1
|
Ou Y, Zhao YL, Su H. Pancreatic β-Cells, Diabetes and Autophagy. Endocr Res 2024:1-16. [PMID: 39429147 DOI: 10.1080/07435800.2024.2413064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/23/2024] [Accepted: 08/18/2024] [Indexed: 10/22/2024]
Abstract
PURPOSE Pancreatic β-cells play a critical role in regulating plasma insulin levels and glucose metabolism balance, with their dysfunction being a key factor in the progression of diabetes. This review aims to explore the role of autophagy, a vital cellular self-maintenance process, in preserving pancreatic β-cell functionality and its implications in diabetes pathogenesis. METHODS We examine the current literature on the role of autophagy in β-cells, highlighting its function in maintaining cell structure, quantity, and function. The review also discusses the effects of both excessive and insufficient autophagy on β-cell dysfunction and glucose metabolism imbalance. Furthermore, we discuss potential therapeutic agents that modulate the autophagy pathway to influence β-cell function, providing insights into therapeutic strategies for diabetes management. RESULTS Autophagy acts as a self-protective mechanism within pancreatic β-cells, clearing damaged organelles and proteins to maintain cellular stability. Abnormal autophagy activity, either overactive or deficient, can disrupt β-cell function and glucose regulation, contributing to diabetes progression. CONCLUSION Autophagy plays a pivotal role in maintaining pancreatic β-cell function, and its dysregulation is implicated in the development of diabetes. Targeting the autophagy pathway offers potential therapeutic strategies for diabetes management, with agents that modulate autophagy showing promise in preserving β-cell function.
Collapse
Affiliation(s)
- Yang Ou
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, P.R. China
- Department of Endocrinology and Metabolism, First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, P.R. China
| | - Yan-Li Zhao
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan Province, China
| | - Heng Su
- Department of Endocrinology and Metabolism, First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, P.R. China
| |
Collapse
|
2
|
Pereira MJ, Mathioudaki A, Otero AG, Duvvuri PP, Vranic M, Sedigh A, Eriksson JW, Svensson MK. Renal sinus adipose tissue: exploratory study of metabolic features and transcriptome compared with omental and subcutaneous adipose tissue. Obesity (Silver Spring) 2024; 32:1870-1884. [PMID: 39210585 DOI: 10.1002/oby.24114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVE The objective was to study metabolic characteristics and transcriptome of renal sinus adipose tissue (RSAT) located around renal arteries and veins. METHODS Adipose tissue biopsies from RSAT, omental (OAT), and subcutaneous (SAT) depots were obtained from healthy kidney donors (20 female, 20 male). Adipocyte glucose uptake rate and cell size were measured, and gene expression analyses using transcriptomics were performed. RESULTS RSAT adipocytes were significantly smaller, with a higher basal glucose uptake rate, than adipocytes from SAT and OAT. Transcriptomic analyses revealed 29 differentially expressed genes between RSAT and OAT (RSAT: 23 lower, 6 higher) and 1214 differentially expressed genes between RSAT and SAT (RSAT: 859 lower, 355 higher). RSAT demonstrated molecular resemblance to OAT, both exhibiting lower metabolic gene expression and higher expression of immune-related pathways, including IL-17, TNFα, and NF-κB signaling than SAT. Weighted gene coexpression network analysis associated RSAT with immune response and nucleic acid transport processes. Despite its location near the renal hilum, RSAT closely resembled OAT and there was a lack of expression in the classical brown adipose tissue genes. Gene enrichment analyses suggest an inflammatory environment in RSAT compared with SAT and, to some extent, OAT. CONCLUSIONS The findings suggest specific RSAT functions that could impact renal function and, possibly, the development of renal and cardiometabolic disorders.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Argyri Mathioudaki
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Alicia G Otero
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Padma Priya Duvvuri
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Milica Vranic
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Amir Sedigh
- Department of Surgical Sciences, Transplantation Surgery, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetology and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria K Svensson
- Department of Medical Sciences, Renal Medicine, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| |
Collapse
|
3
|
Stanciu SM, Jinga M, Miricescu D, Stefani C, Nica RI, Stanescu-Spinu II, Vacaroiu IA, Greabu M, Nica S. mTOR Dysregulation, Insulin Resistance, and Hypertension. Biomedicines 2024; 12:1802. [PMID: 39200267 PMCID: PMC11351979 DOI: 10.3390/biomedicines12081802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/04/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Worldwide, diabetes mellitus (DM) and cardiovascular diseases (CVDs) represent serious health problems associated with unhealthy diet and sedentarism. Metabolic syndrome (MetS) is characterized by obesity, dyslipidemia, hyperglycemia, insulin resistance (IR) and hypertension. The mammalian target of rapamycin (mTOR) is a serine/threonine kinase with key roles in glucose and lipid metabolism, cell growth, survival and proliferation. mTOR hyperactivation disturbs glucose metabolism, leading to hyperglycemia and further to IR, with a higher incidence in the Western population. Metformin is one of the most used hypoglycemic drugs, with anti-inflammatory, antioxidant and antitumoral properties, having also the capacity to inhibit mTOR. mTOR inhibitors such as rapamycin and its analogs everolimus and temsirolimus block mTOR activity, decrease the levels of glucose and triglycerides, and reduce body weight. The link between mTOR dysregulation, IR, hypertension and mTOR inhibitors has not been fully described. Therefore, the main aim of this narrative review is to present the mechanism by which nutrients, proinflammatory cytokines, increased salt intake and renin-angiotensin-aldosterone system (RAAS) dysregulation induce mTOR overactivation, associated further with IR and hypertension development, and also mTOR inhibitors with higher potential to block the activity of this protein kinase.
Collapse
Affiliation(s)
- Silviu Marcel Stanciu
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania; (S.M.S.); (M.J.)
| | - Mariana Jinga
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania; (S.M.S.); (M.J.)
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Constantin Stefani
- Department of Family Medicine and Clinical Base, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania;
| | - Remus Iulian Nica
- Surgery Department, Central Military Emergency University Hospital, “Dr. Carol Davila”, 010825 Bucharest, Romania;
- Discipline of General Surgery, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanotari Blvd, 054474 Bucharest, Romania
| | - Iulia-Ioana Stanescu-Spinu
- Discipline of Physiology, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Department of Nephrology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | - Maria Greabu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania;
| | - Silvia Nica
- Emergency Discipline, University Hospital of Bucharest, 050098 Bucharest, Romania;
- Department of Emergency and First Aid, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Blvd, 050474 Bucharest, Romania
| |
Collapse
|
4
|
Li MY, Shen HH, Cao XY, Gao XX, Xu FY, Ha SY, Sun JS, Liu SP, Xie F, Li MQ. Targeting a mTOR/autophagy axis: a double-edged sword of rapamycin in spontaneous miscarriage. Biomed Pharmacother 2024; 177:116976. [PMID: 38906022 DOI: 10.1016/j.biopha.2024.116976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/02/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Immune dysfunction is a primary culprit behind spontaneous miscarriage (SM). To address this, immunosuppressive agents have emerged as a novel class of tocolytic drugs, modulating the maternal immune system's tolerance towards the embryo. Rapamycin (PubChem CID:5284616), a dual-purpose compound, functions as an immunosuppressive agent and triggers autophagy by targeting the mTOR pathway. Its efficacy in treating SM has garnered significant research interest in recent times. Autophagy, the cellular process of self-degradation and recycling, plays a pivotal role in numerous health conditions. Research indicates that autophagy is integral to endometrial decidualization, trophoblast invasion, and the proper functioning of decidual immune cells during a healthy pregnancy. Yet, in cases of SM, there is a dysregulation of the mTOR/autophagy axis in decidual stromal cells or immune cells at the maternal-fetal interface. Both in vitro and in vivo studies have highlighted the potential benefits of low-dose rapamycin in managing SM. However, given mTOR's critical role in energy metabolism, inhibiting it could potentially harm the pregnancy. Moreover, while low-dose rapamycin has been deemed safe for treating recurrent implant failure, its potential teratogenic effects remain uncertain due to insufficient data. In summary, rapamycin represents a double-edged sword in the treatment of SM, balancing its impact on autophagy and immune regulation. Further investigation is warranted to fully understand its implications.
Collapse
Affiliation(s)
- Meng-Ying Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Hui Shen
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Yan Cao
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Xiao Gao
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China
| | - Feng-Yuan Xu
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Si-Yao Ha
- Department of Obstetrics and Gynecology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510235, China
| | - Jian-Song Sun
- School of Life Science and Health Engineering, Jiangnan University, Wuxi 214122, People's Republic of China
| | - Song-Ping Liu
- Department of Obstetrics and Gynecology, Jinshan Hospital, Fudan University, Shanghai 201508, People's Republic of China.
| | - Feng Xie
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Shanghai Medical School, Fudan University, Shanghai 200080, People's Republic of China.
| | - Ming-Qing Li
- Institute of Obstetrics and Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China; Department of Gynecologic Endocrinology and Reproductive Immunology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, People's Republic of China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200030, People's Republic of China.
| |
Collapse
|
5
|
Vranic M, Ahmed F, Kristófi R, Hetty S, Mokhtari D, Svensson MK, Eriksson JW, Pereira MJ. Subcutaneous adipose tissue dopamine D2 receptor is increased in prediabetes and T2D. Endocrine 2024; 83:378-391. [PMID: 37752366 PMCID: PMC10850013 DOI: 10.1007/s12020-023-03525-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 09/28/2023]
Abstract
PURPOSE To evaluate the dopaminergic signaling in human adipose tissue in the context of obesity and type 2 diabetes (T2D) and potential direct implications in adipose tissue metabolism. METHODS mRNA and protein expression of dopamine receptors D1 and D2 (DRD1 and DRD2) were determined in subcutaneous adipose tissue from subjects without or with T2D and with different body weight, and correlated with markers of obesity, hyperglycemia, and insulin resistance. Glucose uptake and lipolysis were measured in adipocytes ex vivo following short-term exposure to dopamine, DRD1 receptor agonist (SKF81297), or DRD2 receptor agonist (bromocriptine). RESULTS DRD1 and DRD2 gene expression in subcutaneous adipose tissue correlated positively with clinical markers of insulin resistance (e.g. HOMA-IR, insulin, and triglycerides) and central obesity in subjects without T2D. Protein expression of DRD2 in subcutaneous adipose tissue, but not DRD1, is higher in subjects with impaired fasting glucose and T2D and correlated positively with hyperglycemia, HbA1c, and glucose AUC, independent of obesity status. DRD1 and DRD2 proteins were mainly expressed in adipocytes, compared to stromal vascular cells. Dopamine and dopaminergic agonists did not affect adipocyte glucose uptake ex vivo, but DRD1 and DRD2 agonist treatment inhibited isoproterenol-stimulated lipolysis. CONCLUSION The results suggest that protein expression of DRD2 in subcutaneous adipose tissue is up-regulated with hyperglycemia and T2D. Whether DRD2 protein levels contribute to T2D development or occur as a secondary compensatory mechanism needs further investigation. Additionally, dopamine receptor agonists inhibit adipocyte beta-adrenergic stimulation of lipolysis, which might contribute to the beneficial effects in lipid metabolism as observed in patients taking bromocriptine.
Collapse
Affiliation(s)
- Milica Vranic
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Fozia Ahmed
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Dariush Mokhtari
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria K Svensson
- Department of Medical Sciences, Renal Medicine, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
6
|
Benedet PO, Safikhan NS, Pereira MJ, Lum BM, Botezelli JD, Kuo CH, Wu HL, Craddock BP, Miller WT, Eriksson JW, Yue JTY, Conway EM. CD248 promotes insulin resistance by binding to the insulin receptor and dampening its insulin-induced autophosphorylation. EBioMedicine 2024; 99:104906. [PMID: 38061240 PMCID: PMC10750038 DOI: 10.1016/j.ebiom.2023.104906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/29/2023] Open
Abstract
BACKGROUND In spite of new treatments, the incidence of type 2 diabetes (T2D) and its morbidities continue to rise. The key feature of T2D is resistance of adipose tissue and other organs to insulin. Approaches to overcome insulin resistance are limited due to a poor understanding of the mechanisms and inaccessibility of drugs to relevant intracellular targets. We previously showed in mice and humans that CD248, a pre/adipocyte cell surface glycoprotein, acts as an adipose tissue sensor that mediates the transition from healthy to unhealthy adipose, thus promoting insulin resistance. METHODS Molecular mechanisms by which CD248 regulates insulin signaling were explored using in vivo insulin clamp studies and biochemical analyses of cells/tissues from CD248 knockout (KO) and wild-type (WT) mice with diet-induced insulin resistance. Findings were validated with human adipose tissue specimens. FINDINGS Genetic deletion of CD248 in mice, overcame diet-induced insulin resistance with improvements in glucose uptake and lipolysis in white adipose tissue depots, effects paralleled by increased adipose/adipocyte GLUT4, phosphorylated AKT and GSK3β, and reduced ATGL. The insulin resistance of the WT mice could be attributed to direct interaction of the extracellular domains of CD248 and the insulin receptor (IR), with CD248 acting to block insulin binding to the IR. This resulted in dampened insulin-mediated autophosphorylation of the IR, with reduced downstream signaling/activation of intracellular events necessary for glucose and lipid homeostasis. INTERPRETATION Our discovery of a cell-surface CD248-IR complex that is accessible to pharmacologic intervention, opens research avenues toward development of new agents to prevent/reverse insulin resistance. FUNDING Funded by Canadian Institutes of Health Research (CIHR), Natural Sciences and Engineering Research Council of Canada (NSERC), Canada Foundations for Innovation (CFI), the Swedish Diabetes Foundation, Family Ernfors Foundation and Novo Nordisk Foundation.
