1
|
A Review on Oxidative Stress, Diabetic Complications, and the Roles of Honey Polyphenols. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8878172. [PMID: 33299532 PMCID: PMC7704201 DOI: 10.1155/2020/8878172] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/18/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023]
Abstract
Despite the availability of various antidiabetic drugs, diabetes mellitus (DM) remains one of the world's most prevalent chronic diseases and is a global burden. Hyperglycaemia, a characteristic of type 2 diabetes mellitus (T2DM), substantially leads to the generation of reactive oxygen species (ROS), triggering oxidative stress as well as numerous cellular and molecular modifications such as mitochondrial dysfunction affecting normal physiological functions in the body. In mitochondrial-mediated processes, oxidative pathways play an important role, although the responsible molecular mechanisms remain unclear. The impaired mitochondrial function is evidenced by insulin insensitivity in various cell types. In addition, the roles of master antioxidant pathway nuclear factor erythroid 2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)/antioxidant response elements (ARE) are being deciphered to explain various molecular pathways involved in diabetes. Dietary factors are known to influence diabetes, and many natural dietary factors have been studied to improve diabetes. Honey is primarily rich in carbohydrates and is also abundant in flavonoids and phenolic acids; thus, it is a promising therapeutic antioxidant for various disorders. Various research has indicated that honey has strong wound-healing properties and has antibacterial, anti-inflammatory, antifungal, and antiviral effects; thus, it is a promising antidiabetic agent. The potential antidiabetic mechanisms of honey were proposed based on its major constituents. This review focuses on the various prospects of using honey as an antidiabetic agent and the potential insights.
Collapse
|
2
|
Wong CY, Al-Salami H, Dass CR. C2C12 cell model: its role in understanding of insulin resistance at the molecular level and pharmaceutical development at the preclinical stage. J Pharm Pharmacol 2020; 72:1667-1693. [PMID: 32812252 DOI: 10.1111/jphp.13359] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/17/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES The myoblast cell line, C2C12, has been utilised extensively in vitro as an examination model in understanding metabolic disease progression. Although it is indispensable in both preclinical and pharmaceutical research, a comprehensive review of its use in the investigation of insulin resistance progression and pharmaceutical development is not available. KEY FINDINGS C2C12 is a well-documented model, which can facilitate our understanding in glucose metabolism, insulin signalling mechanism, insulin resistance, oxidative stress, reactive oxygen species and glucose transporters at cellular and molecular levels. With the aid of the C2C12 model, recent studies revealed that insulin resistance has close relationship with various metabolic diseases in terms of disease progression, pathogenesis and therapeutic management. A holistic, safe and effective disease management is highly of interest. Therefore, significant efforts have been paid to explore novel drug compounds and natural herbs that can elicit therapeutic effects in the targeted sites at both cellular (e.g. mitochondria, glucose transporter) and molecular level (e.g. genes, signalling pathway). SUMMARY The use of C2C12 myoblast cell line is meaningful in pharmaceutical and biomedical research due to their expression of GLUT-4 and other features that are representative to human skeletal muscle cells. With the use of the C2C12 cell model, the impact of drug delivery systems (nanoparticles and quantum dots) on skeletal muscle, as well as the relationship between exercise, pancreatic β-cells and endothelial cells, was discovered.
Collapse
Affiliation(s)
- Chun Y Wong
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia.,Curtin Health Innovation Research Institute, Bentley, WA, Australia
| | - Hani Al-Salami
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia.,Curtin Health Innovation Research Institute, Bentley, WA, Australia.,Biotechnology and Drug Development Research Laboratory, Curtin University, Bentley, WA, Australia
| | - Crispin R Dass
- School of Pharmacy and Biomedical Science, Curtin University, Bentley, WA, Australia.,Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
3
|
Jiang B, Zhang X, Di D, Luo G, Shi Y, Zhang J, Berggren-Söderlund M, Nilsson-Ehle P, Xu N. Hyperglycemia-induced downregulation of apolipoprotein M expression is not via the hexosamine pathway. Lipids Health Dis 2015; 14:110. [PMID: 26377577 PMCID: PMC4574082 DOI: 10.1186/s12944-015-0103-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 08/24/2015] [Indexed: 11/10/2022] Open
Abstract
Background We previously demonstrated that hyperglycemia could suppress apolipoprotein M (apoM) synthesis both in vivo and in vitro; however, the mechanism of hyperglycemia-induced downregulation of apoM expression is unknown yet. Methods In the present study we further examined if hexosamine pathway, one of the most important pathways of glucose turnover, being involved in modulating apoM expression in the hyperglycemia condition. We examined the effect of glucosamine, a prominent component of hexosamine pathway and intracellular mediator of insulin resistance, on apoM expression in HepG2 cells and in rat’s models. In the present study we also determined apolipoprotein A1 (apoA1) as a control gene. Results Our results demonstrated that glucosamine could even up-regulate both apoM and apoA1 expressions in HepG2 cell cultures. The glucosamine induced upregulation of apoM expression could be blocked by addition of azaserine, an inhibitor of hexosamine pathway. Moreover, intravenous infusion of glucosamine could enhance hepatic apoM expression in rats, although serum apoM levels were not significantly influences. Conclusions It is concluded that both exogenous and endogenous glucosamine were essential for the over-expression of apoM, which may suggest that the increased intracellular content of glucosamine does not be responsible for the depressed apoM expression at hyperglycemia condition.
Collapse
Affiliation(s)
- Bo Jiang
- Department of Cardiothoracic Surgery in the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Xiaoying Zhang
- Department of Cardiothoracic Surgery in the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China.
| | - Dongmei Di
- Department of Cardiothoracic Surgery in the Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Guanghua Luo
- Comprehensive Laboratory, Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Yuanping Shi
- Comprehensive Laboratory, Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Jun Zhang
- Comprehensive Laboratory, Third Affiliated Hospital, Soochow University, Changzhou, 213003, China
| | - Maria Berggren-Söderlund
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, S-221 85, Lund, Sweden
| | - Peter Nilsson-Ehle
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, S-221 85, Lund, Sweden
| | - Ning Xu
- Division of Clinical Chemistry and Pharmacology, Department of Laboratory Medicine, Lund University, S-221 85, Lund, Sweden.
| |
Collapse
|
4
|
Drake KJ, Shotwell MS, Wikswo JP, Sidorov VY. Glutamine and glutamate limit the shortening of action potential duration in anoxia-challenged rabbit hearts. Physiol Rep 2015; 3:3/9/e12535. [PMID: 26333831 PMCID: PMC4600381 DOI: 10.14814/phy2.12535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
In clinical conditions, amino acid supplementation is applied to improve contractile function, minimize ischemia/reperfusion injury, and facilitate postoperative recovery. It has been shown that glutamine enhances myocardial ATP/APD (action potential duration) and glutathione/oxidized glutathione ratios, and can increase hexosamine biosynthesis pathway flux, which is believed to play a role in cardioprotection. Here, we studied the effect of glutamine and glutamate on electrical activity in Langendorff-perfused rabbit hearts. The hearts were supplied by Tyrode's media with or without 2.5 mmol/L glutamine and 150 μmol/L glutamate, and exposed to two 6-min anoxias with 20-min recovery in between. Change in APD was detected using a monophasic action potential probe. A nonlinear mixed-effects regression technique was used to evaluate the effect of amino acids on APD over the experiment. Typically, the dynamic of APD change encompasses three phases: short transient increase (more prominent in the first episode), slow decrease, and fast increase (starting with the beginning of recovery). The effect of both anoxic challenge and glutamine/glutamate was cumulative, being more pronounced in the second anoxia. The amino acids' protective effect became largest by the end of anoxia – 20.0% (18.9, 95% CI: [2.6 ms, 35.1 ms]), during the first anoxia and 36.6% (27.1, 95% CI: [7.7 ms, 46.6 ms]), during the second. Following the second anoxia, APD difference between control and supplemented hearts progressively increased, attaining 10.8% (13.6, 95% CI: [4.1 ms, 23.1 ms]) at the experiments' end. Our data reveal APD stabilizing and suggest an antiarrhythmic capacity of amino acid supplementation in anoxic/ischemic conditions.
