant could contribute to the multifactorial origin of this pathology.
Collapse
Affiliation(s)
- Pablo Pánico
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Myrian Velasco
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Ana María Salazar
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Arturo Picones
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Rosa Isela Ortiz-Huidobro
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Guerrero-Palomo
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Manuel Eduardo Salgado-Bernabé
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Patricia Ostrosky-Wegman
- Department of Genomic Medicine and Environmental Toxicology. Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Marcia Hiriart
- Department of Cognitive Neurosciences, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- *Correspondence: Marcia Hiriart,
| |
Collapse
|
31
|
Rao XS, Cong XX, Gao XK, Shi YP, Shi LJ, Wang JF, Ni CY, He MJ, Xu Y, Yi C, Meng ZX, Liu J, Lin P, Zheng LL, Zhou YT. AMPK-mediated phosphorylation enhances the auto-inhibition of TBC1D17 to promote Rab5-dependent glucose uptake. Cell Death Differ 2021; 28:3214-3234. [PMID: 34045668 PMCID: PMC8630067 DOI: 10.1038/s41418-021-00809-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 05/13/2021] [Accepted: 05/17/2021] [Indexed: 02/04/2023] Open
Abstract
Dysregulation of glucose homeostasis contributes to insulin resistance and type 2 diabetes. Whilst exercise stimulated activation of AMP-activated protein kinase (AMPK), an important energy sensor, has been highlighted for its potential to promote insulin-stimulated glucose uptake, the underlying mechanisms for this remain largely unknown. Here we found that AMPK positively regulates the activation of Rab5, a small GTPase which is involved in regulating Glut4 translocation, in both myoblasts and skeletal muscles. We further verified that TBC1D17, identified as a potential interacting partner of Rab5 in our recent study, is a novel GTPase activating protein (GAP) of Rab5. TBC1D17-Rab5 axis regulates transport of Glut1, Glut4, and transferrin receptor. TBC1D17 interacts with Rab5 or AMPK via its TBC domain or N-terminal 1-306 region (N-Ter), respectively. Moreover, AMPK phosphorylates the Ser 168 residue of TBC1D17 which matches the predicted AMPK consensus motif. N-Ter of TBC1D17 acts as an inhibitory region by directly interacting with the TBC domain. Ser168 phosphorylation promotes intra-molecular interaction and therefore enhances the auto-inhibition of TBC1D17. Our findings reveal that TBC1D17 acts as a molecular bridge that links AMPK and Rab5 and delineate a previously unappreciated mechanism by which the activation of TBC/RabGAP is regulated.
Collapse
Affiliation(s)
- Xi Sheng Rao
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Xia Cong
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiu Kui Gao
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yin Pu Shi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Jing Shi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Feng Wang
- grid.13402.340000 0004 1759 700XDepartment of Respiratory Medicine, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chen-Yao Ni
- grid.35403.310000 0004 1936 9991The School of Molecular and Cellular Biology, University of Illinois at Urbana Champaign, Urbana, IL USA
| | - Ming Jie He
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yingke Xu
- grid.13402.340000 0004 1759 700XDepartment of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Endocrinology, the Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cong Yi
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhuo-Xian Meng
- grid.13402.340000 0004 1759 700XDepartment of Pathology and Pathophysiology and Zhejiang Provincial Key Laboratory of Pancreatic Disease of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinling Liu
- grid.13402.340000 0004 1759 700XDepartment of Pulmonology, the Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Peng Lin
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Ling Zheng
- grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Ting Zhou
- grid.13402.340000 0004 1759 700XDepartment of Biochemistry and Department of Orthopaedic Surgery of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XKey Laboratory of Tissue Engineering and Regenerative Medicine of Zhejiang Province, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XZJU-UoE Institute, Zhejiang University School of Medicine, Hangzhou, China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
32
|
Szałabska-Rąpała K, Borymska W, Kaczmarczyk-Sedlak I. Effectiveness of Magnolol, a Lignan from Magnolia Bark, in Diabetes, Its Complications and Comorbidities-A Review. Int J Mol Sci 2021; 22:10050. [PMID: 34576213 PMCID: PMC8467064 DOI: 10.3390/ijms221810050] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a chronic metabolic disease characterized by disturbances in carbohydrate, protein, and lipid metabolism, often accompanied by oxidative stress. Diabetes treatment is a complicated process in which, in addition to the standard pharmacological action, it is necessary to append a comprehensive approach. Introducing the aspect of non-pharmacological treatment of diabetes allows one to alleviate its many adverse complications. Therefore, it seems important to look for substances that, when included in the daily diet, can improve diabetic parameters. Magnolol, a polyphenolic compound found in magnolia bark, is known for its health-promoting activities and multidirectional beneficial effects on the body. Accordingly, the goal of this review is to systematize the available scientific literature on its beneficial effects on type 2 diabetes and its complications. Taking the above into consideration, the article collects data on the favorable effects of magnolol on parameters related to glycemia, lipid metabolism, or oxidative stress in the course of diabetes. After careful analysis of many scientific articles, it can be concluded that this lignan is a promising agent supporting the conventional therapies with antidiabetic drugs in order to manage diabetes and diabetes-related diseases.