Collapse
Affiliation(s)
- Patricia O Benedet
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Nooshin S Safikhan
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Maria J Pereira
- Department of Medical Sciences, Clinical Diabetology & Metabolism, Uppsala University, Sweden
| | - Bryan M Lum
- Department of Physiology, Alberta Diabetes Institute and Group on Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - José Diego Botezelli
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada
| | - Cheng-Hsiang Kuo
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Hua-Lin Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Barbara P Craddock
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA; Veterans Affairs Medical Center, Northport, NY, USA
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetology & Metabolism, Uppsala University, Sweden
| | - Jessica T Y Yue
- Department of Physiology, Alberta Diabetes Institute and Group on Molecular and Cell Biology of Lipids, University of Alberta, Canada
| | - Edward M Conway
- Centre for Blood Research, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada; Departments of Medicine and Pathology and Laboratory Medicine, Life Sciences Institute, Faculty of Medicine, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
7
|
Du Y, Cai X. Therapeutic potential of natural compounds from herbs and nutraceuticals in spinal cord injury: Regulation of the mTOR signaling pathway. Biomed Pharmacother 2023; 163:114905. [PMID: 37207430 DOI: 10.1016/j.biopha.2023.114905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
Spinal cord injury (SCI) is a disease in which the spinal cord is subjected to various external forces that cause it to burst, shift, or, in severe cases, injure the spinal tissue, resulting in nerve injury. SCI includes not only acute primary injury but also delayed and persistent spinal tissue injury (i.e., secondary injury). The pathological changes post-SCI are complex, and effective clinical treatment strategies are lacking. The mammalian target of rapamycin (mTOR) coordinates the growth and metabolism of eukaryotic cells in response to various nutrients and growth factors. The mTOR signaling pathway has multiple roles in the pathogenesis of SCI. There is evidence for the beneficial effects of natural compounds and nutraceuticals that regulate the mTOR signaling pathways in a variety of diseases. Therefore, the effects of natural compounds on the pathogenesis of SCI were evaluated by a comprehensive review using electronic databases, such as PubMed, Web of Science, Scopus, and Medline, combined with our expertise in neuropathology. In particular, we reviewed the pathogenesis of SCI, including the importance of secondary nerve injury after the primary mechanical injury, the roles of the mTOR signaling pathways, and the beneficial effects and mechanisms of natural compounds that regulate the mTOR signaling pathway on pathological changes post-SCI, including effects on inflammation, neuronal apoptosis, autophagy, nerve regeneration, and other pathways. This recent research highlights the value of natural compounds in regulating the mTOR pathway, providing a basis for developing novel therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Yan Du
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China
| | - Xue Cai
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning, PR China.
| |
Collapse
|
8
|
Granata S, Mercuri S, Troise D, Gesualdo L, Stallone G, Zaza G. mTOR-inhibitors and post-transplant diabetes mellitus: a link still debated in kidney transplantation. Front Med (Lausanne) 2023; 10:1168967. [PMID: 37250653 PMCID: PMC10213242 DOI: 10.3389/fmed.2023.1168967] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023] Open
Abstract
The mammalian target of rapamycin inhibitors (mTOR-Is, Sirolimus, and Everolimus) are immunosuppressive drugs widely employed in kidney transplantation. Their main mechanism of action includes the inhibition of a serine/threonine kinase with a pivotal role in cellular metabolism and in various eukaryotic biological functions (including proteins and lipids synthesis, autophagy, cell survival, cytoskeleton organization, lipogenesis, and gluconeogenesis). Moreover, as well described, the inhibition of the mTOR pathway may also contribute to the development of the post-transplant diabetes mellitus (PTDM), a major clinical complication that may dramatically impact allograft survival (by accelerating the development of the chronic allograft damage) and increase the risk of severe systemic comorbidities. Several factors may contribute to this condition, but the reduction of the beta-cell mass, the impairment of the insulin secretion and resistance, and the induction of glucose intolerance may play a pivotal role. However, although the results of several in vitro and in animal models, the real impact of mTOR-Is on PTDM is still debated and the entire biological machinery is poorly recognized. Therefore, to better elucidate the impact of the mTOR-Is on the risk of PTDM in kidney transplant recipients and to potentially uncover future research topics (particularly for the clinical translational research), we decided to review the available literature evidence regarding this important clinical association. In our opinion, based on the published reports, we cannot draw any conclusion and PTDM remains a challenge. However, also in this case, the administration of the lowest possible dose of mTOR-I should also be recommended.
Collapse
Affiliation(s)
- Simona Granata
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Silvia Mercuri
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Dario Troise
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Loreto Gesualdo
- Renal, Dialysis and Transplantation Unit, Department of Precision and Regenerative Medicine and Ionian Area (DIMEPRE-J), University of Bari, Bari, Italy
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| | - Gianluigi Zaza
- Nephrology, Dialysis and Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
| |
Collapse
|
9
|
Mohammadi-Motlagh HR, Sadeghalvad M, Yavari N, Primavera R, Soltani S, Chetty S, Ganguly A, Regmi S, Fløyel T, Kaur S, Mirza AH, Thakor AS, Pociot F, Yarani R. β Cell and Autophagy: What Do We Know? Biomolecules 2023; 13:biom13040649. [PMID: 37189396 DOI: 10.3390/biom13040649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 05/17/2023] Open
Abstract
Pancreatic β cells are central to glycemic regulation through insulin production. Studies show autophagy as an essential process in β cell function and fate. Autophagy is a catabolic cellular process that regulates cell homeostasis by recycling surplus or damaged cell components. Impaired autophagy results in β cell loss of function and apoptosis and, as a result, diabetes initiation and progress. It has been shown that in response to endoplasmic reticulum stress, inflammation, and high metabolic demands, autophagy affects β cell function, insulin synthesis, and secretion. This review highlights recent evidence regarding how autophagy can affect β cells' fate in the pathogenesis of diabetes. Furthermore, we discuss the role of important intrinsic and extrinsic autophagy modulators, which can lead to β cell failure.
Collapse
Affiliation(s)
- Hamid-Reza Mohammadi-Motlagh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 67155-1616, Iran
| | - Mona Sadeghalvad
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1416634793, Iran
| | - Niloofar Yavari
- Department of Cellular and Molecular Medicine, The Panum Institute, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Rosita Primavera
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Setareh Soltani
- Clinical Research Development Center, Taleghani and Imam Ali Hospital, Kermanshah University of Medical Sciences, Kermanshah 67145-1673, Iran
| | - Shashank Chetty
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Abantika Ganguly
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Shobha Regmi
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Tina Fløyel
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Simranjeet Kaur
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| | - Aashiq H Mirza
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Department of Pharmacology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Avnesh S Thakor
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
- Institute for Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Reza Yarani
- Interventional Regenerative Innovation at Stanford (IRIS), Department of Radiology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, 2730 Herlev, Denmark
| |
Collapse
|
10
|
A mixed blessing for liver transplantation patients - Rapamycin. Hepatobiliary Pancreat Dis Int 2023; 22:14-21. [PMID: 36328894 DOI: 10.1016/j.hbpd.2022.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/14/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Liver transplantation (LT) is an effective treatment option for end-stage liver disease. Mammalian target of rapamycin (mTOR) inhibitors, such as rapamycin, are widely used post LT. DATA SOURCES In this review, we focused on the anti-cancer activities and metabolic side effects of rapamycin after LT. The literature available on PubMed for the period of January 1999-September 2022 was reviewed. The key words were rapamycin, sirolimus, liver transplantation, hepatocellular carcinoma, diabetes, and lipid metabolism disorder. RESULTS Rapamycin has shown excellent effects and is safer than other immunosuppressive regimens. It has exhibited excellent anti-cancer activity and has the potential in preventing hepatocellular carcinoma (HCC) recurrence post LT. Rapamycin is closely related to two long-term complications after LT, diabetes and lipid metabolism disorders. CONCLUSIONS Rapamycin prevents HCC recurrence post LT in some patients, but it also induces metabolic disorders. Reasonable use of rapamycin benefits the liver recipients.
Collapse
|
11
|
Interleukin-33 inhibits glucose uptake in human adipocytes and its expression in adipose tissue is elevated in insulin resistance and type 2 diabetes. Cytokine 2023; 161:156080. [PMID: 36368230 DOI: 10.1016/j.cyto.2022.156080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/05/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Interleukin-33 (IL-33) is associated with obesity-related inflammation. We aim to investigate IL-33 expression in subcutaneous adipose tissue (SAT) in type 2 diabetes (T2D) subjects and its effects on human adipocyte glucose uptake. METHODS Expression of IL-33 was analysed in SAT from cohort studies including subjects with and without obesity and T2D and correlated with insulin resistance and obesity markers. Magnetic resonance imaging (MRI) of tissue fat volumes was performed. We investigated the effects of IL-33 treatment on ex vivo adipocyte glucose uptake. RESULTS T2D subjects had higher IL-33 gene and protein expression in SAT than the control subjects. IL-33 mRNA expression was positively correlated with markers of dysglycemia (e.g. HbA1c), insulin resistance (e.g. HOMA-IR) and adiposity (BMI, visceral adipose tissue volume, liver and pancreas fat %). In multiple linear regression analyses, insulin resistance and T2D status were the strongest predictors of IL-33, independent of BMI. IL-33 mRNA expression was negatively correlated with expression of genes regulating adipocyte glucose uptake, lipid storage, and adipogenesis (e.g.glucose transporter 1 and 4 (GLUT1/4), fatty acid binding protein 4 (FABP4), and PPARG). Additionally, incubation of SAT with IL-33 reduced adipocyte glucose uptake and GLUT4 gene and protein expression. CONCLUSIONS Our findings suggest that T2D subjects have higher IL-33 gene and protein expressionin SATthan control subjects, which is associated with insulin resistance and reduced gene expression of lipid storage and adipogenesis markers. IL-33 may reduce adipocyte glucose uptake. This opens up a potential pharmacological route for reversing insulin resistance in T2D and prediabetes.
Collapse
|
12
|
Castro‐Guarda M, Arancibia Y, Chipón C, Matamala C, Oyarzo P, Vargas G, Reyes A, Salas M, Morera FJ, Zambrano A. Metabolic changes induced by DNA damage in Ramos cells: exploring the role of mTORC1 complex. FEBS Open Bio 2022; 12:1509-1522. [PMID: 35538662 PMCID: PMC9340868 DOI: 10.1002/2211-5463.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 02/26/2022] [Accepted: 05/09/2022] [Indexed: 11/09/2022] Open
Abstract
DNA damage induces the activation of many different signals associated with repair or cell death, but it is also connected with physiological events, such as adult neurogenesis and B-cell differentiation. DNA damage induces different signaling pathways, some of them linked to important metabolic changes. The mTORC1 pathway has a central role in the regulation of growth processes and cell division in response to environmental changes and also controls protein synthesis, lipid biogenesis, nucleotide synthesis, and expression of glycolytic genes. Here, we report that double-strand breaks induced with etoposide affect the expression of genes encoding different enzymes associated with specific metabolic pathways in Ramos cells. We also analyzed the role of mTOR signaling, demonstrating that double-strand breaks induce downregulation of mTOR signaling. Specific inhibition of mTORC1 using rapamycin also induced changes in the expression of metabolic genes. Finally, we demonstrated that DNA damage and rapamycin can regulate glucose uptake. In summary, our findings show that etoposide and rapamycin affect the expression of metabolic genes as well as apoptotic and proliferation markers in Ramos cells, increasing our understanding of cancer metabolism.