Collapse
Affiliation(s)
- Kenneth J Drake
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee
| | - Matthew S Shotwell
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee
| | - John P Wikswo
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee Department of Physics and Astronomy, Vanderbilt University, Nashville, Tennessee Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Veniamin Y Sidorov
- Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University, Nashville, Tennessee Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
5
|
Joseph D, Kimar C, Symington B, Milne R, Essop MF. The detrimental effects of acute hyperglycemia on myocardial glucose uptake. Life Sci 2014; 105:31-42. [PMID: 24747137 DOI: 10.1016/j.lfs.2014.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/20/2014] [Accepted: 04/07/2014] [Indexed: 01/14/2023]
Abstract
AIMS Although acute hyperglycemic (AHG) episodes are linked to lower glucose uptake, underlying mechanisms remain unclear. We hypothesized that AHG triggers reactive oxygen species (ROS) production and increases non-oxidative glucose pathway (NOGP) activation, i.e. stimulation of advanced glycation end products (AGE), polyol pathway (PP), hexosamine biosynthetic pathway (HBP), PKC; thereby decreasing cardiac glucose uptake. MAIN METHODS H9c2 cardiomyoblasts were exposed to 25 mM glucose for 24h vs. 5.5mM controls ± modulating agents during the last hour of glucose exposure: a) antioxidant #1 for mitochondrial ROS (250 μM 4-OHCA), b) antioxidant #2 for NADPH oxidase-generated ROS (100 μM DPI), c) NOGP inhibitors - 100 μM aminoguanidine (AGE), 5 μM chelerythrine (PKC); 40 μM DON (HBP); and 10 μM zopolrestat (PP). ROS levels (mitochondrial, intracellular) and glucose uptake were evaluated by flow cytometry. KEY FINDINGS AHG elevated ROS, activated NOGPs and blunted glucose uptake. Transketolase activity (pentose phosphate pathway [PPP] marker) did not change. Respective 4-OHCA and DPI treatment blunted ROS production, diminished NOGP activation and normalized glucose uptake. NOGP inhibitory studies identified PKCβII as a key downstream player in lowering insulin-mediated glucose uptake. When we employed an agent (benfotiamine) known to shunt flux away from NOGPs (into PPP), it decreased ROS generation and NOGP activation, and restored glucose uptake under AHG conditions. SIGNIFICANCE This study demonstrates that AHG elicits maladaptive events that function in tandem to reduce glucose uptake, and that antioxidant treatment and/or attenuation of NOGP activation (PKC, polyol pathway) may limit the onset of insulin resistance.
Collapse
Affiliation(s)
- Danzil Joseph
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Charlene Kimar
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Burger Symington
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Robyn Milne
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - M Faadiel Essop
- Cardio-Metabolic Research Group (CMRG), Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa.
| |
Collapse
|
6
|
Hypoxia upregulates the expression of the O-linked N-acetylglucosamine containing epitope H in human ependymal cells. Pathol Res Pract 2010; 207:91-6. [PMID: 21145174 DOI: 10.1016/j.prp.2010.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2010] [Revised: 09/04/2010] [Accepted: 10/28/2010] [Indexed: 11/20/2022]
Abstract
Epitope H contains an O-linked N-acetylglucosamine (O-GlcNAc) residue in a specific conformation and/or environment recognized by mouse IgM monoclonal antibody H (mabH). Epitope H is present in several types of cells and in several polypeptides outside the CNS. Previous results have shown that in the adult human brains, epitope H is confined mostly to a minority of fibrous astrocytes, and it is greatly upregulated in the reactive astrocytes. Post-translational modification with O-GlcNAc occurs on many proteins involved in several cell processes, such as cell cycle progression, apoptosis, proteasome degradation pathways, and modulation of cellular function in response to nutrition and stress. Hypoxia is one of the major causes of cellular stress. Therefore, in this study, we used the mAbH and the indirect immunoperoxidase method to investigate the expression of epitope H in ependymal cells in brains of persons who died with signs of hypoxic encephalopathy. The results of the present study showed that practically all ependymal cells showed cytoplasmic staining for epitope H in supranuclear cytoplasm in the brain of two premature neonates and in ten infants who died with signs of hypoxic encephalopathy. However, the overwhelming majority of ependymal cells of the nine human embryos taken from legal abortions, ranging from 26 days until 13 weeks of gestational age, and of the ten infants' brains without any sign of hypoxic encephalopathy remained negative. Only occasionally did the ependymal cells show weak cytoplasmic staining in some foci. In addition, the reactive astrocytes in the hypoxic brains showed strong cytoplasmic staining, confirming previous results.
Collapse
|
7
|
Shikhman AR, Brinson DC, Valbracht J, Lotz MK. Differential metabolic effects of glucosamine and N-acetylglucosamine in human articular chondrocytes. Osteoarthritis Cartilage 2009; 17:1022-8. [PMID: 19332174 PMCID: PMC2785807 DOI: 10.1016/j.joca.2009.03.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 02/25/2009] [Accepted: 03/06/2009] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Aminosugars are commonly used to treat osteoarthritis; however, molecular mechanisms mediating their anti-arthritic activities are still poorly understood. This study analyzes facilitated transport and metabolic effects of glucosamine (GlcN) and N-acetylglucosamine (GlcNAc) in human articular chondrocytes. METHODS Human articular chondrocytes were isolated from knee cartilage. Facilitated transport of glucose, GlcN and GlcNAc was measured by uptake of [3H]2-deoxyglucose, [3H]GlcN and [3H]GlcNAc. Glucose transporter (GLUT) expression was analyzed by Western blotting. Production of sulfated glycosaminoglycans (SGAG) was measured using [(35)S]SO4. Hyaluronan was quantified using hyaluronan binding protein. RESULTS Chondrocytes actively import and metabolize GlcN but not GlcNAc and this represents a cell-type specific phenomenon. Similar to facilitated glucose transport, GlcN transport in chondrocytes is accelerated by cytokines and growth factors. GlcN non-competitively inhibits basal glucose transport, which in part depends on GlcN-mediated depletion of ATP stores. In IL-1beta-stimulated chondrocytes, GlcN inhibits membrane translocation of GLUT1 and 6, but does not affect the expression of GLUT3. In contrast to GlcN, GlcNAc accelerates facilitated glucose transport. In parallel with the opposing actions of these aminosugars on glucose transport, GlcN inhibits hyaluronan and SGAG synthesis while GlcNAc stimulates hyaluronan synthesis. GlcNAc-accelerated hyaluronan synthesis is associated with upregulation of hyaluronan synthase-2. CONCLUSION Differences in GlcN and GlcNAc uptake, and their subsequent effects on glucose transport, GLUT expression and SGAG and hyaluronan synthesis, indicate that these two aminosugars have distinct molecular mechanisms mediating their differential biological activities in chondrocytes.