Collapse
Affiliation(s)
- Katarzyna Szałabska-Rąpała
- Doctoral School of the Medical University of Silesia in Katowice, Discipline of Pharmaceutical Sciences, Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Weronika Borymska
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (W.B.); (I.K.-S.)
| | - Ilona Kaczmarczyk-Sedlak
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (W.B.); (I.K.-S.)
| |
Collapse
|
33
|
Hülsemann M, Sanchez C, Verkhusha PV, Des Marais V, Mao SPH, Donnelly SK, Segall JE, Hodgson L. TC10 regulates breast cancer invasion and metastasis by controlling membrane type-1 matrix metalloproteinase at invadopodia. Commun Biol 2021; 4:1091. [PMID: 34531530 PMCID: PMC8445963 DOI: 10.1038/s42003-021-02583-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 08/23/2021] [Indexed: 01/12/2023] Open
Abstract
During breast cancer metastasis, cancer cell invasion is driven by actin-rich protrusions called invadopodia, which mediate the extracellular matrix degradation required for the success of the invasive cascade. In this study, we demonstrate that TC10, a member of a Cdc42 subfamily of p21 small GTPases, regulates the membrane type 1 matrix metalloproteinase (MT1-MMP)-driven extracellular matrix degradation at invadopodia. We show that TC10 is required for the plasma membrane surface exposure of MT1-MMP at these structures. By utilizing our Förster resonance energy transfer (FRET) biosensor, we demonstrate the p190RhoGAP-dependent regulation of spatiotemporal TC10 activity at invadopodia. We identified a pathway that regulates invadopodia-associated TC10 activity and function through the activation of p190RhoGAP and the downstream interacting effector Exo70. Our findings reveal the role of a previously unknown regulator of vesicular fusion at invadopodia, TC10 GTPase, in breast cancer invasion and metastasis.
Collapse
Affiliation(s)
- Maren Hülsemann
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Colline Sanchez
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Polina V Verkhusha
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Vera Des Marais
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Serena P H Mao
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Sara K Donnelly
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jeffrey E Segall
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Louis Hodgson
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
34
|
Zhang L, Gopalasingam G, Herzog H. Ninjin'yoeito, a herbal medicine, enhances glucose tolerance in mice. Neuropeptides 2021; 88:102150. [PMID: 33895618 DOI: 10.1016/j.npep.2021.102150] [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: 10/20/2020] [Revised: 02/03/2021] [Accepted: 03/03/2021] [Indexed: 10/21/2022]
Abstract
The prevalence of Type 2 diabetes increases under conditions of obesity but also due to aging. While a variety of treatment options are being explored there are still many unanswered questions about the underlying mechanisms for the aetiology and progression of this illness. Here we show that pre-treatment with Ninjin'yoeito (NYT), a herbal medicine composed of 12 different ingrediencies, before a glucose challenge results in significantly improved glucose tolerance. This occurs in the absence of significant alterations in insulin excursion compared to vehicle treatment, indicating NYT improves insulin responsiveness and/or insulin-independent glucose disposal. Furthermore, we identify Ginseng - one of the 12 ingredients of NYT - as one key component contributing to NYT's effect on glucose clearance. Importantly, lack of Y4 receptor signalling abolishes the positive effects of NYT on glucose tolerance suggesting Y4 receptor-controlled pathways are crucial in mediating this action of NYT. Using c-fos as neuronal activation marker, we show NYT activates the area postrema - a circumventricular organ in the brainstem that expresses high level of Y4 receptors, supporting an involvement of brainstem Y4 signalling in NYT-activated central networks. Together, these data suggest that NYT is a positive influencer of glucose metabolism in insulin-sensitive tissues and the mechanistic actions of NYT include brainstem Y4 circuitries.
Collapse
Affiliation(s)
- Lei Zhang
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, Australia; St. Vincent's Clinical School, University of NSW, Sydney, Australia.
| | - Gopana Gopalasingam
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Darlinghurst, Sydney, Australia; School of Medical Sciences, University of NSW, Sydney, NSW, Australia; Faculty of Medicine, University of NSW, Sydney, NSW, Australia
| |
Collapse
|
35
|
Yang YS, Yang S, Li D, Li W. Vitamin D affects the Warburg effect and stemness maintenance of non-small-cell lung cancer cells by regulating PI3K/AKT/mTOR signaling pathway. Curr Cancer Drug Targets 2021; 22:86-95. [PMID: 34325639 DOI: 10.2174/1568009621666210729100300] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/16/2021] [Accepted: 04/25/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Non-small-cell lung cancer (NSCLC) is the most prevalent form of lung cancer, accounting for approximately 85% of all lung cancer cases and resulting in high morbidity and mortality. Previous studies have demonstrated that 1,25-dihydroxy-vitamin-D3 (vitamin D) exhibited anti-cancer activity against breast and prostate cancer. OBJECTIVES The aim of the current study is to investigate the effect of vitamin D on NSCLC and its underlying mechanism. METHODS The effects of vitamin D on stemness maintenance and the Warburg effect in NSCLC cells were investigated both in vitro and in vivo. RESULTS & DISCUSSION In vitro experiments revealed that vitamin D inhibited glycolysis and stemness maintenance in A549 and NCI-H1975 cells. Both in vitro and in vivo experiments indicated that vitamin D attenuated the expression of metabolism-related enzymes associated with the Warburg effect (GLUT1, LDHA, HK2, and PKM2). In addition, vitamin D down-regulated the expression of stemness-related genes (Oct-4, SOX-2, and Nanog) and the expression of PI3K, AKT, and mTOR. CONCLUSION Overall, these findings suggest that vitamin D suppresses the Warburg effect and stemness maintenance in NSCLC cells via the inactivation of PI3K/AKT/mTOR signaling, thereby inhibiting the progression of NSCLC. The current study indicates that vitamin D is a potential candidate in therapeutic strategies against NSCLC.