Collapse
Affiliation(s)
- Marcos Castro‐Guarda
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Yennyfer Arancibia
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Carina Chipón
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Christofer Matamala
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Paola Oyarzo
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Gabriela Vargas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Alejandro Reyes
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Universidad Austral de ChileCoyhaiqueChile
| | - Mónica Salas
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
| | - Francisco J. Morera
- Facultad de Ciencias VeterinariasInstituto de Farmacología y MorfofisiologíaUniversidad Austral de ChileValdiviaChile
| | - Angara Zambrano
- Facultad de CienciasInstituto de Bioquímica y MicrobiologíaUniversidad Austral de ChileValdiviaChile
- Center for Interdisciplinary Studies on the Nervous System (CISNe)Universidad Austral de ChileValdiviaChile
| |
Collapse
|
13
|
Su R, Wei X, Wei Q, Lu D, Lin Z, Wang S, Shao C, Xu X. Extrahepatic organs in the development of non-alcoholic fatty liver disease in liver transplant patients. Hepatobiliary Surg Nutr 2022; 11:400-411. [PMID: 35693397 PMCID: PMC9186206 DOI: 10.21037/hbsn-20-568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/23/2020] [Indexed: 08/30/2023]
Abstract
BACKGROUND AND OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is highly prevalent in patients who undergo liver transplantation (LT). Whereas there is huge data on NAFLD, little is known about NAFLD in LT. In this review, we aim to explore extrahepatic organs and their potential mechanisms in the development of NAFLD in LT patients and discuss current limitations in preclinical and clinical scenarios with suggestions for future study. METHODS The following keywords, such as NAFLD, NASH, liver transplant, therapy, pathogenesis and biomarkers, were set for literature retrieval. The articles which were published articles in English till 25th June 2020 in PubMed database were included, and there is no limit for the study design type. KEY CONTENT AND FINDINGS Following LT, there are significant shifts in the microbiota and farnesoid X receptor may be a potential therapeutic target for NAFLD in LT settings. The roles of probiotics and diet on NALFD remain inconclusive in LT background. Nevertheless, the adipokines and cytokines disorder and local insulin resistance of adipose tissue may contribute to NAFLD process. Bariatric surgeries are promising in controlling de novo and recurrent NAFLD with significant reduction in abdominal adipose tissue, despite the optimal timing is inconclusive in LT cases. Furthermore, circumstantial evidence indicates that miRNA-33a may function as a mediator bridging sarcopenia and NAFLD of post-LT. β-Hydroxy-β-Methyl-Butyrate treatment could improve muscle status in graft recipients and shows protective potential for NAFLD in LT settings. CONCLUSIONS Gut, adipose tissue and muscle are intricately intertwined in promoting NAFLD in LT cases. Further animal studies are needed to deepen our understanding of mechanisms in multi-organ crosstalk. High quality clinical trials are warrant for making guidelines and developing management strategies on NAFLD after LT.
Collapse
Affiliation(s)
- Renyi Su
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Wei
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Di Lu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Zuyuan Lin
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Shuo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Shu Lan Hospital, Shu Lan International Medical College of Zhejiang Shuren University, Hangzhou, China
| | - Chuxiao Shao
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Lishui Hospital, Zhejiang University School of Medicine, Lishui, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| |
Collapse
|
14
|
Pereira MJ, Vranic M, Kamble PG, Jernow H, Kristófi R, Holbikova E, Skrtic S, Kullberg J, Svensson MK, Hetty S, Eriksson JW. CDKN2C expression in adipose tissue is reduced in type II diabetes and central obesity: impact on adipocyte differentiation and lipid storage? Transl Res 2022; 242:105-121. [PMID: 34896253 DOI: 10.1016/j.trsl.2021.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/16/2021] [Accepted: 12/06/2021] [Indexed: 10/19/2022]
Abstract
CDKN2C/p18 (Cyclin-Dependent Kinase Inhibitor 2C) is a cell growth regulator that controls cell cycle progression and has previously been associated with increased risk for type II diabetes (T2D) and reduced peripheral adipose tissue (AT) storage capacity. This study explored the role of CDKN2C in AT lipid and glucose metabolism in T2D. Expression of CDKN2C and other genes was analyzed by transcriptomics, or real-time PCR in subcutaneous AT (SAT) samples obtained from T2D and control subjects matched for sex, age and BMI and also in paired SAT and omental AT (OAT) samples. Functional studies included adipocyte glucose uptake and lipolysis rates. CRISPR/Cas9 CDKN2C gene knockdown was performed in human preadipocytes to assess adipogenesis. CDKN2C mRNA expression in SAT and OAT was reduced in T2D and obese subjects compared to controls. CDKN2C expression in SAT was inversely correlated with measures of hyperglycemia, insulin resistance and visceral adiposity and positively correlated with expression of genes in several metabolic pathways, including insulin signaling and fatty acid and carbohydrate metabolism. CDKN2C protein was mainly expressed in adipocytes compared to stromal vascular cells, and its gene and protein expression was up-regulated during adipocyte differentiation. Knockdown of CDKN2C did not affect the percentage of differentiating cells compared to wild type cultures. However, CDKN2C knockdown cultures had significantly lower expression of differentiation markers CEBPA, ADIPOQ and FASN and transiently reduced lipid accumulation per adipocyte during differentiation. Our findings suggest that adipose CDKN2C expression might be reduced as a consequence of insulin resistance and obesity, and this can further contribute to impairment of SAT lipid storage.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Milica Vranic
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Prasad G Kamble
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Henning Jernow
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Robin Kristófi
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Ema Holbikova
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- Innovation Strategies & External Liaison, Pharmaceutical Technologies & Development, AstraZeneca, Gothenburg, Sweden; Institute of Medicine at Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Joel Kullberg
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria K Svensson
- Department of Medical Sciences, Renal Medicine, Uppsala University, Uppsala, Sweden
| | - Susanne Hetty
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
PINHO ARYANEC, BURGEIRO ANA, PEREIRA MARIAJOÃO, CARVALHO EUGENIA. Drug-induced metabolic alterations in adipose tissue - with an emphasis in epicardial adipose tissue. AN ACAD BRAS CIENC 2022. [DOI: 10.1590/0001-3765202220201819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Affiliation(s)
| | | | | | - EUGENIA CARVALHO
- University of Coimbra, Portugal; University of Coimbra, Portugal; APDP-Portuguese Diabetes Association, Portugal
| |
Collapse
|
16
|
Yang G, Francis D, Krycer JR, Larance M, Zhang Z, Novotny CJ, Diaz-Vegas A, Shokat KM, James DE. Dissecting the biology of mTORC1 beyond rapamycin. Sci Signal 2021; 14:eabe0161. [PMID: 34546793 DOI: 10.1126/scisignal.abe0161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Guang Yang
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Deanne Francis
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - James R Krycer
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Mark Larance
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Ziyang Zhang
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - Chris J Novotny
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - Alexis Diaz-Vegas
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia
| | - Kevan M Shokat
- Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, 600 16th Street, San Francisco, CA 94143, USA
| | - David E James
- University of Sydney, School of life and Environmental Sciences, Charles Perkins Centre, Sydney, New South Wales 2006, Australia.,University of Sydney, Sydney Medical School, Sydney, New South Wales 2006, Australia
| |
Collapse
|
17
|
Querfurth H, Lee HK. Mammalian/mechanistic target of rapamycin (mTOR) complexes in neurodegeneration. Mol Neurodegener 2021; 16:44. [PMID: 34215308 PMCID: PMC8252260 DOI: 10.1186/s13024-021-00428-5] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/01/2021] [Indexed: 12/12/2022] Open
Abstract
Novel targets to arrest neurodegeneration in several dementing conditions involving misfolded protein accumulations may be found in the diverse signaling pathways of the Mammalian/mechanistic target of rapamycin (mTOR). As a nutrient sensor, mTOR has important homeostatic functions to regulate energy metabolism and support neuronal growth and plasticity. However, in Alzheimer's disease (AD), mTOR alternately plays important pathogenic roles by inhibiting both insulin signaling and autophagic removal of β-amyloid (Aβ) and phospho-tau (ptau) aggregates. It also plays a role in the cerebrovascular dysfunction of AD. mTOR is a serine/threonine kinase residing at the core in either of two multiprotein complexes termed mTORC1 and mTORC2. Recent data suggest that their balanced actions also have implications for Parkinson's disease (PD) and Huntington's disease (HD), Frontotemporal dementia (FTD) and Amyotrophic Lateral Sclerosis (ALS). Beyond rapamycin; an mTOR inhibitor, there are rapalogs having greater tolerability and micro delivery modes, that hold promise in arresting these age dependent conditions.
Collapse
Affiliation(s)
- Henry Querfurth
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA.
| | - Han-Kyu Lee
- Department of Neurology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Chevallier E, Jouve T, Rostaing L, Malvezzi P, Noble J. pre-existing diabetes and PTDM in kidney transplant recipients: how to handle immunosuppression. Expert Rev Clin Pharmacol 2020; 14:55-66. [PMID: 33196346 DOI: 10.1080/17512433.2021.1851596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Preexisting diabetes (PD) and post-transplant diabetes mellitus (PTDM) are common and severe comorbidities posttransplantation. The immunosuppressive regimens are modifiable risk factors. AREAS COVERED We reviewed Pubmed and Cochrane database and we summarize the mechanisms and impacts of available immunosuppressive treatments on the risk of PD and PTDM. We also assess the possible management of these drugs to improve glycemic parameters while considering risks inherent in transplantation. EXPERT OPINION PD i) increases the risk of sepsis, ii) is an independent risk factor for infection-related mortality, and iii) increases acute rejection risk. Regarding PTDM development i) immunosuppressive strategies without corticosteroids significantly reduce the risk but the price may be a higher incidence of rejection; ii) minimization or rapid withdrawal of steroids are two valuable approaches; iii) the diabetogenic role of calcineurin inhibitors(CNIs) is also well-described and is more important for tacrolimus than for cyclosporine. Reducing tacrolimus-exposure may improve glycemic parameters but also has a higher risk of rejection. PTDM risk is higher in patients that receive sirolimus compared to mycophenolate mofetil. Finally, conversion from CNIs to belatacept may offer the best benefits to PTDM-recipients in terms of glycemic parameters, graft and patient-outcomes.
Collapse
Affiliation(s)
- Eloi Chevallier
- Service De Néphrologie, Hémodialyse, Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes , Grenoble, France
| | - Thomas Jouve
- Service De Néphrologie, Hémodialyse, Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes , Grenoble, France.,Université Grenoble Alpes , Grenoble, France
| | - Lionel Rostaing
- Service De Néphrologie, Hémodialyse, Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes , Grenoble, France.,Université Grenoble Alpes , Grenoble, France
| | - Paolo Malvezzi
- Service De Néphrologie, Hémodialyse, Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes , Grenoble, France
| | - Johan Noble
- Service De Néphrologie, Hémodialyse, Aphérèses Et Transplantation Rénale, CHU Grenoble-Alpes , Grenoble, France
| |
Collapse
|
19
|
Pereira MJ, Thombare K, Sarsenbayeva A, Kamble PG, Almby K, Lundqvist M, Eriksson JW. Direct effects of glucagon on glucose uptake and lipolysis in human adipocytes. Mol Cell Endocrinol 2020; 503:110696. [PMID: 31891768 DOI: 10.1016/j.mce.2019.110696] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/25/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
We aim to investigate the expression of the glucagon receptor (GCGR) in human adipose tissue, and the impact of glucagon in glucose uptake and lipolysis in human adipocytes. GCGR gene expression in human subcutaneous and visceral adipose tissue was demonstrated, albeit at low levels and with an inter-individual variation. Furthermore, GCGR expression was not significantly different between subjects with T2D and matched controls, and we found no significant association with BMI. Glucagon only at a supra-physiological concentration (10-100 nM) significantly increased basal and insulin-stimulated glucose uptake by up to 1.5-fold. Also, glucagon (0.01 and 1 nM) dose-dependently increased basal and isoproterenol-stimulated lipolysis up to 3.7- and 1.7-fold, respectively, compared to control. In addition, glucagon did not change insulin sensitivity to stimulate glucose uptake or inhibit lipolysis. In conclusion, we show that the GCGR gene is expressed at low levels in human adipose tissue, and glucagon at high concentrations can increase both glucose uptake and lipolysis in human adipocytes. Taken together, our data suggest that glucagon at physiological levels has minor direct effects on the regulation of adipocyte metabolism, but does not antagonize the insulin effect to stimulate glucose uptake and inhibit lipolysis in human adipocytes.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Ketan Thombare
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Prasad G Kamble
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Kristina Almby
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Martin Lundqvist
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
20
|
Zafar MI. Suitability of APINCH high-risk medications use in diabetes mellitus. Eur J Pharmacol 2020; 867:172845. [DOI: 10.1016/j.ejphar.2019.172845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
|
21
|
Edén D, Panagiotou G, Mokhtari D, Eriksson JW, Åberg M, Siegbahn A. Adipocytes express tissue factor and FVII and are procoagulant in a TF/FVIIa-dependent manner. Ups J Med Sci 2019; 124:158-167. [PMID: 31407948 PMCID: PMC6758637 DOI: 10.1080/03009734.2019.1645248] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Background: Tissue factor (TF) combined with its ligand FVII initiates blood coagulation and intracellular signaling. Obese and type 2 diabetic subjects have increased TF expression in their adipose tissue and an increased risk for thrombotic complications. Here we address the role of TF/FVII on adipocyte functions. Materials and methods: Subcutaneous fat was obtained by means of needle aspiration from healthy volunteers, and adipocytes were isolated after collagenase digestion. 3T3-L1 fibroblasts kept in culture were differentiated into adipocytes by addition of IBMX, dexamethasone, rosiglitazone, and insulin to the media. Proteins and mRNA were analyzed by western blot and RT-PCR. Coagulation activity was determined by a colorimetric FX-assay. Lipolysis was measured as free glycerol using a colorimetric method. Glucose uptake was evaluated by scintillation counting of D-[U-14C] glucose. Results: In isolated human primary adipocytes we found expression of TF and FVII. TF expression was confirmed in 3T3-L1 adipocytes, and both cell types were found to be procoagulant in a TF/FVIIa-dependent manner. FXa was generated without FVIIa added to the coagulation assay, and active site-inhibited FVIIa blocked FXa formation, supporting our finding of FVII production by human primary adipocytes. There was no evidence for a role of TF in either lipolysis or glucose uptake in our experimental settings. Conclusion: Human primary adipocytes express active TF and FVII, and the TF/FVIIa complex formed on the adipocyte surface can activate substrate FX. Whether the TF/FVIIa complex conveys signaling pathways leading to biological functions and has any biological activity in adipocytes beyond coagulation remains to be elucidated.