Collapse
Affiliation(s)
- A R Shikhman
- Division of Arthritis Research, The Scripps Research Institute, Division of Rheumatology, Scripps Clinic, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
8
|
Abstract
An early and rapid response to severe injury or trauma is the development of hyperglycemia, which has long been thought to be an essential survival response by providing fuel for vital organ systems and facilitating mobilization of interstitial fluid reserves by increasing osmolarity. However, glucose can also be metabolized via the hexosamine biosynthesis pathway (HBP), leading to the synthesis of uridine diphosphate N-acetyl-glucosamine(UDP-GlcNAc). UDP-GlcNAc is a substrate for the addition, via an O-linkage, of a single N-acetylglucosamine to serine or threonine residues of nuclear and cytoplasmic proteins (O-glycosylation, O-GlcNAc). There is increasing appreciation that protein O-glycosylation is a highly dynamic posttranslational modification that plays a key role in signal transduction pathways. Sustained increases in O-GlocNAc have been implicated in the development of diabetes and diabetic complications; however, recent studies have demonstrated that stress leads to a transient increase in O-GlcNAc levels that is associated with increased tolerance to stress. Indeed, activation of pathways leading to O-GlcNAc formation improves cell survival after I/R injury, whereas inhibition of O-GlcNAc formation decreases cell survival. In addition, in rodent models of trauma-hemorrhage, increasing O-GlcNAc levels during resuscitation improves cardiac function and organ perfusion and attenuates the inflammatory response. At the cellular level, increasing O-GlcNAc levels attenuates nuclear factor-kappaB activation. It is noteworthy that other metabolic-based treatments for severe injury such as glucose-insulin-potassium and glutamine also lead to increased HBP flux and O-GlcNAc levels. The goal of this review is to summarize our current understanding of the role of the HBP and O-GlcNAc on the regulation of cell function and survival and to present evidence to support the notion that activation of these pathways represents a novel treatment strategy for severe injury and trauma.
Collapse
Affiliation(s)
- John C Chatham
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA.
| | | | | | | |
Collapse
|
9
|
Zhu A, Huang JB, Clark A, Romero R, Petty HR. 2,5-Deoxyfructosazine, a D-glucosamine derivative, inhibits T-cell interleukin-2 production better than D-glucosamine. Carbohydr Res 2007; 342:2745-9. [PMID: 17892867 PMCID: PMC2758268 DOI: 10.1016/j.carres.2007.08.025] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 08/28/2007] [Accepted: 08/30/2007] [Indexed: 10/22/2022]
Abstract
D-Glucosamine has been widely reported to have immunosuppressive actions on neutrophils, lymphocytes, and other cells of the immune system. However, under conditions used in biological experiments (e.g., neutral pH, and phosphate buffers), we have found that D-glucosamine self-reacts to form 2,5-deoxyfructosazine [2-(D-arabino-tetrahydroxybutyl)-5-(D-erythro-2,3,4-trihydroxybutyl)pyrazine] (1) and 2,5-fructosazine [2,5-bis(D-arabino-tetrahydroxybutyl)pyrazine] (2). When tested for bioactivity at nontoxic concentrations, these D-glucosamine derivatives were more effective inhibitors of IL-2 release from PHA-activated T cells than d-glucosamine. Hence, fructosazines constitute a novel class of immunomodulators.
Collapse
Affiliation(s)
- Aiping Zhu
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI 48105
| | - Ji-Biao Huang
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI 48105
| | - Andrea Clark
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI 48105
| | - Roberto Romero
- Department of Perinatology Research Branch, National Institute of Child Health and Human Development, Hutzel Hospital, 4707 St. Antoine Blvd., Detroit, MI 48201
| | - Howard R. Petty
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, Ann Arbor, MI 48105
- Department of Microbiology and Immunology, The University of Michigan Medical School, Ann Arbor, MI 48105
- Corresponding author. Tel.: 734-647-0384/fax: 734-936-3815/e-mail:
| |
Collapse
|
10
|
Renström F, Burén J, Svensson M, Eriksson JW. Insulin resistance induced by high glucose and high insulin precedes insulin receptor substrate 1 protein depletion in human adipocytes. Metabolism 2007; 56:190-8. [PMID: 17224332 DOI: 10.1016/j.metabol.2006.09.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2006] [Accepted: 09/08/2006] [Indexed: 11/25/2022]
Abstract
The aim of this study was to investigate whether high glucose and/or high insulin produces cellular insulin resistance in human adipocytes and, if so, to evaluate the time course and content of key proteins in the insulin signaling pathway. Subcutaneous fat biopsies were taken from 27 nondiabetic subjects. Insulin action in vitro was studied by measurement of glucose uptake after incubation at a physiologic glucose level (6 mmol/L) for 24 hours or with the last 2, 6, or 24 hours at a high glucose level (20 mmol/L) with or without high insulin (10(4)microU/mL). High glucose alone for 24 hours produced a small but significant impairment (by approximately 20%, P < .05) of insulin's effect to stimulate glucose transport, whereas nonstimulated glucose uptake was left intact. In contrast, the combination of high glucose and high insulin for 6 hours or more reduced basal glucose uptake by approximately 40% (P < .05). In addition, insulin-stimulated glucose uptake capacity was reduced by approximately 40% already after 2 hours (P < .05) and reached a maximal decline (by approximately 50%, P < .05) after a 6-hour culture in high glucose and high insulin. Treatment with high glucose and high insulin in combination for at least 6 hours reduced cellular insulin receptor substrate (IRS)-1, but not IRS-2, protein content by approximately 45% or more (P < .05). Moreover, after 24 hours, the ability of insulin to activate protein kinase B (ie, the phosphorylated protein kinase B [pPKB]-protein kinase B ratio) was decreased by approximately 50% (P < .05). No significant effects were seen on insulin signaling proteins or glucose transporter 4 after a long-term high-glucose culture. Culture with high insulin alone (and low glucose, 6 mmol/L) decreased basal and insulin-stimulated glucose uptake in conformity with the high-glucose/high-insulin setting. However, IRS-1 protein content remained unchanged. We conclude that, in adipocytes from healthy humans, high insulin alone for 2 hours or more decrease glucose uptake capacity. Likewise, high glucose and high insulin in combination for 2 hours or more decrease glucose uptake to the same extent as when cells were cultured with high insulin alone but, in addition, with a diminishment in IRS-1 protein lagging behind. Thus, IRS-1 depletion appears to be a secondary phenomenon in this model of insulin resistance. High glucose alone induces only a minor insulin resistance in human fat cells.