Collapse
Affiliation(s)
- Yiyan Song Yang
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Songyisha Yang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Dejia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, Wuhan 430071, China
| | - Wen Li
- Department of Emergency, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
36
|
Zhou L, Han X, Li W, Wang N, Yao L, Zhao Y, Zhang L. N6-methyladenosine Demethylase FTO Induces the Dysfunctions of Ovarian Granulosa Cells by Upregulating Flotillin 2. Reprod Sci 2021; 29:1305-1315. [PMID: 34254281 DOI: 10.1007/s43032-021-00664-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
Polycystic ovarian syndrome (PCOS) is often accompanied by overweight/obesity and insulin resistance. The dysfunctions of ovarian granulosa cells (GCs) are closely linked with the pathogenesis of PCOS. Fat mass and obesity-associated gene (FTO), an N6-methyladenosine (m6A) demethylase, has been reported to be implicated in the risks and insulin resistance of PCOS. However, the roles of FTO in the development of GCs along with its m6A-related regulatory mechanisms are poorly defined. Cell proliferative ability was detected by MTT assay. Cell apoptotic rate was measured via flow cytometry. Insulin resistance was assessed by GLUT4 transport potential. The mRNA and protein levels of FTO and flotillin 2 (FLOT2) were determined by RT-qPCR and western blot assays, respectively. FLOT2 was screened out to be a potential FTO target through differential expression analysis for the GSE95728 dataset and target prediction analysis by POSTAR2 and STARBASE databases. The interaction between FTO and FLOT2 was analyzed by RNA immunoprecipitation (RIP) assay. The effect of FTO upregulation on FLOT2 m6A level was measured by methylated RIP (meRIP) assay. FLOT2 mRNA stability was examined by actinomycin D assay. FTO overexpression facilitated cell proliferation, hindered cell apoptosis, and induced insulin resistance in GCs. FTO promoted FLOT2 expression by reducing m6A level on FLOT2 mRNA and increasing FLOT2 mRNA stability. FLOT2 loss weakened the effects of FTO overexpression on cell proliferation/apoptosis and insulin resistance in GCs. FTO induced the dysfunctions of GCs by upregulating FLOT2, suggesting that FTO/FLOT2 might play a role in the pathophysiology of PCOS.
Collapse
Affiliation(s)
- Li Zhou
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China
| | - Xiao Han
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China
| | - Wei Li
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China
| | - Ning Wang
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China
| | - Lan Yao
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China
| | - Yunhe Zhao
- Department of Gynaecology and Obstetrics, Xiangyang Central Hospital, The Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Liqun Zhang
- Department of Gynecology, The First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin, China.
| |
Collapse
|
37
|
Activated Alpha-2 Macroglobulin Improves Insulin Response via LRP1 in Lipid-Loaded HL-1 Cardiomyocytes. Int J Mol Sci 2021; 22:ijms22136915. [PMID: 34203120 PMCID: PMC8268138 DOI: 10.3390/ijms22136915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Activated alpha-2 Macroglobulin (α2M*) is specifically recognized by the cluster I/II of LRP1 (Low-density lipoprotein Receptor-related Protein-1). LRP1 is a scaffold protein for insulin receptor involved in the insulin-induced glucose transporter type 4 (GLUT4) translocation to plasma membrane and glucose uptake in different types of cells. Moreover, the cluster II of LRP1 plays a critical role in the internalization of atherogenic lipoproteins, such as aggregated Low-density Lipoproteins (aggLDL), promoting intracellular cholesteryl ester (CE) accumulation mainly in arterial intima and myocardium. The aggLDL uptake by LRP1 impairs GLUT4 traffic and the insulin response in cardiomyocytes. However, the link between CE accumulation, insulin action, and cardiac dysfunction are largely unknown. Here, we found that α2M* increased GLUT4 expression on cell surface by Rab4, Rab8A, and Rab10-mediated recycling through PI3K/Akt and MAPK/ERK signaling activation. Moreover, α2M* enhanced the insulin response increasing insulin-induced glucose uptake rate in the myocardium under normal conditions. On the other hand, α2M* blocked the intracellular CE accumulation, improved the insulin response and reduced cardiac damage in HL-1 cardiomyocytes exposed to aggLDL. In conclusion, α2M* by its agonist action on LRP1, counteracts the deleterious effects of aggLDL in cardiomyocytes, which may have therapeutic implications in cardiovascular diseases associated with hypercholesterolemia.
Collapse
|
38
|
A-769662 inhibits adipocyte glucose uptake in an AMPK-independent manner. Biochem J 2021; 478:633-646. [PMID: 33493298 DOI: 10.1042/bcj20200659] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/22/2022]
Abstract
Activation of AMP-activated protein kinase (AMPK) is considered a valid strategy for the treatment of type 2 diabetes. However, despite the importance of adipose tissue for whole-body energy homeostasis, the effect of AMPK activation in adipocytes has only been studied to a limited extent and mainly with the AMP-mimetic 5-aminoimidazole-4-carboxamide-1-β-d-ribofuranoside (AICAR), which has limited specificity. The aim of this study was to evaluate the effect of the allosteric AMPK activators A-769662 and 991 on glucose uptake in adipocytes. For this purpose, primary rat or human adipocytes, and cultured 3T3-L1 adipocytes, were treated with either of the allosteric activators, or AICAR, and basal and insulin-stimulated glucose uptake was assessed. Additionally, the effect of AMPK activators on insulin-stimulated phosphorylation of Akt and Akt substrate of 160 kDa was assessed. Furthermore, primary adipocytes from ADaM site binding drug-resistant AMPKβ1 S108A knock-in mice were employed to investigate the specificity of the drugs for the observed effects. Our results show that insulin-stimulated adipocyte glucose uptake was significantly reduced by A-769662 but not 991, yet neither activator had any clear effects on basal or insulin-stimulated Akt/AS160 signaling. The use of AMPKβ1 S108A mutant-expressing adipocytes revealed that the observed inhibition of glucose uptake by A-769662 is most likely AMPK-independent, a finding which is supported by the rapid inhibitory effect A-769662 exerts on glucose uptake in 3T3-L1 adipocytes. These data suggest that AMPK activation per se does not inhibit glucose uptake in adipocytes and that the effects of AICAR and A-769662 are AMPK-independent.