Collapse
Affiliation(s)
- Desirée Edén
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Grigorios Panagiotou
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Dariush Mokhtari
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W. Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Mikael Åberg
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
- CONTACT Agneta Siegbahn Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| |
Collapse
|
22
|
Katsogiannos P, Kamble PG, Boersma GJ, Karlsson FA, Lundkvist P, Sundbom M, Pereira MJ, Eriksson JW. Early Changes in Adipose Tissue Morphology, Gene Expression, and Metabolism After RYGB in Patients With Obesity and T2D. J Clin Endocrinol Metab 2019; 104:2601-2613. [PMID: 30689903 DOI: 10.1210/jc.2018-02165] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 01/18/2019] [Indexed: 02/13/2023]
Abstract
CONTEXT Roux-en-Y gastric bypass (RYGB) surgery effectively prevents or treats type 2 diabetes (T2D). Adipose tissue (AT) mechanisms may be of importance. OBJECTIVE To assess the relationship between early changes in whole-body and AT metabolism in surgically treated patients with T2D. DESIGN AND SETTING A randomized single-center study. PATIENTS Nineteen patients with T2D with body mass index 30 to 45 kg/m2. INTERVENTIONS Thirteen patients were assessed at baseline and 4 and 24 weeks after RYGB (preceded by a 4-week low-calorie diet) and compared with 6 control patients continuing standard medical treatment: oral glucose tolerance test, subcutaneous AT biopsies for gene expression, adipocyte size, glucose uptake, lipolysis, and insulin action. RESULTS At 4 and 24 weeks post-RYGB, all patients but one had stopped diabetes medication. Fasting glucose, HbA1c, and insulin levels decreased and the Matsuda index increased compared with baseline (P < 0.01 for all), indicating improved whole-body insulin sensitivity. Mean adipocyte size significantly reduced, more at 4 than at 24 weeks; at 4 weeks, glucose uptake per adipocyte was lowered, and isoproterenol-stimulated lipolysis tended to increase, whereas the fold insulin effects on glucose uptake and lipolysis were unchanged. Expression of genes involved in fatty acid oxidation, CPT1b and adiponectin, was increased at 4 weeks, whereas leptin and E2F1 (involved in cell proliferation) were reduced (P < 0.05 for all). CONCLUSION Glycemic control and in vivo insulin sensitivity improved 4 weeks after RYGB, but adipocyte insulin sensitivity did not change despite a marked reduction in adipocyte size. Thus, mechanisms for a rapid improvement of T2D after RYGB may occur mainly in other tissues than adipose.
Collapse
MESH Headings
- Adipocytes/metabolism
- Adult
- Biopsy
- Blood Glucose/analysis
- Blood Glucose/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/etiology
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/surgery
- Female
- Follow-Up Studies
- Gastric Bypass
- Humans
- Insulin/metabolism
- Insulin Resistance
- Male
- Middle Aged
- Obesity, Morbid/blood
- Obesity, Morbid/complications
- Obesity, Morbid/metabolism
- Obesity, Morbid/surgery
- Subcutaneous Fat, Abdominal/cytology
- Subcutaneous Fat, Abdominal/metabolism
- Subcutaneous Fat, Abdominal/pathology
- Treatment Outcome
Collapse
Affiliation(s)
| | - Prasad G Kamble
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Gretha J Boersma
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Per Lundkvist
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Chronic AICAR treatment prevents metabolic changes in cardiomyocytes exposed to free fatty acids. Pflugers Arch 2019; 471:1219-1234. [PMID: 31152240 DOI: 10.1007/s00424-019-02285-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/27/2019] [Accepted: 05/15/2019] [Indexed: 01/09/2023]
Abstract
The stimulation of glucose transport by metabolic stress is an important determinant of myocardial susceptibility to ischemia and reperfusion injury. Stimulation of glucose transport is markedly impaired in cardiomyocytes chronically exposed to excess free fatty acids (FFA), as occurs in vivo in type 2 diabetes. To determine whether chronic low-grade activation of AMP-activated kinase (AMPK) improves substrate metabolism in cardiomyocytes exposed to FFA, isolated cultured cardiomyocytes were exposed for 7 days to FFA ± the AMPK agonist 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR). Glucose transport and glycolysis were then measured during acute metabolic stress provoked by oligomycin. Chronic treatment with AICAR improved basal and oligomycin-stimulated glucose transport in FFA-exposed but not in control cardiomyocytes. Similarly, basal and oligomycin-stimulated glycolysis was reduced in FFA-exposed cardiomyocytes but restored by chronic AICAR treatment. Conversely, fatty acid oxidation was increased in FFA-exposed cardiomyocytes and reduced by chronic AICAR treatment. Chronic AICAR treatment induced in FFA-exposed cardiomyocytes the biogenesis of numerous lipid droplets. Curiously, whereas acute treatment of cardiomyocytes with AICAR increased phosphorylation of the AMPKα subunit on T172, a classical marker of AMPK activation, chronic AICAR treatment almost completely obliterated T172 phosphorylation. However, phosphorylation of the AMPK target protein raptor on S792 was reduced in FFA-exposed cardiomyocytes but restored by AICAR treatment. In conclusion, chronic AICAR treatment induces a metabolic shift in FFA-exposed cardiomyocytes, characterized by improved glucose transport and glycolysis and redirection of fatty acids towards neutral storage. Such metabolic changes in vivo could protect the hearts of patients with type 2 diabetes against ischemia-reperfusion injury.
Collapse
|
24
|
Brewer PD, Romenskaia I, Mastick CC. A high-throughput chemical-genetics screen in murine adipocytes identifies insulin-regulatory pathways. J Biol Chem 2018; 294:4103-4118. [PMID: 30591588 DOI: 10.1074/jbc.ra118.006986] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Pathways linking activation of the insulin receptor to downstream targets of insulin have traditionally been studied using a candidate gene approach. To elucidate additional pathways regulating insulin activity, we performed a forward chemical-genetics screen based on translocation of a glucose transporter 4 (Glut4) reporter expressed in murine 3T3-L1 adipocytes. To identify compounds with known targets, we screened drug-repurposing and natural product libraries. We identified, confirmed, and validated 64 activators and 65 inhibitors that acutely increase or rapidly decrease cell-surface Glut4 in adipocytes stimulated with submaximal insulin concentrations. These agents were grouped by target, chemical class, and mechanism of action. All groups contained multiple hits from a single drug class, and several comprised multiple structurally unrelated hits for a single target. Targets include the β-adrenergic and adenosine receptors. Agonists of these receptors increased and inverse agonists/antagonists decreased cell-surface Glut4 independently of insulin. Additional activators include insulin sensitizers (thiazolidinediones), insulin mimetics, dis-inhibitors (the mTORC1 inhibitor rapamycin), cardiotonic steroids (the Na+/K+-ATPase inhibitor ouabain), and corticosteroids (dexamethasone). Inhibitors include heterocyclic amines (tricyclic antidepressants) and 21 natural product supplements and herbal extracts. Mechanisms of action include effects on Glut4 trafficking, signal transduction, inhibition of protein synthesis, and dissipation of proton gradients. Two pathways that acutely regulate Glut4 translocation were discovered: de novo protein synthesis and endocytic acidification. The mechanism of action of additional classes of activators (tanshinones, dalbergiones, and coumarins) and inhibitors (flavonoids and resveratrol) remains to be determined. These tools are among the most sensitive, responsive, and reproducible insulin-activity assays described to date.
Collapse
Affiliation(s)
- Paul Duffield Brewer
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| | - Irina Romenskaia
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| | - Cynthia Corley Mastick
- From the Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, Nevada 89557
| |
Collapse
|
25
|
Therapeutic Use of mTOR Inhibitors in Renal Diseases: Advances, Drawbacks, and Challenges. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3693625. [PMID: 30510618 PMCID: PMC6231362 DOI: 10.1155/2018/3693625] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 09/07/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
The mammalian (or mechanistic) target of rapamycin (mTOR) pathway has a key role in the regulation of a variety of biological processes pivotal for cellular life, aging, and death. Impaired activity of mTOR complexes (mTORC1/mTORC2), particularly mTORC1 overactivation, has been implicated in a plethora of age-related disorders, including human renal diseases. Since the discovery of rapamycin (or sirolimus), more than four decades ago, advances in our understanding of how mTOR participates in renal physiological and pathological mechanisms have grown exponentially, due to both preclinical studies in animal models with genetic modification of some mTOR components as well as due to evidence coming from the clinical experience. The main clinical indication of rapamycin is as immunosuppressive therapy for the prevention of allograft rejection, namely, in renal transplantation. However, considering the central participation of mTOR in the pathogenesis of other renal disorders, the use of rapamycin and its analogs meanwhile developed (rapalogues) everolimus and temsirolimus has been viewed as a promising pharmacological strategy. This article critically reviews the use of mTOR inhibitors in renal diseases. Firstly, we briefly overview the mTOR components and signaling as well as the pharmacological armamentarium targeting the mTOR pathway currently available or in the research and development stages. Thereafter, we revisit the mTOR pathway in renal physiology to conclude with the advances, drawbacks, and challenges regarding the use of mTOR inhibitors, in a translational perspective, in four classes of renal diseases: kidney transplantation, polycystic kidney diseases, renal carcinomas, and diabetic nephropathy.
Collapse
|
26
|
Bouillet B, Buffier P, Smati S, Archambeaud F, Cariou B, Vergès B. Expert opinion on the metabolic complications of mTOR inhibitors. ANNALES D'ENDOCRINOLOGIE 2018; 79:583-590. [PMID: 30144939 DOI: 10.1016/j.ando.2018.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Using mTOR inhibitors (mTORi) as anticancer drugs led to hyperglycemia (12-50%) and hyperlipidemia (7-73%) in phase-III trials. These high rates require adapted treatment in cancer patients. Before initiating mTORi treatment, lipid profile screening should be systematic, with fasting glucose assay in non-diabetic patients and HbA1C in diabetic patients. After initiation, lipid profile monitoring should be systematic, with fasting glucose assay in non-diabetic patients, every 2 weeks for the first month and then monthly. The HbA1C target is≤8%, before and after treatment initiation in known diabetic patients and in case of onset of diabetes under mTORi. LDL-cholesterol targets should be adapted to general health status and cardiovascular and oncologic prognosis. If treatment is indicated, pravastatin should be prescribed in first line; atorvastatin and simvastatin are contraindicated. Fenofibrate should be prescribed for hypertriglyceridemia>5g/l resisting dietary measures adapted to oncologic status. In non-controllable hypertriglyceridemia exceeding 10g/l, mTORi treatment should be interrupted and specialist opinion should be sought.