Collapse
Affiliation(s)
- Frida Renström
- Department of Public Health and Clinical Medicine, Medicine, Umeå University Hospital, S-901 85 Umeå, Sweden
| | | | | | | |
Collapse
|
11
|
Huang JB, Clark AJ, Petty HR. The hexosamine biosynthesis pathway negatively regulates IL-2 production by Jurkat T cells. Cell Immunol 2007; 245:1-6. [PMID: 17481598 PMCID: PMC3178408 DOI: 10.1016/j.cellimm.2007.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Revised: 03/21/2007] [Accepted: 03/26/2007] [Indexed: 10/23/2022]
Abstract
To test the hypothesis that the hexosamine biosynthesis pathway (HBP) affects cytokine production, we studied IL-2 production by Jurkat cells in response to PHA. We found that the HBP activator glucosamine (GlcN), but not glucose (Glc), dose-dependently reduced IL-2 production. Importantly, GlcN blocked trafficking of a GFP-NFAT chimeric protein to the nucleus of stimulated transfectants. Not surprisingly, changes in O-GlcNAc protein modifications were noted during cell activation with and without GlcN addition. These findings could not be explained by some non-specific change in cell metabolism because ATP concentrations did not significantly change. We speculate that HBP-active compounds may contribute to patient care in certain inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Ji-Biao Huang
- Department of Ophthalmology and Visual Sciences, The University of Michigan Medical School, 1000 Wall Street, Ann Arbor, MI 48105, USA
| | | | | |
Collapse
|
12
|
Matthews JA, Belof JL, Acevedo-Duncan M, Potter RL. Glucosamine-induced increase in Akt phosphorylation corresponds to increased endoplasmic reticulum stress in astroglial cells. Mol Cell Biochem 2006; 298:109-23. [PMID: 17136481 DOI: 10.1007/s11010-006-9358-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Accepted: 10/25/2006] [Indexed: 11/28/2022]
Abstract
Increased glucose flux through the hexosamine biosynthetic pathway (HBP) is known to affect the activity of a number of signal transduction pathways and lead to insulin resistance. Although widely studied in insulin responsive tissues, the effect of increased HBP activity on largely insulin unresponsive tissues, such as the brain, remains relatively unknown. Herein, we investigate the effects of increased HBP flux on Akt activation in a human astroglial cells line using glucosamine, a compound commonly used to mimic hyperglycemic conditions by increasing HBP flux. Cellular treatment with 8 mM glucosamine resulted in a 96.8% +/- 24.6 increase in Akt phosphorylation after 5 h of treatment that remained elevated throughout the 9-h time course. Glucosamine treatment also resulted in modest increases in global levels of the O-GlcNAc protein modification. Increasing O-GlcNAc levels using the O-GlcNAcase inhibitor streptozotocin (STZ) also increased Akt phosphorylation by 96.8% +/- 11.0 after only 3 h although for a shorter duration than glucosamine; however, the more potent O-GlcNAcase inhibitors O-(2-acetamido-2-deoxy-D-glucopyranosylidene)amino-N-phenylcarbamate (PUGNAc) and 1,2-dideoxy-2'-propyl-alpha-D-glucopyranoso-[2,1-d]-Delta2'-thiazoline (NAGBT) failed to mimic the increases in phospho-Akt indicating that the Akt phosphorylation is not a result of increased O-GlcNAc protein modification. Further analysis indicated that this increased phosphorylation was also not due to increased osmotic stress and was not attenuated by N-acetylcysteine eliminating the potential role of oxidative stress in the observed phospho-Akt increases. Glucosamine treatment, but not STZ treatment, did correlate with a large increase in the expression of the endoplasmic reticulum (ER) stress marker GRP 78. Altogether, these results indicate that increased HBP flux in human astroglial cells results in a rapid, short-term phosphorylation of Akt that is likely a result of increased ER stress. The mechanism by which STZ increases Akt phosphorylation, however, remains unknown.
Collapse
Affiliation(s)
- J Aaron Matthews
- Department of Chemistry, University of South Florida, 4202 East Fowler Ave, SCA 400, Tampa, FL 33620, USA
| | | | | | | |
Collapse
|
13
|
Zachara NE, Hart GW. Cell signaling, the essential role of O-GlcNAc! Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:599-617. [PMID: 16781888 DOI: 10.1016/j.bbalip.2006.04.007] [Citation(s) in RCA: 295] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2005] [Revised: 04/20/2006] [Accepted: 04/24/2006] [Indexed: 11/28/2022]
Abstract
An increasing body of evidence points to a central regulatory role for glucose in mediating cellular processes and expands the role of glucose well beyond its traditional role(s) in energy metabolism. Recently, it has been recognized that one downstream effector produced from glucose is UDP-GlcNAc. Levels of UDP-GlcNAc, and the subsequent addition of O-linked beta-N-acetylglucosamine (O-GlcNAc) to Ser/Thr residues, is involved in regulating nuclear and cytoplasmic proteins in a manner analogous to protein phosphorylation. O-GlcNAc protein modification is essential for life in mammalian cells, highlighting the importance of this simple post-translational modification in basic cellular regulation. Recent research has highlighted key roles for O-GlcNAc serving as a nutrient sensor in regulating insulin signaling, the cell cycle, and calcium handling, as well as the cellular stress response.
Collapse
Affiliation(s)
- Natasha E Zachara
- Department of Biological Chemistry, Johns Hopkins Singapore, 31 Biopolis Way, #02-01 The Nanos, 138669 Singapore
| | | |
Collapse
|
14
|
|
15
|
Renström F, Burén J, Eriksson JW. Insulin receptor substrates-1 and -2 are both depleted but via different mechanisms after down-regulation of glucose transport in rat adipocytes. Endocrinology 2005; 146:3044-51. [PMID: 15845625 DOI: 10.1210/en.2004-1675] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Alterations in muscle and adipose tissue insulin receptor substrate (IRS)-1 and IRS-2 are associated with, and commonly believed to contribute to, development of insulin resistance. In this study, we investigated the mechanisms behind previously observed reductions in IRS levels due to high concentrations of glucose and insulin and their significance in the impairment of glucose uptake capacity in primary rat adipocytes. Semiquantitative RT-PCR analysis showed that insulin (10(4) microU/ml) alone or in combination with glucose (15 mm) markedly suppressed IRS-2 gene expression, whereas IRS-1 mRNA was unaffected by the culture conditions. The negative effect of a high glucose/high insulin setting on IRS-1 protein level was still exerted when protein synthesis was inhibited with cycloheximide. Impairment of glucose uptake capacity after treatment with high glucose and insulin was most pronounced after 3 h, whereas IRS-1 and IRS-2 protein levels were unaffected up to 6 h but were reduced after 16 h. Moreover, impaired glucose uptake capacity could only partially be reversed by subsequent incubation at physiological conditions. These novel results suggest that: 1) in a high glucose/high insulin setting depletion of IRS-1 and IRS-2 protein, respectively, occurs via different mechanisms, and IRS-2 gene expression is suppressed, whereas IRS-1 depletion is due to posttranslational mechanisms; 2) IRS-1 and IRS-2 protein depletion is a secondary event in the development of insulin resistance in this model of hyperglycemia/hyperinsulinemia; and 3) depletion of cellular IRS in adipose tissue may be a consequence rather than a cause of insulin resistance and hyperinsulinemia in type 2 diabetes.
Collapse
Affiliation(s)
- Frida Renström
- Department of Medicine, Umeå University Hospital, SE-901 85 Umeå, Sweden
| | | | | |
Collapse
|
16
|
Fujita T, Furukawa S, Morita K, Ishihara T, Shiotani M, Matsushita Y, Matsuda M, Shimomura I. Glucosamine induces lipid accumulation and adipogenic change in C2C12 myoblasts. Biochem Biophys Res Commun 2005; 328:369-74. [PMID: 15694357 DOI: 10.1016/j.bbrc.2004.12.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Indexed: 11/28/2022]
Abstract
Hyperglycemia-induced activation of hexosamine biosynthesis pathway (HBP) has been implicated in the development of insulin resistance in skeletal muscles. In the present study, the content of uridine-5'-diphospho-N-acetylglucosamine, the end product of the HBP, was elevated in skeletal muscle of obese diabetic KKA(y) mice, compared with control mice. To elucidate the effect of elevated HBP in the skeletal muscle, we treated C2C12 myoblasts with glucosamine, an intermediate metabolite of the HBP. Glucosamine induced lipid accumulation and significantly increased the mRNA expression levels of peroxisome proliferator-activated receptor gamma, adiponectin, and aP2 in C2C12 myoblasts. Similar mRNA changes were observed in skeletal muscles of Sprague-Dawley rats treated with glucosamine infusion. Our results provide a possible explanation of hyperglycemia-induced insulin resistance in skeletal muscle.