Collapse
|
39
|
Wagner G, Fenzl A, Lindroos-Christensen J, Einwallner E, Husa J, Witzeneder N, Rauscher S, Gröger M, Derdak S, Mohr T, Sutterlüty H, Klinglmüller F, Wolkerstorfer S, Fondi M, Hoermann G, Cao L, Wagner O, Kiefer FW, Esterbauer H, Bilban M. LMO3 reprograms visceral adipocyte metabolism during obesity. J Mol Med (Berl) 2021; 99:1151-1171. [PMID: 34018016 PMCID: PMC8313462 DOI: 10.1007/s00109-021-02089-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 05/05/2021] [Accepted: 05/10/2021] [Indexed: 01/02/2023]
Abstract
Abstract Obesity and body fat distribution are important risk factors for the development of type 2 diabetes and metabolic syndrome. Evidence has accumulated that this risk is related to intrinsic differences in behavior of adipocytes in different fat depots. We recently identified LIM domain only 3 (LMO3) in human mature visceral adipocytes; however, its function in these cells is currently unknown. The aim of this study was to determine the potential involvement of LMO3-dependent pathways in the modulation of key functions of mature adipocytes during obesity. Based on a recently engineered hybrid rAAV serotype Rec2 shown to efficiently transduce both brown adipose tissue (BAT) and white adipose tissue (WAT), we delivered YFP or Lmo3 to epididymal WAT (eWAT) of C57Bl6/J mice on a high-fat diet (HFD). The effects of eWAT transduction on metabolic parameters were evaluated 10 weeks later. To further define the role of LMO3 in insulin-stimulated glucose uptake, insulin signaling, adipocyte bioenergetics, as well as endocrine function, experiments were conducted in 3T3-L1 adipocytes and newly differentiated human primary mature adipocytes, engineered for transient gain or loss of LMO3 expression, respectively. AAV transduction of eWAT results in strong and stable Lmo3 expression specifically in the adipocyte fraction over a course of 10 weeks with HFD feeding. LMO3 expression in eWAT significantly improved insulin sensitivity and healthy visceral adipose tissue expansion in diet-induced obesity, paralleled by increased serum adiponectin. In vitro, LMO3 expression in 3T3-L1 adipocytes increased PPARγ transcriptional activity, insulin-stimulated GLUT4 translocation and glucose uptake, as well as mitochondrial oxidative capacity in addition to fatty acid oxidation. Mechanistically, LMO3 induced the PPARγ coregulator Ncoa1, which was required for LMO3 to enhance glucose uptake and mitochondrial oxidative gene expression. In human mature adipocytes, LMO3 overexpression promoted, while silencing of LMO3 suppressed mitochondrial oxidative capacity. LMO3 expression in visceral adipose tissue regulates multiple genes that preserve adipose tissue functionality during obesity, such as glucose metabolism, insulin sensitivity, mitochondrial function, and adiponectin secretion. Together with increased PPARγ activity and Ncoa1 expression, these gene expression changes promote insulin-induced GLUT4 translocation, glucose uptake in addition to increased mitochondrial oxidative capacity, limiting HFD-induced adipose dysfunction. These data add LMO3 as a novel regulator improving visceral adipose tissue function during obesity. Key messages LMO3 increases beneficial visceral adipose tissue expansion and insulin sensitivity in vivo. LMO3 increases glucose uptake and oxidative mitochondrial activity in adipocytes. LMO3 increases nuclear coactivator 1 (Ncoa1). LMO3-enhanced glucose uptake and mitochondrial gene expression requires Ncoa1.
Supplementary Information The online version contains supplementary material available at 10.1007/s00109-021-02089-9.
Collapse
Affiliation(s)
- Gabriel Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Anna Fenzl
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Josefine Lindroos-Christensen
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Novo Nordisk, Maaloev, Denmark
| | - Elisa Einwallner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Julia Husa
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Nadine Witzeneder
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Sabine Rauscher
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Marion Gröger
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, 1090, Vienna, Austria
| | - Thomas Mohr
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Hedwig Sutterlüty
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian Klinglmüller
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, 1090, Vienna, Austria.,Austrian Medicines & Medical Devices Agency, 1200, Vienna, Austria
| | - Silviya Wolkerstorfer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria.,Institute of Cardiovascular Prevention, Ludwig-Maximilians-University, 80336, Munich, Germany
| | - Martina Fondi
- University of Applied Sciences, FH Campus Wien, 1100, Vienna, Austria
| | - Gregor Hoermann
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria.,Central Institute of Medical and Chemical Laboratory Diagnostics, University Hospital Innsbruck, 6020, Innsbruck, Austria
| | - Lei Cao
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - Oswald Wagner
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Florian W Kiefer
- Clinical Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, 1090, Vienna, Austria
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria. .,Core Facilities, Medical University of Vienna, 1090, Vienna, Austria.
| |
Collapse
|
40
|
Drissi F, Lahfa F, Gonzalez T, Peiretti F, Tanti JF, Haddad M, Fabre N, Govers R. A Citrullus colocynthis fruit extract acutely enhances insulin-induced GLUT4 translocation and glucose uptake in adipocytes by increasing PKB phosphorylation. JOURNAL OF ETHNOPHARMACOLOGY 2021; 270:113772. [PMID: 33418030 DOI: 10.1016/j.jep.2020.113772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/14/2020] [Accepted: 12/24/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Citrullus colocynthis (L.) Schrad is a common fruit in traditional medicine and used as remedy against various diseases, especially diabetes. Up to now, its anti-diabetic effects have been fully attributed to its enhancement of pancreatic insulin secretion. Whether C. colocynthis also ameliorates insulin action in peripheral tissues has not been investigated. AIM OF THE STUDY In the present study, using 3T3-L1 adipocytes as cell model, we have investigated whether colocynth fruit extracts affect insulin action. MATERIALS AND METHODS Various extracts were prepared from the C. colocynthis fruit and screened using a cell-based 96 well plate GLUT4 translocation assay. Promising extracts were further studied for their effects on glucose uptake and cell viability. The effect on insulin signal transduction was determined by Western blot and the molecular composition was established by LC-MS. RESULTS The ethyl acetate fractions of aqueous non-defatted extracts of seed and pulp, designated Sna1 and Pna1, acutely enhanced insulin-induced GLUT4 translocation. In accordance, both extracts increased insulin-stimulated cellular glucose uptake. Pna1, which displayed greater effects on GLUT4 and glucose uptake than Sna1, was further investigated and was demonstrated to increase GLUT4 translocation without changing the half-maximum dose (ED50) of insulin, nor changing GLUT4 translocation kinetics. At the molecular level, Pna1 was found to enhance insulin-induced PKB phosphorylation without changing phosphorylation of the insulin receptor. Pna1 appeared not to be toxic to cells and, like insulin, restored cell viability during serum starvation. By investigating the molecular composition of Pna1, nine compounds were identified that made up 87% of the mass of the extract, one of which is likely to be responsible for the insulin-enhancing effects of Pna1. CONCLUSIONS The C. colocynthis fruit possesses insulin-enhancing activity. This activity may explain in part its anti-diabetic effects in traditional medicine. It also identifies the C. colocynthis as a source of a potential novel insulin enhancer that may prove to be useful to reduce hyperglycemia in type 2 diabetes.