Collapse
Affiliation(s)
- Benjamin Bouillet
- Service d'endocrinologie, diabétologie, maladies métaboliques, CHU de Dijon, 2, boulevard du Maréchal-de-Lattre, BP 77908, 21000 Dijon, France; Unité Inserm, LNC-UMR 1231, université de Bourgogne, Dijon, France.
| | - Perrine Buffier
- Service d'endocrinologie, diabétologie, maladies métaboliques, CHU de Dijon, 2, boulevard du Maréchal-de-Lattre, BP 77908, 21000 Dijon, France
| | - Sarra Smati
- Clinique d'endocrinologie, Institut du Thorax, CHU de Nantes, Nantes, France
| | | | - Bertrand Cariou
- Clinique d'endocrinologie, Institut du Thorax, CHU de Nantes, Nantes, France
| | - Bruno Vergès
- Service d'endocrinologie, diabétologie, maladies métaboliques, CHU de Dijon, 2, boulevard du Maréchal-de-Lattre, BP 77908, 21000 Dijon, France; Unité Inserm, LNC-UMR 1231, université de Bourgogne, Dijon, France
| |
Collapse
|
27
|
Kamble PG, Pereira MJ, Gustafsson S, Lundkvist P, Castillejo-López C, Fall T, Ingelsson E, Eriksson JW. Role of peroxisome proliferator-activated receptor gamma Pro12Ala polymorphism in human adipose tissue: assessment of adipogenesis and adipocyte glucose and lipid turnover. Adipocyte 2018; 7:285-296. [PMID: 30064293 PMCID: PMC6768277 DOI: 10.1080/21623945.2018.1503030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The protective mechanisms of peroxisome proliferator-activated receptor gamma (PPARγ) Pro12Ala polymorphism in type 2 diabetes (T2D) are unclear. We obtained subcutaneous adipose tissue (AT) before and 3 h after oral glucose (OGTT) in carriers and non-carriers of the Ala allele (12 Pro/Pro, 15 Pro/Ala, and 13 Ala/Ala). Adipogenesis, adipocyte glucose uptake and lipolysis as well as PPARγ target gene expression were investigated and compared between the genotype groups. During fasting and post-OGTT, neither basal nor insulin-stimulated adipocyte glucose uptake differed between genotypes. Compared to fasting, a decreased hormone-sensitive lipase gene expression in Pro/Pro (p < 0.05) was accompanied with a higher antilipolytic effect of insulin post-OGTT (p < 0.01). The adipocyte size was similar across groups. Preadipocyte differentiation rates between Pro/Pro and Ala/Ala were unchanged. In conclusion, no major differences in AT differentiation, glucose uptake, lipolysis or expression of PPARγ target genes were observed between different PPARγ Pro12Ala genotypes. Albeit small, our study may suggest that other pathways in AT or effects exerted in other tissues might contribute to the Pro12Ala-mediated protection against T2D.
Collapse
Affiliation(s)
- Prasad G. Kamble
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Maria J. Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Stefan Gustafsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Per Lundkvist
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Casimiro Castillejo-López
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Erik Ingelsson
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jan W. Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
28
|
Role of mTOR in Glucose and Lipid Metabolism. Int J Mol Sci 2018; 19:ijms19072043. [PMID: 30011848 PMCID: PMC6073766 DOI: 10.3390/ijms19072043] [Citation(s) in RCA: 174] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin, mTOR is the master regulator of a cell’s growth and metabolic state in response to nutrients, growth factors and many extracellular cues. Its dysregulation leads to a number of metabolic pathological conditions, including obesity and type 2 diabetes. Here, we review recent findings on the role of mTOR in major metabolic organs, such as adipose tissues, liver, muscle, pancreas and brain. And their potentials as the mTOR related pharmacological targets will be also discussed.
Collapse
|
29
|
Inhibition of FOXO1 transcription factor in primary human adipocytes mimics the insulin-resistant state of type 2 diabetes. Biochem J 2018; 475:1807-1820. [PMID: 29724916 DOI: 10.1042/bcj20180144] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is characterized by insulin resistance in the expanding adipose tissue of obesity. The insulin resistance manifests in human adipocytes as system-wide impairment of insulin signalling. An exception is the regulation of transcription factor FOXO1 (forkhead box protein O1), which is phosphorylated downstream of mTORC2 (mammalian/mechanistic target of rapamycin in complex with raptor) and is therefore not exhibiting impaired response to insulin. However, the abundance, and activity, of FOXO1 is reduced by half in adipocytes from patients with diabetes. To elucidate the effect of reduced FOXO1 activity, we here transduced human adipocytes with a dominant-negative construct of FOXO1 (DN-FOXO1). Inhibition of FOXO1 reduced the abundance of insulin receptor, glucose transporter-4, ribosomal protein S6, mTOR and raptor. Functionally, inhibition of FOXO1 induced an insulin-resistant state network-wide, a state that qualitatively and quantitatively mimicked adipocytes from patients with type 2 diabetes. In contrast, and in accordance with these effects of DN-FOXO1, overexpression of wild-type FOXO1 appeared to augment insulin signalling. We combined experimental data with mathematical modelling to show that the impaired insulin signalling in FOXO1-inhibited cells to a large extent can be explained by reduced mTORC1 activity - a mechanism that defines much of the diabetic state in human adipocytes. Our findings demonstrate that FOXO1 is critical for maintaining normal insulin signalling of human adipocytes.
Collapse
|
30
|
Abstract
The mechanistic target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that senses and integrates environmental information into cellular regulation and homeostasis. Accumulating evidence has suggested a master role of mTOR signalling in many fundamental aspects of cell biology and organismal development. mTOR deregulation is implicated in a broad range of pathological conditions, including diabetes, cancer, neurodegenerative diseases, myopathies, inflammatory, infectious, and autoimmune conditions. Here, we review recent advances in our knowledge of mTOR signalling in mammalian physiology. We also discuss the impact of mTOR alteration in human diseases and how targeting mTOR function can treat human diseases.
Collapse
Affiliation(s)
- Yassine El Hiani
- a Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS B3H 4R2, Canada
| | - Emmanuel Eroume-A Egom
- b Jewish General Hospital and Lady Davis Institute for Medical Research, Montreal, QC H3T 1E2, Canada
| | - Xian-Ping Dong
- a Department of Physiology and Biophysics, Dalhousie University, PO Box 15000, Halifax, NS B3H 4R2, Canada
| |
Collapse
|
31
|
Ooi DJ, Azmi NH, Imam MU, Alitheen NB, Ismail M. Curculigoside and polyphenol-rich ethyl acetate fraction of Molineria latifolia rhizome improved glucose uptake via potential mTOR/AKT activated GLUT4 translocation. J Food Drug Anal 2018; 26:1253-1264. [PMID: 30249324 PMCID: PMC9298560 DOI: 10.1016/j.jfda.2018.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/14/2018] [Accepted: 03/13/2018] [Indexed: 02/08/2023] Open
Abstract
Adipose tissue is one of the major organs responsible for rapid restoration of postprandial glucose fluxes. Being the major isoform of glucose transporter in adipose tissue, regulations of insulin-dependent GLUT4 trafficking have always been of research interest. The present study aimed to examine the molecular mechanisms underlying the efficacy of curculigoside and polyphenol-rich ethyl acetate fraction (EAF) of Molineria latifolia rhizome in triggering glucose uptake. We assessed the adipogenic potential and glucose uptake stimulatory activity of curculigoside and EAF by employing a murine 3T3-L1 adipocyte model. The transcriptional and translational expressions of selected intermediates in the insulin signalling pathway were evaluated. While curculigoside neither promoted adipogenesis nor activated peroxisome proliferator activated receptor gamma, treatment with polyphenol-rich EAF resulted otherwise. However, both treatments enhanced insulin-stimulated uptake of glucose. This was coupled with increased availability of GLUT4 at the plasma membrane of the differentiated adipocytes although the total GLUT4 protein level was unaffected. In addition, the treatment increased the phosphorylation of both AKT and mTOR, which have been reported to be associated with GLUT4 translocation. The present findings proposed that curculigoside and EAF increased glucose transport activity of 3T3-L1 adipocytes via GLUT4 translocation as a result of potential mTOR/AKT activation. The more potent efficacy observed with EAF suggested potential synergistic and multi-targeted action.
Collapse
Affiliation(s)
- Der Jiun Ooi
- Nutri-Cosmeceuticals, Nutrigenomics & Nanodelivery Programme, Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Nur Hanisah Azmi
- Nutri-Cosmeceuticals, Nutrigenomics & Nanodelivery Programme, Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Mustapha Umar Imam
- Nutri-Cosmeceuticals, Nutrigenomics & Nanodelivery Programme, Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia
| | - Maznah Ismail
- Nutri-Cosmeceuticals, Nutrigenomics & Nanodelivery Programme, Laboratory of Molecular Biomedicine, Institute of Bioscience, Universiti Putra Malaysia, 43400, Serdang, Selangor, Malaysia.
| |
Collapse
|
32
|
Abstract
Mechanistic target of rapamycin (mTOR) is the kinase subunit of two structurally and functionally distinct large multiprotein complexes, referred to as mTOR complex 1 (mTORC1) and mTORC2. mTORC1 and mTORC2 play key physiological roles as they control anabolic and catabolic processes in response to external cues in a variety of tissues and organs. However, mTORC1 and mTORC2 activities are deregulated in widespread human diseases, including cancer. Cancer cells take advantage of mTOR oncogenic signaling to drive their proliferation, survival, metabolic transformation, and metastatic potential. Therefore, mTOR lends itself very well as a therapeutic target for innovative cancer treatment. mTOR was initially identified as the target of the antibiotic rapamycin that displayed remarkable antitumor activity in vitro Promising preclinical studies using rapamycin and its derivatives (rapalogs) demonstrated efficacy in many human cancer types, hence supporting the launch of numerous clinical trials aimed to evaluate the real effectiveness of mTOR-targeted therapies. However, rapamycin and rapalogs have shown very limited activity in most clinical contexts, also when combined with other drugs. Thus, novel classes of mTOR inhibitors with a stronger antineoplastic potency have been developed. Nevertheless, emerging clinical data suggest that also these novel mTOR-targeting drugs may have a weak antitumor activity. Here, we summarize the current status of available mTOR inhibitors and highlight the most relevant results from both preclinical and clinical studies that have provided valuable insights into both their efficacy and failure.
Collapse
|
33
|
Lee HK, Kwon B, Lemere CA, de la Monte S, Itamura K, Ha AY, Querfurth HW. mTORC2 (Rictor) in Alzheimer's Disease and Reversal of Amyloid-β Expression-Induced Insulin Resistance and Toxicity in Rat Primary Cortical Neurons. J Alzheimers Dis 2018; 56:1015-1036. [PMID: 28035937 DOI: 10.3233/jad-161029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Mammalian target of rapamycin complex 1 (mTORC1), a nutrient sensor and central controller of cell growth and proliferation, is altered in various models of Alzheimer's disease (AD). Even less studied or understood in AD is mammalian target of rapamycin complex 2 (mTORC2) that influences cellular metabolism, in part through the regulations of Akt/PKB and SGK. Dysregulation of insulin/PI3K/Akt signaling is another important feature of AD pathogenesis. We found that both total mTORC1 and C2 protein levels and individual C1 and C2 enzymatic activities were decreased in human AD brain samples. In two rodent AD models, mTORC1 and C2 activities were also decreased. In a neuronal culture model of AD characterized by accumulation of cellular amyloid-β (Aβ)42, mTORC1 activity was reduced. Autophagic vesicles and markers were correspondingly increased and new protein synthesis was inhibited, consistent with mTORC1 hypofunction. Interestingly, mTORC2 activity in neural culture seemed resistant to the effects of intracellular amyloid. In various cell lines, Aβ expression provoked insulin resistance, characterized by inhibition of stimulated Akt phosphorylation, and an increase in negative mTORC1 regular, p-AMPK, itself a nutrient sensor. Rapamycin decreased phospho-mTOR and to lesser degree p-Rictor. This further suppression of mTORC1 activity protected cells from Aβ-induced toxicity and insulin resistance. More striking, Rictor over-expression fully reversed the Aβ-effects on primary neuronal cultures. Finally, using in vitro assay, Rictor protein addition completely overcame oligomeric Aβ-induced inhibition of the PDK-Akt activation step. We conclude that striking a new balance by restoring mTORC2 abundance and/or inhibition of mTORC1 has therapeutic potential in AD.