Collapse
Affiliation(s)
- Takuya Fujita
- Department of Medicine and Pathophysiology, Graduate School of Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Marshall S, Yamasaki K, Okuyama R. Glucosamine induces rapid desensitization of glucose transport in isolated adipocytes by increasing GlcN-6-P levels. Biochem Biophys Res Commun 2005; 329:1155-61. [PMID: 15752775 DOI: 10.1016/j.bbrc.2005.02.084] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Indexed: 11/19/2022]
Abstract
We have examined the hypothesis that glucosamine (GlcN) can rapidly induce insulin resistance through an allosteric mechanism. When insulin-treated adipocytes were exposed to 2mM GlcN, glucose uptake was rapidly reduced by approximately 60% with a T(1/2) of 2 min. We also observed an increase in intracellular GlcN-6-P (at 5 min) from undetectable levels to approximately 260 nmol/g. Continued GlcN treatment resulted in additional accumulation of GlcN-6-P (>1200 nmol/g at 2h), but caused no further decrease in glucose uptake. Although the acute inhibitory action of GlcN could be completely reversed by removing extracellular GlcN, a slow and progressive decrease in insulin-stimulated glucose transport was observed with longer treatment times (T(1/2) of 45 min, 62% loss by 5h). From these data, we conclude that: (1) GlcN elevates intracellular GlcN-6-P levels within minutes, resulting in desensitization of the glucose transport system through allosteric inhibition of hexokinase; (2) prolonged treatment elevates GlcN-6-P to levels that cannot be effectively lowered by cell washing; and (3) residual levels of GlcN-6-P continue to allosterically inhibit glucose uptake, resulting in a slower rate of desensitization that is temporally similar to glucose-induced desensitization, but mechanistically different.
Collapse
|
18
|
Matthews JA, Acevedo-Duncan M, Potter RL. Selective decrease of membrane-associated PKC-alpha and PKC-epsilon in response to elevated intracellular O-GlcNAc levels in transformed human glial cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2004; 1743:305-15. [PMID: 15843043 DOI: 10.1016/j.bbamcr.2004.11.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Revised: 10/19/2004] [Accepted: 11/03/2004] [Indexed: 11/19/2022]
Abstract
Increased flux through the hexosamine biosynthetic pathway (HBP) has been shown to affect the activity and translocation of certain protein kinase C (PKC) isoforms. It has been suggested that this effect is due to increases in the beta-O-linked N-acetylglucosamine (O-GlcNAc) modification. Herein, we demonstrate the effect of increasing the O-GlcNAc modification on the translocation of select PKC isozymes in a human astroglial cell line. Treating cells with either 8 mM d-glucosamine (GlcN), 5 mM streptozotocin (STZ), or 80 muM O-(2-acetamido-2-deoxy-d-glucopyranosylidene)amino-N-phenylcarbamate (PUGNAc) produced a significant increase in the O-GlcNAc modification on both cytosolic and membrane proteins; however, both the level and rate of O-GlcNAc increase varied with the compound. GlcN treatment resulted in a rapid, transient translocation of PKC-betaII that was maximal after 3 h (73+/-8%) and also produced a 48+/-15% decrease in membrane-associated PKC-epsilon after 9 h of treatment. Similar to GlcN treatment, STZ and PUGNAc treatment also resulted in decreased levels of PKC-epsilon in the membrane fraction. Significant decreases were seen as early as 5 h and, by 9 h of treatment, had decreased by 87+/-6% with STZ and 73+/-7% with PUGNAc. Unlike GlcN, both STZ and PUGNAc produced a decrease in PKC-alpha membrane levels by 9 h posttreatment (78+/-10% with STZ and 66+/-8% with PUGNAc) while neither compound produced any changes in PKC-betaII translocation. In addition, none of the three compounds affected membrane levels of PKC-iota. Altogether, these results demonstrate a novel link between increased levels of the O-GlcNAc modification and the regulation of specific PKC isoforms.
Collapse
Affiliation(s)
- Jason A Matthews
- Department of Chemistry, University of South Florida, 4202 East Fowler Ave, SCA 400, Tampa, FL, 33620, USA
| | | | | |
Collapse
|
19
|
Lamont BJ, Andrikopoulos S, Funkat A, Favaloro J, Ye JM, Kraegen EW, Howlett KF, Zajac JD, Proietto J. Peripheral insulin resistance develops in transgenic rats overexpressing phosphoenolpyruvate carboxykinase in the kidney. Diabetologia 2003; 46:1338-47. [PMID: 12898008 DOI: 10.1007/s00125-003-1180-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2003] [Revised: 05/26/2003] [Indexed: 12/22/2022]
Abstract
AIMS/HYPOTHESIS To study the secondary consequences of impaired suppression of endogenous glucose production (EGP) we have created a transgenic rat overexpressing the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK) in the kidney. The aim of this study was to determine whether peripheral insulin resistance develops in these transgenic rats. METHODS Whole body rate of glucose disappearance (R(d)) and endogenous glucose production were measured basally and during a euglycaemic/hyperinsulinaemic clamp in phosphoenolpyruvate carboxykinase transgenic and control rats using [6-(3)H]-glucose. Glucose uptake into individual tissues was measured in vivo using 2-[1-(14)C]-deoxyglucose. RESULTS Phosphoenolpyruvate carboxykinase transgenic rats were heavier and had increased gonadal and infrarenal fat pad weights. Under basal conditions, endogenous glucose production was similar in phosphoenolpyruvate carboxykinase transgenic and control rats (37.4+/-1.1 vs 34.6+/-2.6 micromol/kg/min). Moderate hyperinsulinaemia (810 pmol/l) completely suppressed EGP in control rats (-0.6+/-5.5 micromol/kg/min, p<0.05) while there was no suppression in phosphoenolpyruvate carboxykinase rats (45.2+/-7.9 micromol/kg/min). Basal R(d) was comparable between PEPCK transgenic and control rats (37.4+/-1.1 vs 34.6+/-2.6 micromol/kg/min) but under insulin-stimulated conditions the increase in R(d) was greater in control compared to phosphoenolpyruvate carboxykinase transgenic rats indicative of insulin resistance (73.4+/-11.2 vs 112.0+/-8.0 micromol/kg/min, p<0.05). Basal glucose uptake was reduced in white and brown adipose tissue, heart and soleus while insulin-stimulated transport was reduced in white and brown adipose tissue, white quadriceps, white gastrocnemius and soleus in phosphoenolpyruvate carboxykinase transgenic compared to control rats. The impairment in both white and brown adipose tissue glucose uptake in phosphoenolpyruvate carboxykinase transgenic rats was associated with a decrease in GLUT4 protein content. In contrast, muscle GLUT4 protein, triglyceride and long-chain acylCoA levels were comparable between PEPCK transgenic and control rats. CONCLUSIONS/INTERPRETATION A primary defect in suppression of EGP caused adipose tissue and muscle insulin resistance.