Collapse
Affiliation(s)
- Farah Drissi
- Department of Synthesis and Biological Activities, University of Abou Bekr Belkaïd, 119 13000, Tlemcen, Algeria.
| | - Farid Lahfa
- Department of Synthesis and Biological Activities, University of Abou Bekr Belkaïd, 119 13000, Tlemcen, Algeria.
| | - Teresa Gonzalez
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| | - Franck Peiretti
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| | - Jean-François Tanti
- Université Côte D'Azur, INSERM, C3M, Team "Cellular and Molecular Physiopathology of Obesity", 06204, Nice, France.
| | - Mohamed Haddad
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400, Toulouse, France.
| | - Nicolas Fabre
- UMR 152 Pharma Dev, Université de Toulouse, IRD, UPS, 31400, Toulouse, France.
| | - Roland Govers
- Aix Marseille Université, INSERM, INRAE, C2VN, 13385, Marseille, France.
| |
Collapse
|
41
|
Machin PA, Tsonou E, Hornigold DC, Welch HCE. Rho Family GTPases and Rho GEFs in Glucose Homeostasis. Cells 2021; 10:cells10040915. [PMID: 33923452 PMCID: PMC8074089 DOI: 10.3390/cells10040915] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/11/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of glucose homeostasis leading to metabolic syndrome and type 2 diabetes is the cause of an increasing world health crisis. New intriguing roles have emerged for Rho family GTPases and their Rho guanine nucleotide exchange factor (GEF) activators in the regulation of glucose homeostasis. This review summates the current knowledge, focusing in particular on the roles of Rho GEFs in the processes of glucose-stimulated insulin secretion by pancreatic β cells and insulin-stimulated glucose uptake into skeletal muscle and adipose tissues. We discuss the ten Rho GEFs that are known so far to regulate glucose homeostasis, nine of which are in mammals, and one is in yeast. Among the mammalian Rho GEFs, P-Rex1, Vav2, Vav3, Tiam1, Kalirin and Plekhg4 were shown to mediate the insulin-stimulated translocation of the glucose transporter GLUT4 to the plasma membrane and/or insulin-stimulated glucose uptake in skeletal muscle or adipose tissue. The Rho GEFs P-Rex1, Vav2, Tiam1 and β-PIX were found to control the glucose-stimulated release of insulin by pancreatic β cells. In vivo studies demonstrated the involvement of the Rho GEFs P-Rex2, Vav2, Vav3 and PDZ-RhoGEF in glucose tolerance and/or insulin sensitivity, with deletion of these GEFs either contributing to the development of metabolic syndrome or protecting from it. This research is in its infancy. Considering that over 80 Rho GEFs exist, it is likely that future research will identify more roles for Rho GEFs in glucose homeostasis.
Collapse
Affiliation(s)
- Polly A. Machin
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
| | - Elpida Tsonou
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - David C. Hornigold
- Bioscience Metabolism, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Cambridge CB22 3AT, UK;
| | - Heidi C. E. Welch
- Signalling Programme, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK; (P.A.M.); (E.T.)
- Correspondence: ; Tel.: +44-(0)1223-496-596
| |
Collapse
|
42
|
Lanzillotta C, Tramutola A, Di Giacomo G, Marini F, Butterfield DA, Di Domenico F, Perluigi M, Barone E. Insulin resistance, oxidative stress and mitochondrial defects in Ts65dn mice brain: A harmful synergistic path in down syndrome. Free Radic Biol Med 2021; 165:152-170. [PMID: 33516914 DOI: 10.1016/j.freeradbiomed.2021.01.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 01/12/2021] [Accepted: 01/22/2021] [Indexed: 02/06/2023]
Abstract
Dysregulation of brain insulin signaling with reduced downstream neuronal survival and plasticity mechanisms are fundamental abnormalities observed in Alzheimer disease (AD). This phenomenon, known as brain insulin resistance, is associated with poor cognitive performance and is driven by the inhibition of IRS1. Since Down syndrome (DS) and AD neuropathology share many common features, we investigated metabolic aspects of neurodegeneration in DS and whether they contribute to early onset AD in DS. We evaluated levels and activation of proteins belonging to the insulin signaling pathway (IR, IRS1, BVR-A, MAPK, PTEN, Akt, GSK3β, PKCζ, AS160, GLUT4) in the frontal cortex of Ts65dn (DS model) (n = 5-6/group) and euploid mice (n = 6/group) at different ages (1, 3, 9 and 18 months). Furthermore, we analyzed whether changes of brain insulin signaling were associated with alterations of: (i) proteins regulating brain energy metabolism (mitochondrial complexes, hexokinase-II, Sirt1); (ii) oxidative stress (OS) markers (iii) APP cleavage; and (iv) proteins mediating synaptic plasticity mechanisms (PSD95, syntaxin-1 and BDNF). Ts65dn mice showed an overall impairment of the above-mentioned pathways, mainly characterized by defects of proteins activation state. Such alterations start early in life (at 1 month, during brain maturation). In particular, accumulation of inhibited IRS1, together with the uncoupling among the proteins downstream from IRS1 (brain insulin resistance), characterize Ts65dn mice. Furthermore, reduced levels of mitochondrial complexes and Sirt1, as well as increased indices of OS also were observed. These alterations precede the accumulation of APP-C99 in Ts65dn mice. Tellingly, oxidative stress levels were negatively associated with IR, IRS1 and AS160 activation as well as mitochondrial complexes levels in Ts65dn mice, suggesting a role for oxidative stress in the observed alterations. We propose that a close link exists among brain insulin resistance, mitochondrial defects and OS that contributes to brain dysfunctions observed in DS, likely favoring the development of AD in DS.