Collapse
Affiliation(s)
- Han-Kyu Lee
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Bumsup Kwon
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Cynthia A Lemere
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Suzanne de la Monte
- Department of Pathology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Kyohei Itamura
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Austin Y Ha
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| | - Henry W Querfurth
- Department of Neurology, Rhode Island Hospital and Brown University Warren Alpert Medical School, Providence, RI, USA
| |
Collapse
|
34
|
Fonseca ACRG, Carvalho E, Eriksson JW, Pereira MJ. Calcineurin is an important factor involved in glucose uptake in human adipocytes. Mol Cell Biochem 2018; 445:157-168. [PMID: 29380240 PMCID: PMC6060758 DOI: 10.1007/s11010-017-3261-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/23/2017] [Indexed: 11/24/2022]
Abstract
Calcineurin inhibitors are used in immunosuppressive therapy applied after transplantation, but they are associated with major metabolic side effects including the development of new onset diabetes. Previously, we have shown that the calcineurin inhibiting drugs tacrolimus and cyclosporin A reduce adipocyte and myocyte glucose uptakes by reducing the amount of glucose transporter type 4 (GLUT4) at the cell surface, due to an increased internalization rate. However, this happens without alteration in total protein and phosphorylation levels of key proteins involved in insulin signalling or in the total amount of GLUT4. The present study evaluates possible pathways involved in the altered internalization of GLUT4 and consequent reduction of glucose uptake provoked by calcineurin inhibitors in human subcutaneous adipose tissue. Short- and long-term treatments with tacrolimus, cyclosporin A or another CNI deltamethrin (herbicide) decreased basal and insulin-dependent glucose uptake in adipocytes, without any additive effects observed when added together. However, no tacrolimus effects were observed on glucose uptake when gene transcription and protein translation were inhibited. Investigation of genes potentially involved in GLUT4 trafficking showed only a small effect on ARHGEF11 gene expression (p < 0.05). In conlusion, the specific inhibition of calcineurin, but not that of protein phosphatases, decreases glucose uptake in human subcutaneous adipocytes, suggesting that calcineurin is an important regulator of glucose transport. This inhibitory effect is mediated via gene transcription or protein translation; however, expression of genes potentially involved in GLUT4 trafficking and endocytosis appears not to be involved in these effects.
Collapse
Affiliation(s)
- Ana Catarina R G Fonseca
- Department of Medical Sciences, University of Uppsala, 751 85, Uppsala, Sweden.,Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Eugénia Carvalho
- Center of Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,The Portuguese Diabetes Association (APDP), 1250-203, Lisbon, Portugal.,Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.,Arkansas Children's Research Institute, Little Rock, AR, 72202, USA
| | - Jan W Eriksson
- Department of Medical Sciences, University of Uppsala, 751 85, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, University of Uppsala, 751 85, Uppsala, Sweden.
| |
Collapse
|
35
|
Burgeiro A, Cerqueira MG, Varela-Rodríguez BM, Nunes S, Neto P, Pereira FC, Reis F, Carvalho E. Glucose and Lipid Dysmetabolism in a Rat Model of Prediabetes Induced by a High-Sucrose Diet. Nutrients 2017. [PMID: 28635632 PMCID: PMC5490617 DOI: 10.3390/nu9060638] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Glucotoxicity and lipotoxicity are key features of type 2 diabetes mellitus, but their molecular nature during the early stages of the disease remains to be elucidated. We aimed to characterize glucose and lipid metabolism in insulin-target organs (liver, skeletal muscle, and white adipose tissue) in a rat model treated with a high-sucrose (HSu) diet. Two groups of 16-week-old male Wistar rats underwent a 9-week protocol: HSu diet (n = 10)-received 35% of sucrose in drinking water; Control (n = 12)-received vehicle (water). Body weight, food, and beverage consumption were monitored and glucose, insulin, and lipid profiles were measured. Serum and liver triglyceride concentrations, as well as the expression of genes and proteins involved in lipid biosynthesis were assessed. The insulin-stimulated glucose uptake and isoproterenol-stimulated lipolysis were also measured in freshly isolated adipocytes. Even in the absence of obesity, this rat model already presented the main features of prediabetes, with fasting normoglycemia but reduced glucose tolerance, postprandial hyperglycemia, compensatory hyperinsulinemia, as well as decreased insulin sensitivity (resistance) and hypertriglyceridemia. In addition, impaired hepatic function, including altered gluconeogenic and lipogenic pathways, as well as increased expression of acetyl-coenzyme A carboxylase 1 and fatty acid synthase in the liver, were observed, suggesting that liver glucose and lipid dysmetabolism may play a major role at this stage of the disease.
Collapse
Affiliation(s)
- Ana Burgeiro
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Manuela G Cerqueira
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Bárbara M Varela-Rodríguez
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
| | - Sara Nunes
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Paula Neto
- Service of Anatomical Pathology, Coimbra University Hospital Centre (CHUC), 3000-075 Coimbra, Portugal.
| | - Frederico C Pereira
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Flávio Reis
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
- Laboratory of Pharmacology and Experimental Therapeutics, Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal.
| | - Eugénia Carvalho
- Center of Neuroscience and Cell Biology (CNC) and CNC.IBILI Research Consortium, University of Coimbra, 3004-504 Coimbra, Portugal.
- The Portuguese Diabetes Association (APDP), 1250-203 Lisbon, Portugal.
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
- Arkansas Children's Hospital Research Institute, Little Rock, AR 72202, USA.
| |
Collapse
|
36
|
Dahlman I, Belarbi Y, Laurencikiene J, Pettersson AM, Arner P, Kulyté A. Comprehensive functional screening of miRNAs involved in fat cell insulin sensitivity among women. Am J Physiol Endocrinol Metab 2017; 312:E482-E494. [PMID: 28270439 DOI: 10.1152/ajpendo.00251.2016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 01/18/2017] [Accepted: 02/22/2017] [Indexed: 01/12/2023]
Abstract
The key pathological link between obesity and type 2 diabetes is insulin resistance, but the molecular mechanisms are not entirely identified. micro-RNAs (miRNA) are dysregulated in obesity and may contribute to insulin resistance. Our objective was to detect and functionally investigate miRNAs linked to insulin sensitivity in human subcutaneous white adipose tissue (scWAT). Subjects were selected based on the insulin-stimulated lipogenesis response of subcutaneous adipocytes. Global miRNA profiling was performed in abdominal scWAT of 18 obese insulin-resistance (OIR), 21 obese insulin-sensitive (OIS), and 9 lean women. miRNAs demonstrating differential expression between OIR and OIS women were overexpressed in human in vitro-differentiated adipocytes followed by assessment of lipogenesis and identification of miRNA targets by measuring mRNA/protein expression and 3'-untranslated region analysis. Eleven miRNAs displayed differential expression between OIR and OIS states. Overexpression of miR-143-3p and miR-652-3p increased insulin-stimulated lipogenesis in human in vitro differentiated adipocytes and directly or indirectly affected several genes/proteins involved in insulin signaling at transcriptional or posttranscriptional levels. Adipose expression of miR-143-3p and miR-652-3p was positively associated with insulin-stimulated lipogenesis in scWAT independent of body mass index. In conclusion, miR-143-3p and miR-652-3p are linked to scWAT insulin resistance independent of obesity and influence insulin-stimulated lipogenesis by interacting at different steps with insulin-signaling pathways.
Collapse
Affiliation(s)
- Ingrid Dahlman
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Yasmina Belarbi
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jurga Laurencikiene
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Annie M Pettersson
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Peter Arner
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Agné Kulyté
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
37
|
Moderate lifelong overexpression of tuberous sclerosis complex 1 (TSC1) improves health and survival in mice. Sci Rep 2017; 7:834. [PMID: 28400571 PMCID: PMC5429778 DOI: 10.1038/s41598-017-00970-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The tuberous sclerosis complex 1/2 (TSC1/2) is an endogenous regulator of the mechanistic target of rapamycin (mTOR). While mTOR has been shown to play an important role in health and aging, the role of TSC1/2 in aging has not been fully investigated. In the current study, a constitutive TSC1 transgenic (Tsc1tg) mouse model was generated and characterized. mTORC1 signaling was reduced in majority of the tissues, except the brain. In contrast, mTORC2 signaling was enhanced in Tsc1tg mice. Tsc1tg mice are more tolerant to exhaustive exercises and less susceptible to isoproterenol-induced cardiac hypertrophy at both young and advanced ages. Tsc1tg mice have less fibrosis and inflammation in aged as well as isoproterenol-challenged heart than age-matched wild type mice. The female Tsc1tg mice exhibit a higher fat to lean mass ratio at advanced ages than age-matched wild type mice. More importantly, the lifespan increased significantly in female Tsc1tg mice, but not in male Tsc1tg mice. Collectively, our data demonstrated that moderate increase of TSC1 expression can enhance overall health, particularly cardiovascular health, and improve survival in a gender-specific manner.
Collapse
|
38
|
Sidibeh CO, Pereira MJ, Lau Börjesson J, Kamble PG, Skrtic S, Katsogiannos P, Sundbom M, Svensson MK, Eriksson JW. Role of cannabinoid receptor 1 in human adipose tissue for lipolysis regulation and insulin resistance. Endocrine 2017; 55:839-852. [PMID: 27858284 PMCID: PMC5316391 DOI: 10.1007/s12020-016-1172-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/03/2016] [Indexed: 12/31/2022]
Abstract
We recently showed that the peripheral cannabinoid receptor type 1 (CNR1) gene is upregulated by the synthetic glucocorticoid dexamethasone. CNR1 is highly expressed in the central nervous system and has been a drug target for the treatment of obesity. Here we explore the role of peripheral CNR1 in states of insulin resistance in human adipose tissue. Subcutaneous adipose tissue was obtained from well-controlled type 2 diabetes subjects and controls. Subcutaneous adipose tissue gene expression levels of CNR1 and endocannabinoid synthesizing and degrading enzymes were assessed. Furthermore, paired human subcutaneous adipose tissue and omental adipose tissue from non-diabetic volunteers undergoing kidney donation or bariatric surgery, was incubated with or without dexamethasone. Subcutaneous adipose tissue obtained from volunteers through needle biopsy was incubated with or without dexamethasone and in the presence or absence of the CNR1-specific antagonist AM281. CNR1 gene and protein expression, lipolysis and glucose uptake were evaluated. Subcutaneous adipose tissue CNR1 gene expression levels were 2-fold elevated in type 2 diabetes subjects compared with control subjects. Additionally, gene expression levels of CNR1 and endocannabinoid-regulating enzymes from both groups correlated with markers of insulin resistance. Dexamethasone increased CNR1 expression dose-dependently in subcutaneous adipose tissue and omental adipose tissue by up to 25-fold. Dexamethasone pre-treatment of subcutaneous adipose tissue increased lipolysis rate and reduced glucose uptake. Co-incubation with the CNR1 antagonist AM281 prevented the stimulatory effect on lipolysis, but had no effect on glucose uptake. CNR1 is upregulated in states of type 2 diabetes and insulin resistance. Furthermore, CNR1 is involved in glucocorticoid-regulated lipolysis. Peripheral CNR1 could be an interesting drug target in type 2 diabetes and dyslipidemia.
Collapse
Affiliation(s)
- Cherno O Sidibeh
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | - Prasad G Kamble
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- AstraZeneca R&D, Mölndal, Sweden
- Department of Endocrinology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Magnus Sundbom
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Maria K Svensson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Cross-talks via mTORC2 can explain enhanced activation in response to insulin in diabetic patients. Biosci Rep 2017; 37:BSR20160514. [PMID: 27986865 PMCID: PMC5271673 DOI: 10.1042/bsr20160514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 12/08/2016] [Accepted: 12/16/2016] [Indexed: 01/01/2023] Open
Abstract
The molecular mechanisms of insulin resistance in Type 2 diabetes have been
extensively studied in primary human adipocytes, and mathematical modelling has
clarified the central role of attenuation of mammalian target of rapamycin
(mTOR) complex 1 (mTORC1) activity in the diabetic state. Attenuation of mTORC1
in diabetes quells insulin-signalling network-wide, except for the mTOR in
complex 2 (mTORC2)-catalysed phosphorylation of protein kinase B (PKB) at
Ser473 (PKB-S473P), which is increased. This unique increase
could potentially be explained by feedback and interbranch cross-talk signals.
To examine if such mechanisms operate in adipocytes, we herein analysed data
from an unbiased phosphoproteomic screen in 3T3-L1 adipocytes. Using a
mathematical modelling approach, we showed that a negative signal from
mTORC1-p70 S6 kinase (S6K) to rictor–mTORC2 in combination with a
positive signal from PKB to SIN1–mTORC2 are compatible with the
experimental data. This combined cross-branch signalling predicted an increased
PKB-S473P in response to attenuation of mTORC1 – a distinguishing feature
of the insulin resistant state in human adipocytes. This aspect of insulin
signalling was then verified for our comprehensive model of insulin signalling
in human adipocytes. Introduction of the cross-branch signals was compatible
with all data for insulin signalling in human adipocytes, and the resulting
model can explain all data network-wide, including the increased PKB-S473P in
the diabetic state. Our approach was to first identify potential mechanisms in
data from a phosphoproteomic screen in a cell line, and then verify such
mechanisms in primary human cells, which demonstrates how an unbiased approach
can support a direct knowledge-based study.