Collapse
Affiliation(s)
- B J Lamont
- University of Melbourne Department of Medicine, Royal Melbourne Hospital, 3050 Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Bosch RR, Janssen SWJ, Span PN, Olthaar A, van Emst-de Vries SE, Willems PHGM, Martens J M G, Hermus ARMM, Sweep CCJ. Exploring levels of hexosamine biosynthesis pathway intermediates and protein kinase C isoforms in muscle and fat tissue of Zucker Diabetic Fatty rats. Endocrine 2003; 20:247-52. [PMID: 12721503 DOI: 10.1385/endo:20:3:247] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2002] [Revised: 12/02/2002] [Accepted: 01/09/2003] [Indexed: 11/11/2022]
Abstract
Many studies suggest that insulin resistance develops and/or is maintained by an increased flux of glucose through the hexosamine biosynthesis pathway. This pathway may attenuate insulin-stimulated glucose uptake by activating protein kinase C (PKC). Therefore, we investigated whether the concentrations of the major hexosamine metabolites, uridine diphosphate- N-acetyl-glucosamine (UDP-GlcNAc) and uridine diphosphate- N-acetyl-galactosamine (UDP-GalNAc), and the expression levels of PKC isoforms were affected in Zucker Diabetic Fatty (ZDF) rats, an animal model widely used to study type 2 diabetes mellitus. At the age of 6 wk, control and ZDF rats were normoglycemic. Whereas control rats remained normoglycemic, the ZDF rats became hyperglycemic. The amount of UDP-GlcNAc and UDP-GalNAc in muscle tissue of ZDF rats was similar at 6, 12, 18, and 24 wk of age. Moreover, the concentration of both hexosamines did not differ among ZDF, phlorizin-treated ZDF, and control rats. Western blot analysis revealed that PKCalpha, delta, epsilon, andzeta, but not PKCbeta and gamma, were expressed in muscle and fat tissues from 6- and 24-wk-old control and ZDF rats. In addition, we did not observe changes in the expression levels of the PKC isoforms following prolonged hyperglycemia. Taken together, these findings indicate that the amounts of several metabolites from the hexosamine biosynthesis pathway and PKC isoforms, both hypothesized to be important in the development and/or maintenance of the insulin-resistant state of muscle and fat tissue, are not different in ZDF compared with nondiabetic rats.
Collapse
Affiliation(s)
- Remko R Bosch
- Department of Chemical Endocrinology, University Medical Centre Nijmegen, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Burt DJ, Gruden G, Thomas SM, Tutt P, Dell'Anna C, Viberti GC, Gnudi L. P38 mitogen-activated protein kinase mediates hexosamine-induced TGFbeta1 mRNA expression in human mesangial cells. Diabetologia 2003; 46:531-7. [PMID: 12679868 DOI: 10.1007/s00125-003-1075-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2002] [Revised: 09/12/2002] [Indexed: 10/20/2022]
Abstract
AIMS/HYPOTHESIS The hexosamine pathway has been implicated in the induction of TGFbeta1 expression and in the pathophysiology of diabetic glomerulopathy. Glucose-induced TGFbeta1 expression is mediated by p38 mitogen-activated-protein-kinase (p38-MAPK) and this kinase is activated in the diabetic glomeruli. We examined whether the p38-MAPK is implicated in hexosamine-induced TGFbeta1 mRNA expression in human mesangial cells. GFAT overexpression induced an increase in p38-MAPK activation after 6 and 12 h incubation in normal glucose, and this was prevented by the GFAT inhibitor azaserine. Furthermore, high glucose enhanced p38-MAPK activation in GFAT tranfected cells ( p</=0.04). P38-MAPK inhibition using SB202190 (1 micro mol/l) reduced hexosamine-induced TGFbeta1 expression in normal and high glucose. The activation of the p38-MAPK was dependent on protein kinase-C. METHODS The products of the hexosamine biosynthetic pathway were increased by the addition of glucosamine or by the overexpression of the rate-limiting enzyme of the hexosamine pathway, glutamine: fructose-6-phosphate amidotransferase (GFAT). RESULTS Glucosamine addition resulted in cell death. UDP-N-Acetylglucosamine, one of the major hexosamine end-products, was increased in normal (7 mmol/l) and high (25 mmol/l) glucose conditions in GFAT-transfected cells compared to control transfected cells by twofold and 1.7-fold respectively ( p</=0.04) and this was accompanied by a 1.6- and 2.3-fold increase ( p</=0.02) in TGFbeta1 mRNA expression. Addition of the GFAT inhibitor azaserine (10 micro mol/l) prevented the induction of TGFbeta1 in GFAT transfected cells. CONCLUSION/INTERPRETATION Overexpression of GFAT increases hexosamine accumulation which mediates TGFbeta1 expression via a protein kinase-C and p38-MAPK dependent mechanism. Increased glucose concentrations magnify these effects.
Collapse
Affiliation(s)
- D J Burt
- Department of Diabetes, Endocrinology and Internal Medicine, Guys Hospital, King's College, 5th Floor Thomas Guy House, SE1 9RT, London, UK
| | | | | | | | | | | | | |
Collapse
|
22
|
Burén J, Lindmark S, Renström F, Eriksson JW. In vitro reversal of hyperglycemia normalizes insulin action in fat cells from type 2 diabetes patients: is cellular insulin resistance caused by glucotoxicity in vivo? Metabolism 2003; 52:239-45. [PMID: 12601640 DOI: 10.1053/meta.2003.50041] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Chronic hyperglycemia promotes the development of insulin resistance. The aim of this study was to investigate whether cellular insulin resistance is secondary to the diabetic state in human type 2 diabetes. Subcutaneous fat biopsies were taken from 3 age-, sex-, and body mass index (BMI)-matched groups with 10 subjects in each group: type 2 diabetes patients with either good (hemoglobin A(1c) [HbA(1c)] < 7%, G) or poor (HbA(1c) > 7.5%, P) metabolic control and healthy control subjects (C). Insulin action in vitro was studied by measurements of glucose uptake both directly after cell isolation and following a 24-hour incubation at a physiological glucose level (6 mmol/L). The relationship with insulin action in vivo was addressed by employing the euglycemic clamp technique. Freshly isolated fat cells from type 2 diabetes patients with poor metabolic control had approximately 55% lower maximal insulin response (1,000 microU/mL) on glucose uptake (P <.05) compared to C. Cells from P were more insulin-resistant (P <.05) than cells from G at a low (5 microU/mL) but not at a high (1,000 microU/mL) insulin concentration, suggesting insulin insensitivity. However, following 24 hours of incubation at physiological glucose levels, insulin resistance was completely reversed in the diabetes cells and no differences in insulin-stimulated glucose uptake were found among the 3 groups. Insulin sensitivity in vivo assessed with hyperinsulinemic, euglycemic clamp (M-value) was significantly associated with insulin action on glucose uptake in fresh adipocytes in vitro (r = 0.50, P <.01). Fasting blood glucose at the time of biopsy and HbA(1c), but not serum insulin, were negatively correlated to insulin's effect to stimulate glucose uptake in vitro (r = -0.36, P =.064 and r = - 0.41, P <.05, respectively) in all groups taken together. In the in vivo situation, fasting blood glucose, HbA(1c), and serum insulin were all negatively correlated to insulin sensitivity (M-value; r = -0.62, P<.001, r= -0.61, P<.001, and r = -0.56, p <.01, respectively). Cell size, waist-to-hip ration (WHR), and BMI correlated negatively with insulin's effect to stimulate glucose uptake both in vitro (r = -0.55, P <.01, r = -0.54, P <.01, and r = -0.43, P <.05, respectively) and in vivo (r = -0.43, P <.05, r = -0.50, P <.01, and r = -0.36, P <.05, respectively). Multiple regression analyses revealed that adipocyte cell size and WHR independently predicted insulin resistance in vitro. Furthermore, insulin sensitivity in vivo could be predicted by fasting blood glucose and serum insulin levels. We conclude that insulin resistance in fat cells from type 2 diabetes patients is fully reversible following incubation at physiological glucose concentrations. Thus, cellular insulin resistance may be mainly secondary to the hyperglycemic state in vivo.