Collapse
Affiliation(s)
- Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Graziella Di Giacomo
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Federico Marini
- Department of Chemistry, Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40506-0055, USA
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi-Fanelli", Sapienza University of Rome, Piazzale A. Moro 5, 00185, Roma, Italy.
| |
Collapse
|
43
|
Chamberlain LH, Shipston MJ, Gould GW. Regulatory effects of protein S-acylation on insulin secretion and insulin action. Open Biol 2021; 11:210017. [PMID: 33784857 PMCID: PMC8061761 DOI: 10.1098/rsob.210017] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/02/2021] [Indexed: 12/23/2022] Open
Abstract
Post-translational modifications (PTMs) such as phosphorylation and ubiquitination are well-studied events with a recognized importance in all aspects of cellular function. By contrast, protein S-acylation, although a widespread PTM with important functions in most physiological systems, has received far less attention. Perturbations in S-acylation are linked to various disorders, including intellectual disability, cancer and diabetes, suggesting that this less-studied modification is likely to be of considerable biological importance. As an exemplar, in this review, we focus on the newly emerging links between S-acylation and the hormone insulin. Specifically, we examine how S-acylation regulates key components of the insulin secretion and insulin response pathways. The proteins discussed highlight the diverse array of proteins that are modified by S-acylation, including channels, transporters, receptors and trafficking proteins and also illustrate the diverse effects that S-acylation has on these proteins, from membrane binding and micro-localization to regulation of protein sorting and protein interactions.
Collapse
Affiliation(s)
- Luke H. Chamberlain
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Michael J. Shipston
- Centre for Discovery Brain Sciences, Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Gwyn W. Gould
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
44
|
Habtemichael EN, Li DT, Camporez JP, Westergaard XO, Sales CI, Liu X, López-Giráldez F, DeVries SG, Li H, Ruiz DM, Wang KY, Sayal BS, González Zapata S, Dann P, Brown SN, Hirabara S, Vatner DF, Goedeke L, Philbrick W, Shulman GI, Bogan JS. Insulin-stimulated endoproteolytic TUG cleavage links energy expenditure with glucose uptake. Nat Metab 2021; 3:378-393. [PMID: 33686286 PMCID: PMC7990718 DOI: 10.1038/s42255-021-00359-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/05/2021] [Indexed: 12/12/2022]
Abstract
TUG tethering proteins bind and sequester GLUT4 glucose transporters intracellularly, and insulin stimulates TUG cleavage to translocate GLUT4 to the cell surface and increase glucose uptake. This effect of insulin is independent of phosphatidylinositol 3-kinase, and its physiological relevance remains uncertain. Here we show that this TUG cleavage pathway regulates both insulin-stimulated glucose uptake in muscle and organism-level energy expenditure. Using mice with muscle-specific Tug (Aspscr1)-knockout and muscle-specific constitutive TUG cleavage, we show that, after GLUT4 release, the TUG C-terminal cleavage product enters the nucleus, binds peroxisome proliferator-activated receptor (PPAR)γ and its coactivator PGC-1α and regulates gene expression to promote lipid oxidation and thermogenesis. This pathway acts in muscle and adipose cells to upregulate sarcolipin and uncoupling protein 1 (UCP1), respectively. The PPARγ2 Pro12Ala polymorphism, which reduces diabetes risk, enhances TUG binding. The ATE1 arginyltransferase, which mediates a specific protein degradation pathway and controls thermogenesis, regulates the stability of the TUG product. We conclude that insulin-stimulated TUG cleavage coordinates whole-body energy expenditure with glucose uptake, that this mechanism might contribute to the thermic effect of food and that its attenuation could promote obesity.
Collapse
Affiliation(s)
- Estifanos N Habtemichael
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Evelo Biosciences, Inc., Cambridge, MA, USA
| | - Don T Li
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | - João Paulo Camporez
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- University of São Paulo, São Paulo, Brazil
| | - Xavier O Westergaard
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Columbia University, New York, NY, USA
| | - Chloe I Sales
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Xinran Liu
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA
| | | | - Stephen G DeVries
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Hanbing Li
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Zhejiang University of Technology, Hangzhou, China
| | - Diana M Ruiz
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Kenny Y Wang
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Bhavesh S Sayal
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sofia González Zapata
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Pamela Dann
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Stacey N Brown
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sandro Hirabara
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Institute of Physical Activity Sciences and Sports, Cruzeiro do Sul University, São Paulo, Brazil
| | - Daniel F Vatner
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Leigh Goedeke
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - William Philbrick
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gerald I Shulman
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan S Bogan
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
45
|
Resveratrol Affects Insulin Signaling in Type 2 Diabetic Goto-Kakizaki Rats. Int J Mol Sci 2021; 22:ijms22052469. [PMID: 33671110 PMCID: PMC7957525 DOI: 10.3390/ijms22052469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
Resveratrol is a biologically active diphenolic compound exerting multiple beneficial effects in the organism, including anti-diabetic properties. This action is, however, not fully elucidated. In the present study, we examined effects of resveratrol on some parameters related to insulin signaling, and also on diabetes-associated dysregulation in Goto-Kakizaki (GK) rats with congenital type 2 diabetes. Resveratrol was given at the dose of 20 mg/kg b.w. for 10 weeks. It was shown that the expression and phosphorylation levels of insulin receptor in the skeletal muscle of GK rats were significantly decreased, compared with control animals. However, these changes were totally prevented by resveratrol. Liver expression of the insulin receptor was also reduced, but in this case, resveratrol was ineffective. Resveratrol was also demonstrated to significantly influence parameters of insulin binding (dissociation constant and binding capacity) in the skeletal muscle and liver. Moreover, it was shown that the expression levels of proteins related to intracellular glucose transport (GLUT4 and TUG) in adipose tissue of GK rats were significantly decreased. However, treatment with resveratrol completely abolished these changes. Resveratrol was found to induce normalization of TUG expression in the skeletal muscle. Blood levels of insulin and GIP were elevated, whereas proinsulin and GLP-1 diminished in GK rats. However, concentrations of these hormones were not affected by resveratrol. These results indicate that resveratrol partially ameliorates diabetes-associated dysregulation in GK rats. The most relevant finding covers the normalization of the insulin receptor expression in the skeletal muscle and also GLUT4 and TUG in adipose tissue.