Collapse
|
40
|
Pereira MJ, Skrtic S, Katsogiannos P, Abrahamsson N, Sidibeh CO, Dahgam S, Månsson M, Risérus U, Kullberg J, Eriksson JW. Impaired adipose tissue lipid storage, but not altered lipolysis, contributes to elevated levels of NEFA in type 2 diabetes. Degree of hyperglycemia and adiposity are important factors. Metabolism 2016; 65:1768-1780. [PMID: 27832864 DOI: 10.1016/j.metabol.2016.09.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/06/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Elevated levels of circulating non-esterified fatty acids (NEFA) mediate many adverse metabolic effects. In this work we aim to determine the impact of type 2 diabetes (T2D), glycemic control and obesity on lipolysis regulation. DESIGN AND PARTICIPANTS 20 control and 20 metformin-treated T2D subjects were matched for sex (10M/10 F), age (58±11 vs 58±9 y) and BMI (30.8±4.6 vs 30.7±4.9kg/m2). In vivo lipolysis was assessed during a 3h-OGTT with plasma glycerol and NEFA levels. Subcutaneous adipose tissue (SAT) biopsies were obtained to measure mRNA and metabolite levels of factors related to lipolysis and lipid storage and to assess in vitro lipolysis in isolated subcutaneous adipocytes. RESULTS Plasma NEFA AUC during the OGTT where higher 30% (P=0.005) in T2D than in control subjects, but plasma glycerol AUC and subcutaneous adipocyte lipolysis in vitro were similar, suggesting that adipose tissue lipolysis is not altered. Expression in SAT of genes involved in lipid storage (FABP4, DGAT1, FASN) were reduced in T2D subjects compared with controls, but no differences were seen for genes involved in lipolysis. T2D subjects had elevated markers of beta-oxidation, α-hydroxybutyrate (1.4-fold, P<0.01) and β-hydroxybutyrate (1.7-fold, P<0.05) in plasma. In multivariate analysis, HbA1c, visceral adipose tissue volume and sex (male) were significantly associated with NEFA AUC in T2D subjects. CONCLUSIONS In T2D subjects, NEFA turnover is impaired, but not due to defects in lipolysis or lipid beta-oxidation. Impaired adipose NEFA re-esterification or de novo lipogenesis is likely to contribute to higher NEFA plasma levels in T2D. The data suggest that hyperglycemia and adiposity are important contributing factors for the regulation of plasma NEFA concentrations.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- AstraZeneca R&D, Mölndal, Sweden; Department of Endocrinology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Cherno O Sidibeh
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Ulf Risérus
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Joel Kullberg
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
41
|
Fu Y, Zheng X, Jia X, Binderiya U, Wang Y, Bao W, Bao L, Zhao K, Fu Y, Hao H, Wang Z. A quantitative transcriptomic analysis of the physiological significance of mTOR signaling in goat fetal fibroblasts. BMC Genomics 2016; 17:879. [PMID: 27821074 PMCID: PMC5098276 DOI: 10.1186/s12864-016-3151-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 10/11/2016] [Indexed: 12/12/2022] Open
Abstract
Background Mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that is a central regulator of cell growth and metabolism. CCI-779 is a specific inhibitor of the mTORC1 signaling pathway. Results We performed comparative transcriptome profiling on Inner Mongolia Cashmere goat fetal fibroblasts (GFbs) that were treated with CCI-779 and untreated cells. A total of 365 differentially expressed genes (DEGs) appeared between untreated and CCI-779-treated GFbs, with an FDR ≤0.001 and fold-change ≥2. These 365 DEGs were associated with mTOR signaling; 144 were upregulated in CCI-779-treated cells, and 221 were downregulated. Additionally, 300 genes were annotated with 43 Gene Ontology (GO) terms, and 293 genes were annotated with 194 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. Three RNA polymerase II and polymerase III subunits, 3 transcription factors, and 5 kinases in mTOR signaling were differentially expressed in CCI-779-treated GFbs. Further 6 DEGs were related to amino acid metabolism, 11 mediated lipid metabolism, 11 participated in carbohydrate metabolism, and 5 were involved in single-nucleotide metabolism. Based on our quantitative transcriptomic analysis, 40 significant DEGs with important function related to metabolism, RNA polymerase, transcription factors and mTOR signaling were selected for qPCR analysis, and the quantitative results between the two analysis methods were concordant. The qPCR data confirmed the differential expression in the RNA-Seq experiments. To validate the translational significance of the findings in certain differentially expressed genes, S6K1 and VEGF were detected by western blot, and these two proteins showed a differential expression between non-treated and treated with CCI-779 groups, which were consistent with mRNA abundance. The data showed a preliminary significance of the findings in the protein levels. Conclusions CCI-779 induces transcriptomic changes, and mTOR signaling might have significant function in the activation of RNA polymerase and certain transcription factors and in the metabolism of amino acids, lipids, carbohydrates, and single nucleotides in GFbs. These data filled the vacancy in the systematical profiling of mTOR signaling on Cashmere goat fetal fibroblasts. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3151-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yuting Fu
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Xu Zheng
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China.,Clinical Laboratory, The Hulunbuir People's Hospital, Hailaer, 021008, China
| | - Xiaoyang Jia
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Uyanga Binderiya
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Yanfeng Wang
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Wenlei Bao
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Lili Bao
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China.,College of Basic Medical Science, Inner Mongolia Medical University, Hohhot, 010021, China
| | - Keyu Zhao
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Yu Fu
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China
| | - Huifang Hao
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China.
| | - Zhigang Wang
- College of Life Sciences, Inner Mongolia University, Hohhot, 010021, China.
| |
Collapse
|
42
|
Han E, Kim MS, Kim YS, Kang ES. Risk assessment and management of post-transplant diabetes mellitus. Metabolism 2016; 65:1559-69. [PMID: 27621191 DOI: 10.1016/j.metabol.2016.07.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/13/2016] [Accepted: 07/21/2016] [Indexed: 02/06/2023]
Abstract
The success rate of organ transplantation has been increasing with advances in surgical and pharmacological techniques. However, the number of solid organ transplant recipients who require metabolic disease management is also growing. Post-transplant diabetes mellitus (PTDM) is a common complication after solid organ transplantation and is associated with risks of graft loss, cardiovascular morbidity, and mortality. Other risk factors for PTDM include older age, genetic background, obesity, hepatitis C virus infection, hypomagnesemia, and use of immunosuppressant agents (corticosteroids, calcineurin inhibitors, and mammalian target of rapamycin inhibitor). Management of PTDM should be started before the transplantation plan to properly screen high-risk patients. Even though PTDM management is similar to that of general type 2 diabetes, therapeutic approaches must be made with consideration of drug interactions between immunosuppressive agents, glucose-lowering medications, and graft rejection and function.
Collapse
Affiliation(s)
- Eugene Han
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Hospital Diabetes Center
| | - Myoung Soo Kim
- Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Yu Seun Kim
- Department of Transplantation Surgery, Severance Hospital, Yonsei University Health System, Seoul, Republic of Korea
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea; Severance Hospital Diabetes Center; Institute of Endocrine Research, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
43
|
Rapamycin negatively impacts insulin signaling, glucose uptake and uncoupling protein-1 in brown adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1929-1941. [PMID: 27686967 DOI: 10.1016/j.bbalip.2016.09.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/20/2016] [Accepted: 09/22/2016] [Indexed: 11/21/2022]
Abstract
New onset diabetes after transplantation (NODAT) is a metabolic disorder that affects 40% of patients on immunosuppressive agent (IA) treatment, such as rapamycin (also known as sirolimus). IAs negatively modulate insulin action in peripheral tissues including skeletal muscle, liver and white fat. However, the effects of IAs on insulin sensitivity and thermogenesis in brown adipose tissue (BAT) have not been investigated. We have analyzed the impact of rapamycin on insulin signaling, thermogenic gene-expression and mitochondrial respiration in BAT. Treatment of brown adipocytes with rapamycin for 16h significantly decreased insulin receptor substrate 1 (IRS1) protein expression and insulin-mediated protein kinase B (Akt) phosphorylation. Consequently, both insulin-induced glucose transporter 4 (GLUT4) translocation to the plasma membrane and glucose uptake were decreased. Early activation of the N-terminal Janus activated kinase (JNK) was also observed, thereby increasing IRS1 Ser 307 phosphorylation. These effects of rapamycin on insulin signaling in brown adipocytes were partly prevented by a JNK inhibitor. In vivo treatment of rats with rapamycin for three weeks abolished insulin-mediated Akt phosphorylation in BAT. Rapamycin also inhibited norepinephrine (NE)-induced lipolysis, the expression of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and uncoupling protein (UCP)-1 in brown adipocytes. Importantly, basal mitochondrial respiration, proton leak and maximal respiratory capacity were significantly decreased in brown adipocytes treated with rapamycin. In conclusion, we demonstrate, for the first time the important role of brown adipocytes as target cells of rapamycin, suggesting that insulin resistance in BAT might play a major role in NODAT development.
Collapse
|
44
|
Effects of insulin on the skin: possible healing benefits for diabetic foot ulcers. Arch Dermatol Res 2016; 308:677-694. [PMID: 27655635 DOI: 10.1007/s00403-016-1686-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 08/04/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
Diabetic foot ulcers affect 15-20 % of all diabetic patients and remain an important challenge since the available therapies have limited efficacy and some of the novel therapeutic approaches, which include growth factors and stem cells, are highly expensive and their safety remains to be evaluated. Despite its low cost and safety, the interest for topical insulin as a healing agent has increased only in the last 20 years. The molecular mechanisms of insulin signaling and its metabolic effects have been well studied in its classical target tissues. However, little is known about the specific effects of insulin in healthy or even diabetic skin. In addition, the mechanisms involved in the effects of insulin on wound healing have been virtually unknown until about 10 years ago. This paper will review the most recent advances in the cellular and molecular mechanisms that underlie the beneficial effects of insulin on skin wound healing in diabetes. Emerging evidence that links dysfunction of key cellular organelles, namely the endoplasmic reticulum and the mitochondria, to changes in the autophagy response, as well as the impaired wound healing in diabetic patients will also be discussed along with the putative mechanisms whereby insulin could regulate/modulate these alterations.
Collapse
|
45
|
Tharp WG, Gupta D, Smith J, Jones KP, Jones AM, Pratley RE. Effects of glucose and insulin on secretion of amyloid-β by human adipose tissue cells. Obesity (Silver Spring) 2016; 24:1471-9. [PMID: 27172814 PMCID: PMC5084817 DOI: 10.1002/oby.21494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Obesity and type 2 diabetes mellitus are risk factors for developing Alzheimer disease. Overlapping patterns of metabolic dysfunction may be common molecular links between these complex diseases. Amyloid-β (Aβ) precursor protein and associated β- and γ-secretases are expressed in adipose tissue. Aβ precursor protein is up-regulated with obesity and correlated to insulin resistance. Aβ may be secreted by adipose tissue, its production may be regulated through metabolic pathways, and Aβ may exert effects on adipose tissue insulin receptor signaling. METHODS Human stromal-vascular cells and differentiated adipocytes were cultured with different combinations of glucose and insulin and then assayed for Aβ in conditioned media. Aβ was measured in vivo using adipose tissue microdialysis. RESULTS Aβ secretion was increased by glucose and insulin in vitro. Adipose tissue microdialysates contained Aβ. Adipocytes treated with Aβ had decreased expression of insulin receptor substrate-2 and reduced Akt-1 phosphorylation. CONCLUSIONS Aβ was made by adipose tissue cells in vitro at concentrations similar to in vivo measurements. Regulation of Aβ production by glucose and insulin and effects of Aβ on the insulin receptor pathway suggest similar cellular mechanisms may exist between neuronal dysfunction in Alzheimer disease and adipose dysfunction in type 2 diabetes.