Collapse
Affiliation(s)
- Jonas Burén
- Department of Medicine, Umeå University Hospital, Umeå, Sweden
| | | | | | | |
Collapse
|
23
|
Goldberg HJ, Whiteside CI, Fantus IG. The hexosamine pathway regulates the plasminogen activator inhibitor-1 gene promoter and Sp1 transcriptional activation through protein kinase C-beta I and -delta. J Biol Chem 2002; 277:33833-41. [PMID: 12105191 DOI: 10.1074/jbc.m112331200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increased flux through the hexosamine biosynthesis pathway (HBP) has been shown to stimulate the expression of a number of genes. We previously demonstrated in glomerular mesangial and endothelial cells that both high glucose concentrations and glucosamine activated the plasminogen activator inhibitor-1 (PAI-1) gene promoter through the transcription factor, Sp1; and that the glutamine:fructose-6-phosphate amidotransferase inhibitor, 6-diazo-5-oxonorleucine, inhibited the effect of high glucose, but not that of glucosamine. Here, we examined the role of protein kinase C (PKC) isoforms in the regulation of the PAI-1 promoter and Sp1 transcriptional activity by the HBP. In transient transfections, exposure to 2 mm glucosamine or 20 mm glucose for 4 days increased the activities of a PAI-1 promoter-luciferase reporter gene as well as the Sp1 transcriptional activation domain fused to the GAL4 DNA-binding domain cotransfected with a GAL4 promoter-luciferase reporter. Cotransfected dominant negative PKC-betaI and -delta completely blocked the induction of PAI-1 promoter transcription by both sugars, whereas only dominant negative PKC-betaI interfered with Sp1-GAL4 activation. Both glucosamine and high glucose stimulated the in vitro kinase activity of immunoprecipitated PKC-betaI and -delta. Furthermore, 6-diazo-5-oxonorleucine suppressed high glucose-induced PKC kinase activity and Sp1-GAL4 transcriptional activation. These findings demonstrate a requirement for the PKC-betaI and -delta signal transduction pathways in HBP-induced transcription.
Collapse
Affiliation(s)
- Howard J Goldberg
- Department of Medicine, Mount Sinai Hospital and University Health Network, 600 University Avenue, Suite 780, Toronto, Ontario M5G 1X5, Canada
| | | | | |
Collapse
|
24
|
Filippis C, Filippis A, Clark S, Proietto J. Activation of PI 3-kinase by the hexosamine biosynthesis pathway. Mol Cell Endocrinol 2002; 194:29-37. [PMID: 12242025 DOI: 10.1016/s0303-7207(02)00213-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
It has been shown that hyperglycaemia-induced defects in glucose transport and insulin action are mediated by increased flux of excess glucose through the hexosamine biosynthesis pathway (HBP). We have previously demonstrated that in rat adipocytes, increased flux through the HBP activates protein kinase C (PKC). The aim of the present study was to explore the mechanism for HBP-mediated activation of PKC. We show that activation of the HBP by either high glucose or glucosamine causes the translocation of PKC-zeta/lambda and PKC-epsilon but not other PKC isoforms tested (alpha, beta, delta). This translocation was inhibited by wortmannin, a PI 3-kinase inhibitor. Both high glucose and glucosamine caused widespread cellular activation of PI 3-kinase. We demonstrate that HBP-mediated activation of PI 3-kinase has an insulin-like effect to translocate GLUT4. We conclude that an acute increase of glucose flux through the HBP activates PI 3-kinase.
Collapse
Affiliation(s)
- Christine Filippis
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | | | | | | |
Collapse
|
25
|
Milewski S. Glucosamine-6-phosphate synthase--the multi-facets enzyme. BIOCHIMICA ET BIOPHYSICA ACTA 2002; 1597:173-92. [PMID: 12044898 DOI: 10.1016/s0167-4838(02)00318-7] [Citation(s) in RCA: 201] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
L-Glutamine: D-fructose-6-phosphate amidotransferase, known under trivial name of glucosamine-6-phosphate synthase, as the only member of the amidotransferase subfamily of enzymes, does not display any ammonia-dependent activity. This enzyme, catalysing the first committed step in a pathway leading to the eventual formation of uridine 5'-diphospho-N-acetyl-D-glucosamine (UDP-GlcNAc), is an important point of metabolic control in biosynthesis of amino sugar-containing macromolecules. The molecular mechanism of reaction catalysed by GlcN-6-P synthase is complex and involves both amino transfer and sugar isomerisation. Substantial alterations to the enzyme structure and properties have been detected in different neoplastic tissues. GlcN-6-P synthase is inflicted in phenomenon of hexosamine-induced insulin resistance in diabetes. Finally, this enzyme has been proposed as a promising target in antifungal chemotherapy. Most of these issues, especially their molecular aspects, have been extensively studied in recent years. This article provides a comprehensive overview of the present knowledge on this multi-facets enzyme.
Collapse
Affiliation(s)
- Sławomir Milewski
- Department of Pharmaceutical Technology and Biochemistry, Technical University of Gdańsk, ul. Narutowicza 11/12, 80-952 Gdańsk, Poland.
| |
Collapse
|
26
|
Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr Rev 2002; 23:201-29. [PMID: 11943743 DOI: 10.1210/edrv.23.2.0461] [Citation(s) in RCA: 767] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The primary genetic, environmental, and metabolic factors responsible for causing insulin resistance and pancreatic beta-cell failure and the precise sequence of events leading to the development of type 2 diabetes are not yet fully understood. Abnormalities of triglyceride storage and lipolysis in insulin-sensitive tissues are an early manifestation of conditions characterized by insulin resistance and are detectable before the development of postprandial or fasting hyperglycemia. Increased free fatty acid (FFA) flux from adipose tissue to nonadipose tissue, resulting from abnormalities of fat metabolism, participates in and amplifies many of the fundamental metabolic derangements that are characteristic of the insulin resistance syndrome and type 2 diabetes. It is also likely to play an important role in the progression from normal glucose tolerance to fasting hyperglycemia and conversion to frank type 2 diabetes in insulin resistant individuals. Adverse metabolic consequences of increased FFA flux, to be discussed in this review, are extremely wide ranging and include, but are not limited to: 1) dyslipidemia and hepatic steatosis, 2) impaired glucose metabolism and insulin sensitivity in muscle and liver, 3) diminished insulin clearance, aggravating peripheral tissue hyperinsulinemia, and 4) impaired pancreatic beta-cell function. The precise biochemical mechanisms whereby fatty acids and cytosolic triglycerides exert their effects remain poorly understood. Recent studies, however, suggest that the sequence of events may be the following: in states of positive net energy balance, triglyceride accumulation in "fat-buffering" adipose tissue is limited by the development of adipose tissue insulin resistance. This results in diversion of energy substrates to nonadipose tissue, which in turn leads to a complex array of metabolic abnormalities characteristic of insulin-resistant states and type 2 diabetes. Recent evidence suggests that some of the biochemical mechanisms whereby glucose and fat exert adverse effects in insulin-sensitive and insulin-producing tissues are shared, thus implicating a diabetogenic role for energy excess as a whole. Although there is now evidence that weight loss through reduction of caloric intake and increase in physical activity can prevent the development of diabetes, it remains an open question as to whether specific modulation of fat metabolism will result in improvement in some or all of the above metabolic derangements or will prevent progression from insulin resistance syndrome to type 2 diabetes.