Collapse
|
46
|
Complexin-2 redistributes to the membrane of muscle cells in response to insulin and contributes to GLUT4 translocation. Biochem J 2021; 478:407-422. [DOI: 10.1042/bcj20200542] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 12/11/2020] [Accepted: 01/04/2021] [Indexed: 11/17/2022]
Abstract
Insulin stimulates glucose uptake in muscle cells by rapidly redistributing vesicles containing GLUT4 glucose transporters from intracellular compartments to the plasma membrane (PM). GLUT4 vesicle fusion requires the formation of SNARE complexes between vesicular VAMP and PM syntaxin4 and SNAP23. SNARE accessory proteins usually regulate vesicle fusion processes. Complexins aide in neuro-secretory vesicle-membrane fusion by stabilizing trans-SNARE complexes but their participation in GLUT4 vesicle fusion is unknown. We report that complexin-2 is expressed and homogeneously distributed in L6 rat skeletal muscle cells. Upon insulin stimulation, a cohort of complexin-2 redistributes to the PM. Complexin-2 knockdown markedly inhibited GLUT4 translocation without affecting proximal insulin signalling of Akt/PKB phosphorylation and actin fiber remodelling. Similarly, complexin-2 overexpression decreased maximal GLUT4 translocation suggesting that the concentration of complexin-2 is finely tuned to vesicle fusion. These findings reveal an insulin-dependent regulation of GLUT4 insertion into the PM involving complexin-2.
Collapse
|
47
|
Counter-regulatory responses to Telfairia occidentalis-induced hypoglycaemia. Metabol Open 2020; 8:100065. [PMID: 33235989 PMCID: PMC7670218 DOI: 10.1016/j.metop.2020.100065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/25/2020] [Accepted: 10/29/2020] [Indexed: 12/27/2022] Open
Abstract
Background Telfairia occidentalis (TO) has many biological activities including blood glucose regulation. Thus, it is being used in the treatment of diabetes mellitus. TO has been shown to cause insulin-mediated hypoglycaemia, which leads to post-hypoglycaemic hyperglycaemia. However, the mechanism involved in the post-hypoglycaemic hyperglycaemia is still poorly understood. Objective This research was designed to determine the response of glucoregulatory hormones and enzymes to TO treatment. Methods Thirty-five male Wistar rats were divided into seven oral treatment groups (n = 5/group), which received either of 100 mg/kg or 200 mg/kg TO for 7-, 10- or 14 days. Results The 7-day treatment with TO significantly increased the levels of insulin, glucagon, and glucose-6-phosphatase (G6Pase) activity but decreased the levels of glucose, adrenaline, and glucokinase (GCK) activity. The 10-day treatment with 100 mg/kg TO increased glucose and decreased GCK activity while 200 mg/kg for the same duration increased glucose, insulin, GCK and G6Pase activities but reduced glucagon. The 14-day treatment with 100 mg/kg TO decreased glucose and glucagon but increased cortisol, while 200 mg/kg TO for same duration increased insulin, but reduced glucagon and GCK activity. Conclusion The TO's post-hypoglycaemic hyperglycaemia results from increased glucagon and G6Pase activity, and reduced GCK activity. Moreover, the glucagon response mainly depends on glucose rather than insulin.
Collapse
Key Words
- ANOVA, Analysis of Variance
- Avidin-HRP, Avidin-Horseradish Peroxidase
- Counter-regulatory hormones
- EGP, Endogenous glucose production
- ELISA, Enzyme-linked immunosorbent assay
- G6P, Glucose-6-phosphate
- G6PD, Glucose-6-phosphate dehydrogenase
- G6Pase, Glucose-6-phosphatase
- GCK, Glucokinase
- Glucoregulatory enzymes
- Insulin
- LDH, Lactate dehydrogenase
- LSD, Least Significance Difference
- NAD, Nicotinamide adenine dinucleotide
- NIH, National Institutes of Health
- Plasma glucose
- SEM, Standard error of mean
- SPSS, Statistical Package for the Social Sciences
- TO, Telfairia occidentalis
- Telfairia occidentalis
- cAMP, Cyclic adenosine monophosphate
Collapse
|
48
|
Gong YJ, Feng Y, Cao YY, Zhao J, Wu W, Zheng YY, Wu JR, Li X, Yang GZ, Zhou X. Huntingtin-associated protein 1 plays an essential role in the pathogenesis of type 2 diabetes by regulating the translocation of GLUT4 in mouse adipocytes. BMJ Open Diabetes Res Care 2020; 8:8/1/e001199. [PMID: 33060070 PMCID: PMC7566288 DOI: 10.1136/bmjdrc-2020-001199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 07/30/2020] [Accepted: 09/05/2020] [Indexed: 01/09/2023] Open
Abstract
OBJECTIVE Glucose disposal by insulin-responsive tissues maintains the body glucose homeostasis and insulin resistance leads to a risk of developing type 2 diabetes (T2DM). Insulin stimulates the translocation of glucose transporter isoform 4 (GLUT4) vesicles from intracellular compartments to the plasma membrane to facilitate glucose uptake. However, the underlying mechanisms of GLUT4 vesicle translocation are not well defined. Here we show the role of huntingtin-associated protein 1 (HAP1) in GLUT4 translocation in adipocytes and the pathogenesis of T2DM. RESEARCH DESIGN AND METHODS The parameters for glucose metabolism including body weight, glucose tolerance and insulin tolerance were assessed in wild-type (WT) and Hap1+/- mice. HAP1 protein expression was verified in adipose tissue. Hap1 mRNA and protein expression was monitored in adipose tissue of high-fat diet (HFD)-induced diabetic mice. Insulin-stimulated GLUT4 vesicle translocation and glucose uptake were detected using immunofluorescence techniques and quantified in primary adipocytes from Hap1-/- mice. The interaction between HAP1 and GLUT4 was assessed