Collapse
Affiliation(s)
- William G. Tharp
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Dhananjay Gupta
- Department of Medicine Endocrine UnitUniversity of Vermont College of MedicineBurlingtonVermontUSA
| | - Joshua Smith
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Karen P. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Amanda M. Jones
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| | - Richard E. Pratley
- Florida Hospital Sanford/Burnham Translational Research Institute for Metabolism and DiabetesOrlandoFloridaUSA
| |
Collapse
|
46
|
Cai M, Shen R, Song L, Lu M, Wang J, Zhao S, Tang Y, Meng X, Li Z, He ZX. Bone Marrow Mesenchymal Stem Cells (BM-MSCs) Improve Heart Function in Swine Myocardial Infarction Model through Paracrine Effects. Sci Rep 2016; 6:28250. [PMID: 27321050 PMCID: PMC4913323 DOI: 10.1038/srep28250] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/01/2016] [Indexed: 12/14/2022] Open
Abstract
Stem cells are promising for the treatment of myocardial infarction (MI) and large animal models should be used to better understand the full spectrum of stem cell actions and preclinical evidences. In this study, bone marrow mesenchymal stem cells (BM-MSCs) were transplanted into swine heart ischemia model. To detect glucose metabolism in global left ventricular myocardium and regional myocardium, combined with assessment of cardiac function, positron emission tomography-computer tomography (PET-CT) and magnetic resonance imaging (MRI) were performed. To study the changes of glucose transporters and glucose metabolism-related enzymes and the signal transduction pathway, RT-PCR, Western-blot, and immunohistochemistry were carried out. Myocardium metabolic evaluation by PET-CT showed that mean signal intensity (MSI) increased in these segments at week 4 compared with that at week 1 after BM-MSCs transplantation. Moreover, MRI demonstrated significant function enhancement in BM-MSCs group. The gene expressions of glucose transporters (GLUT1, GLUT4), glucose metabolism-related enzymes phosphofructokinase (PFK), and glyceraldehyde-3-phosphate dehydrogenase (GAPDH)) and 70-kDa ribosomal protein S6 kinase (p70s6k) in BM-MSCs injected areas were up-regulated at week 4 after BM-MSCs transplantation and this was confirmed by Western-blot and immunohistochemistry. In conclusions, BM-MSCs transplantation could improve cardiac function in swine MI model by activation of mTOR signal transduction pathway.
Collapse
Affiliation(s)
- Min Cai
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China.,Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Shen
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Song
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Minjie Lu
- Department of Radiology, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Jianguang Wang
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Shihua Zhao
- Department of Nuclear Medicine, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Yue Tang
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Xianmin Meng
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| | - Zongjin Li
- Department of Pathophysiology, Nankai University School of Medicine, Tianjin, China
| | - Zuo-Xiang He
- Department of Nuclear Medicine, State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center of Cardiovascular Disease, Peking Union Medical College &Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Rajan MR, Nyman E, Kjølhede P, Cedersund G, Strålfors P. Systems-wide Experimental and Modeling Analysis of Insulin Signaling through Forkhead Box Protein O1 (FOXO1) in Human Adipocytes, Normally and in Type 2 Diabetes. J Biol Chem 2016; 291:15806-19. [PMID: 27226562 DOI: 10.1074/jbc.m116.715763] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Indexed: 01/31/2023] Open
Abstract
Insulin resistance is a major aspect of type 2 diabetes (T2D), which results from impaired insulin signaling in target cells. Signaling to regulate forkhead box protein O1 (FOXO1) may be the most important mechanism for insulin to control transcription. Despite this, little is known about how insulin regulates FOXO1 and how FOXO1 may contribute to insulin resistance in adipocytes, which are the most critical cell type in the development of insulin resistance. We report a detailed mechanistic analysis of insulin control of FOXO1 in human adipocytes obtained from non-diabetic subjects and from patients with T2D. We show that FOXO1 is mainly phosphorylated through mTORC2-mediated phosphorylation of protein kinase B at Ser(473) and that this mechanism is unperturbed in T2D. We also demonstrate a cross-talk from the MAPK branch of insulin signaling to stimulate phosphorylation of FOXO1. The cellular abundance and consequently activity of FOXO1 are halved in T2D. Interestingly, inhibition of mTORC1 with rapamycin reduces the abundance of FOXO1 to the levels in T2D. This suggests that the reduction of the concentration of FOXO1 is a consequence of attenuation of mTORC1, which defines much of the diabetic state in human adipocytes. We integrate insulin control of FOXO1 in a network-wide mathematical model of insulin signaling dynamics based on compatible data from human adipocytes. The diabetic state is network-wide explained by attenuation of an mTORC1-to-insulin receptor substrate-1 (IRS1) feedback and reduced abundances of insulin receptor, GLUT4, AS160, ribosomal protein S6, and FOXO1. The model demonstrates that attenuation of the mTORC1-to-IRS1 feedback is a major mechanism of insulin resistance in the diabetic state.
Collapse
Affiliation(s)
| | - Elin Nyman
- Biomedical Engineering, Linköping University, SE58185 Linköping, Sweden and Cardiovascular and Metabolic Diseases, Innovative Medicines, and Drug Metabolism and Pharmacokinetics, AstraZeneca Research and Development, 43150 Gothenburg, Sweden
| | - Preben Kjølhede
- From the Departments of Clinical and Experimental Medicine and
| | - Gunnar Cedersund
- From the Departments of Clinical and Experimental Medicine and Biomedical Engineering, Linköping University, SE58185 Linköping, Sweden and
| | - Peter Strålfors
- From the Departments of Clinical and Experimental Medicine and
| |
Collapse
|
48
|
Burgeiro A, Fuhrmann A, Cherian S, Espinoza D, Jarak I, Carvalho RA, Loureiro M, Patrício M, Antunes M, Carvalho E. Glucose uptake and lipid metabolism are impaired in epicardial adipose tissue from heart failure patients with or without diabetes. Am J Physiol Endocrinol Metab 2016; 310:E550-64. [PMID: 26814014 PMCID: PMC4824138 DOI: 10.1152/ajpendo.00384.2015] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/20/2016] [Indexed: 01/25/2023]
Abstract
Type 2 diabetes mellitus is a complex metabolic disease, and cardiovascular disease is a leading complication of diabetes. Epicardial adipose tissue surrounding the heart displays biochemical, thermogenic, and cardioprotective properties. However, the metabolic cross-talk between epicardial fat and the myocardium is largely unknown. This study sought to understand epicardial adipose tissue metabolism from heart failure patients with or without diabetes. We aimed to unravel possible differences in glucose and lipid metabolism between human epicardial and subcutaneous adipocytes and elucidate the potential underlying mechanisms involved in heart failure. Insulin-stimulated [(14)C]glucose uptake and isoproterenol-stimulated lipolysis were measured in isolated epicardial and subcutaneous adipocytes. The expression of genes involved in glucose and lipid metabolism was analyzed by reverse transcription-polymerase chain reaction in adipocytes. In addition, epicardial and subcutaneous fatty acid composition was analyzed by high-resolution proton nuclear magnetic resonance spectroscopy. The difference between basal and insulin conditions in glucose uptake was significantly decreased (P= 0.006) in epicardial compared with subcutaneous adipocytes. Moreover, a significant (P< 0.001) decrease in the isoproterenol-stimulated lipolysis was also observed when the two fat depots were compared, and it was strongly correlated with lipolysis, lipid storage, and inflammation-related gene expression. Moreover, the fatty acid composition of these tissues was significantly altered by diabetes. These results emphasize potential metabolic differences between both fat depots in the presence of heart failure and highlight epicardial fat as a possible therapeutic target in situ in the cardiac microenvironment.
Collapse
Affiliation(s)
- Ana Burgeiro
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Amelia Fuhrmann
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sam Cherian
- Faculty of Integrative Sciences and Technology, Quest International University Perak, Perak, Malaysia
| | - Daniel Espinoza
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ivana Jarak
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Rui A Carvalho
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra, Coimbra, Portugal;
| | - Marisa Loureiro
- Laboratory of Biostatistics and Medical Informatics, IBILI - Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Patrício
- Laboratory of Biostatistics and Medical Informatics, IBILI - Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Manuel Antunes
- Cardiothroracic Surgery Unit at the University Hospital of Coimbra, Coimbra, Portugal
| | - Eugénia Carvalho
- Center of Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Portuguese Diabetes Association, Lisbon, Portugal; Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and Arkansas Children's Hospital Research Institute, Little Rock, Arkansas
| |
Collapse
|
49
|
Röhling M, Herder C, Stemper T, Müssig K. Influence of Acute and Chronic Exercise on Glucose Uptake. J Diabetes Res 2016; 2016:2868652. [PMID: 27069930 PMCID: PMC4812462 DOI: 10.1155/2016/2868652] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 01/31/2016] [Accepted: 02/03/2016] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance plays a key role in the development of type 2 diabetes. It arises from a combination of genetic predisposition and environmental and lifestyle factors including lack of physical exercise and poor nutrition habits. The increased risk of type 2 diabetes is molecularly based on defects in insulin signaling, insulin secretion, and inflammation. The present review aims to give an overview on the molecular mechanisms underlying the uptake of glucose and related signaling pathways after acute and chronic exercise. Physical exercise, as crucial part in the prevention and treatment of diabetes, has marked acute and chronic effects on glucose disposal and related inflammatory signaling pathways. Exercise can stimulate molecular signaling pathways leading to glucose transport into the cell. Furthermore, physical exercise has the potential to modulate inflammatory processes by affecting specific inflammatory signaling pathways which can interfere with signaling pathways of the glucose uptake. The intensity of physical training appears to be the primary determinant of the degree of metabolic improvement modulating the molecular signaling pathways in a dose-response pattern, whereas training modality seems to have a secondary role.
Collapse
Affiliation(s)
- Martin Röhling
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich, 85764 Neuherberg, Germany
| | - Christian Herder
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich, 85764 Neuherberg, Germany
| | - Theodor Stemper
- Department Fitness and Health, University Wuppertal, 42119 Wuppertal, Germany
| | - Karsten Müssig
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
- German Center for Diabetes Research (DZD), Munich, 85764 Neuherberg, Germany
- Department of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| |
Collapse
|
50
|
Bermudez-Silva FJ, Romero-Zerbo SY, Haissaguerre M, Ruz-Maldonado I, Lhamyani S, El Bekay R, Tabarin A, Marsicano G, Cota D. The cannabinoid CB1 receptor and mTORC1 signalling pathways interact to modulate glucose homeostasis in mice. Dis Model Mech 2015; 9:51-61. [PMID: 26563389 PMCID: PMC4728331 DOI: 10.1242/dmm.020750] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 11/02/2015] [Indexed: 12/31/2022] Open
Abstract
The endocannabinoid system (ECS) is an intercellular signalling mechanism that is present in the islets of Langerhans and plays a role in the modulation of insulin secretion and expansion of the β-cell mass. The downstream signalling pathways mediating these effects are poorly understood. Mammalian target of rapamycin complex 1 (mTORC1) signalling is a key intracellular pathway involved in energy homeostasis and is known to importantly affect the physiology of pancreatic islets. We investigated the possible relationship between cannabinoid type 1 (CB1) receptor signalling and the mTORC1 pathway in the endocrine pancreas of mice by using pharmacological analysis as well as mice genetically lacking the CB1 receptor or the downstream target of mTORC1, the kinase p70S6K1. In vitro static secretion experiments on islets, western blotting, and in vivo glucose and insulin tolerance tests were performed. The CB1 receptor antagonist rimonabant decreased glucose-stimulated insulin secretion (GSIS) at 0.1 µM while increasing phosphorylation of p70S6K1 and ribosomal protein S6 (rpS6) within the islets. Specific pharmacological blockade of mTORC1 by 3 nM rapamycin, as well as genetic deletion of p70S6K1, impaired the CB1-antagonist-mediated decrease in GSIS. In vivo experiments showed that 3 mg/kg body weight rimonabant decreased insulin levels and induced glucose intolerance in lean mice without altering peripheral insulin sensitivity; this effect was prevented by peripheral administration of low doses of rapamycin (0.1 mg/kg body weight), which increased insulin sensitivity. These findings suggest a functional interaction between the ECS and the mTORC1 pathway within the endocrine pancreas and at the whole-organism level, which could have implications for the development of new therapeutic approaches for pancreatic β-cell diseases. Summary: Evidence supporting a functional interaction between the endocannabinoid system and the mTORC1 pathway within the endocrine pancreas, which could have implications for the development of new therapeutic approaches for diabetes.
Collapse
Affiliation(s)
- Francisco J Bermudez-Silva
- Unidad de Gestion Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga 29009, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Málaga 29009, Spain INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France
| | - Silvana Y Romero-Zerbo
- Unidad de Gestion Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga 29009, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Málaga 29009, Spain
| | - Magalie Haissaguerre
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France
| | - Inmaculada Ruz-Maldonado
- Unidad de Gestion Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga 29009, Spain Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Málaga 29009, Spain
| | - Said Lhamyani
- Unidad de Gestion Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga 29009, Spain
| | - Rajaa El Bekay
- Unidad de Gestion Clínica Intercentros de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga/Universidad de Málaga, Málaga 29009, Spain
| | - Antoine Tabarin
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Service d'endocrinologie, diabétologie, maladies métaboliques et nutrition, Hôpital Haut-Lévêque, Pessac F-33604, France
| | - Giovanni Marsicano
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France
| | - Daniela Cota
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U862, Bordeaux F-33000, France
| |
Collapse
|