by immunofluorescence colocalization and co-immunoprecipitation in HEK293 cells and adipose tissue. The role of sortilin in HAP1 and GLUT4 interaction was approved by co-immunoprecipitation and RNA interference. RESULTS The expression of Hap1 mRNA and protein was detected in WT mouse adipose tissue and downregulated in adipose tissue of HFD-induced diabetic mice. Hap1+/- mice exhibited increased body weight, pronounced glucose tolerance and significant insulin intolerance compared with the WT mice. HAP1 colocalized with GLUT4 in mouse adipocytes and cotransfected HEK293 cells. Furthermore, the insulin-stimulated GLUT4 vesicle translocation and glucose uptake were defective in Hap1-/- adipocytes. Finally, sortilin mediated the interaction of HAP1 and GLUT4. CONCLUSIONS Our study showed that HAP1 formed a protein complex with GLUT4 and sortilin, and played a critical role in insulin-stimulated GLUT4 translocation in adipocytes. Its downregulation may contribute to the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Yan-Ju Gong
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Ying Feng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Yuan-Yuan Cao
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jia Zhao
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wu
- Institute of Biology, National Institute of Measurement and Testing Technology, Chengdu, Sichuan, China
| | - Ya-Yun Zheng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jia-Rui Wu
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xin Li
- Department of Pathophysiology, School of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Gui-Zhi Yang
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Xue Zhou
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
49
|
Astaxanthin Inhibits p70 S6 Kinase 1 Activity to Sensitize Insulin Signaling. Mar Drugs 2020; 18:md18100495. [PMID: 32998286 PMCID: PMC7600478 DOI: 10.3390/md18100495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/16/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022] Open
Abstract
Astaxanthin (AST) is a carotenoid with therapeutic values on hyperglycemia and diabetic complications. The mechanisms of action of AST remain incompletely understood. p70 S6 kinase 1 (S6K1) is a serine/threonine kinase that phosphorylates insulin receptor substrate 1 (IRS-1)S1101 and desensitizes the insulin receptor (IR). Our present study aims to determine if AST improves glucose metabolisms by targeting S6K1. Western blot analysis revealed that AST inhibited the phosphorylation of two S6K1 substrates, S6S235/236 and IRS-1S1101, but enhanced the phosphorylation of AKTT308, AKTS473, and S6K1T389 by feedback activation of the phosphatidylinositol-3 (PI-3) kinase in 3T3-L1 adipocytes and L6 myotubes. In vitro kinase assays revealed that AST inhibited S6K1 activity with an IC50 value of approximately 13.8 μM. AST increased insulin-induced IR tyrosine phosphorylation and IRS-1 binding to the p85 subunit of PI-3 kinase. Confocal microscopy revealed that AST increased the translocation of the glucose transporter 4 (GLUT4) to the plasma membrane in L6 cells. Glucose uptake assays using a fluorescent dye, 2-NBDG (2-N-(Nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose), revealed that AST increased glucose uptake in 3T3-L1 adipocytes and L6 myotubes under insulin resistance conditions. Our study identifies S6K1 as a previously unrecognized molecular target of AST and provides novel insights into the mechanisms of action of AST on IR sensitization.
Collapse
|
50
|
Zhang Y, Wang Y, Li X, Gu K, Li M, Zhang Y, Zhang Z, Wang S, Li Z. WSF-7 Inhibits Obesity-Mediated PPARγ Phosphorylation and Improves Insulin Sensitivity in 3T3-L1 Adipocytes. Biol Pharm Bull 2020; 43:526-532. [PMID: 32115511 DOI: 10.1248/bpb.b19-00986] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ), the molecular target for antidiabetic thiazolidinediones (TZDs), is a master regulator of preadipocyte differentiation and lipid metabolism. The adverse side effects of TZDs, arising from their potent agonistic activity, can be minimized by PPARγ partial agonists or PPARγ non-agonists without loss of insulin sensitization. In this study, we reported that WSF-7, a synthetic chemical derived from natural monoterpene α-pinene, is a partial PPARγ agonist. We found that WSF-7 binds directly to PPARγ. Activation of PPARγ by WSF-7 promotes adipogenesis, adiponectin oligomerization and insulin-induced glucose uptake. WSF-7 also inhibits obesity-mediated PPARγ phosphorylation at serine (Ser)273 and improves insulin sensitivity of 3T3-L1 adipocytes. Our study suggested that WSF-7 activates PPARγ transcription by a mechanism different from that of rosiglitazone or luteolin. Therefore, WSF-7 might be a potential therapeutic drug to treat type 2 diabetes.
Collapse
Affiliation(s)
- Yudian Zhang
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University
| | - Yunyun Wang
- College of Chemical Engineering, Nanjing Forestry University
| | - Xiaochuan Li
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University
| | - Kerui Gu
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University
| | - Mingxin Li
- College of Chemical Engineering, Nanjing Forestry University
| | - Yan Zhang
- College of Chemical Engineering, Nanjing Forestry University
| | - Zhijie Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences
| | - Shifa Wang
- College of Chemical Engineering, Nanjing Forestry University
| | - Zhen Li
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University
| |
Collapse
|