Collapse
Affiliation(s)
- Gary F Lewis
- Department of Medicine, Division of Endocrinology, University of Toronto, Canada M5G 2C4.
| | | | | | | |
Collapse
|
27
|
Tannock LR, Little PJ, Wight TN, Chait A. Arterial smooth muscle cell proteoglycans synthesized in the presence of glucosamine demonstrate reduced binding to LDL. J Lipid Res 2002. [DOI: 10.1016/s0022-2275(20)30198-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
28
|
Kawanaka K, Han DH, Gao J, Nolte LA, Holloszy JO. Development of glucose-induced insulin resistance in muscle requires protein synthesis. J Biol Chem 2001; 276:20101-7. [PMID: 11274201 DOI: 10.1074/jbc.m010599200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Muscles and fat cells develop insulin resistance when exposed to high concentrations of glucose and insulin. We used an isolated muscle preparation incubated with high levels of glucose and insulin to further evaluate how glucose-induced insulin resistance (GIIR) is mediated. Incubation with 2 milliunits/ml insulin and 36 mm glucose for 5 h resulted in an approximately 50% decrease in insulin-stimulated muscle glucose transport. The decrease in insulin responsiveness of glucose transport induced by glucose was not due to impaired insulin signaling, as insulin-stimulated phosphatidylinositol 3-kinase activity and protein kinase B phosphorylation were not reduced. It has been hypothesized that entry of glucose into the hexosamine biosynthetic pathway with accumulation of UDP-N-acetylhexosamines (UDP-HexNAcs) mediates GIIR. However, inhibition of the rate-limiting enzyme GFAT (glutamine:fructose-6-phosphate amidotransferase) did not protect against GIIR despite a marked reduction of UDP-HexNAcs. The mRNA synthesis inhibitor actinomycin D and the protein synthesis inhibitor cycloheximide both completely protected against GIIR despite the massive increases in UDP-HexNAcs and glycogen that resulted from increased glucose entry. Activation of AMP-activated protein kinase also protected against GIIR. These results provide evidence that GIIR can occur in muscle without increased accumulation of hexosamine pathway end products, that neither high glycogen concentration nor impaired insulin signaling is responsible for GIIR, and that synthesis of a protein with a short half-life mediates GIIR. They also suggest that dephosphorylation of a transcription factor may be involved in the induction of GIIR.
Collapse
Affiliation(s)
- K Kawanaka
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | | | |
Collapse
|
29
|
Schmitz-Peiffer C. Signalling aspects of insulin resistance in skeletal muscle: mechanisms induced by lipid oversupply. Cell Signal 2000; 12:583-94. [PMID: 11080610 DOI: 10.1016/s0898-6568(00)00110-8] [Citation(s) in RCA: 179] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A reduced capacity for insulin to elicit increases in glucose uptake and metabolism in target tissues such as skeletal muscle is a common feature of obesity and diabetes. The association between lipid oversupply and such insulin resistance is well established, and evidence for mechanisms through which lipids could play a causative role in the generation of muscle insulin resistance is reviewed. While the effects of lipids may in part be mediated by substrate competition through the glucose-fatty acid cycle, interference with insulin signal transduction by lipid-activated signalling pathways is also likely to play an important role. Thus, studies of insulin resistance in Type 2 diabetes, obesity, fat-fed animals and lipid-treated cells have identified defects both at the level of insulin receptor-mediated tyrosine phosphorylation and at downstream sites such as protein kinase B (PKB) activation. Lipid signalling molecules can be derived from free fatty acids, and include diacylglycerol, which activates isozymes of the protein kinase C (PKC) family, and ceramide, which has several effectors including PKCs and a protein phosphatase. In addition, elevated lipid availability can increase flux through the hexosamine biosynthesis pathway which can also lead to activation of PKC as well as protein glycosylation and modulation of gene expression. The mechanisms giving rise to decreased insulin signalling include serine/threonine phosphorylation of insulin receptor substrate-1, but also direct inhibition of components such as PKB. Thus lipids can inhibit glucose disposal by causing interference with insulin signal transduction, and most likely by more than one pathway depending on the prevalent species of fatty acids.
Collapse
Affiliation(s)
- C Schmitz-Peiffer
- Garvan Institute of Medical Research, 384 Victoria Street, NSW 2010, Darlinghurst, Australia.
| |
Collapse
|
30
|
Schleicher ED, Weigert C. Role of the hexosamine biosynthetic pathway in diabetic nephropathy. KIDNEY INTERNATIONAL. SUPPLEMENT 2000; 77:S13-8. [PMID: 10997685 DOI: 10.1046/j.1523-1755.2000.07703.x] [Citation(s) in RCA: 168] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The hexosamine biosynthetic pathway has been hypothesized to be involved in the development of insulin resistance and diabetic vascular complications. In particular, it was demonstrated that hyperglycemia-induced production of transforming growth factor-beta (TGF-beta1), a prosclerotic cytokine causally involved in the development of diabetic nephropathy. Several lines of evidence indicate that TGF-beta1 induction is mediated by the hexosamine pathway. In cultured mesangial cells, high glucose levels induce TGF-beta1 production. This effect is eliminated by inhibition of glutamine: fructose-6-phosphate-amidotransferase (GFAT), the rate-limiting enzyme of this pathway. Furthermore, stable overexpression of GFAT increased levels of TGF-beta1 protein, mRNA, and promoter activity. Inasmuch as stimulation or inhibition of GFAT increased or decreased high glucose-stimulated activity of protein kinase C (PKC), respectively, the observed effects appear to be transduced by PKC. In similar experiments, involvement of the hexosamine pathway in hyperglycemia-induced production of cytokines (TGF-alpha and basic fibroblast growth factor [bFGF]) was demonstrated in vascular smooth muscle cells. These studies also revealed a rapid increase in GFAT activity by treatment with agents that elevated levels of cyclic adenosine 3',5' monophosphate (cAMP), thus indicating that GFAT activity is tightly regulated by cAMP-dependent phosphorylation. Using immunohistochemistry and in situ hybridization, high expression of GFAT was found in human adipocytes, skeletal muscle, vascular smooth muscle cells, and renal tubular epithelial cells. whereas glomerular cells remained essentially unstained. However, significant staining occurred in glomerular cells of patients with diabetic nephropathy. Current data indicate that the flux through the hexosamine pathway, regulated by GFAT, may be causally involved in the development of diabetic vascular disease, particularly diabetic nephropathy.
Collapse
Affiliation(s)
- E D Schleicher
- Department of Internal Medicine, University of Tübingen, Germany.
| | | |
Collapse
|
31
|
Ross SA, Chen X, Hope HR, Sun S, McMahon EG, Broschat K, Gulve EA. Development and comparison of two 3T3-L1 adipocyte models of insulin resistance: increased glucose flux vs glucosamine treatment. Biochem Biophys Res Commun 2000; 273:1033-41. [PMID: 10891367 DOI: 10.1006/bbrc.2000.3082] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance can be induced in vivo by intravenous infusion of glucosamine or in cells by incubation with glucosamine. However, a publication (Hresko, R. C., et al. (1998) J. Biol. Chem. 273, 20658-20668) suggests a trivial explanation of glucosamine-induced insulin resistance whereby intracellular ATP pools are depleted presumably due to the phosphorylation of glucosamine to glucosamine 6-phosphate, a hexosamine pathway intermediate. The reduced ATP level impaired insulin receptor (IR) autophosphorylation and tyrosine kinase activity toward substrates. The present work describes the development and comparison of two methods for inducing insulin resistance, by treating 3T3-L1 adipocytes overnight using either 25 mM glucose/5 nM insulin or 2 mM glucosamine. Under these conditions basal glucose transport rates were comparable with controls. Insulin-stimulated 2-deoxyglucose uptake, however, was reduced by approximately 45% in response to both high glucose/insulin and glucosamine treatment, relative to control cells. The total relative amounts of the insulin-responsive glucose transporter, Glut4, remained constant under both treatment conditions. The relative phosphotyrosine (Tyr(P)) contents of the insulin receptor and its substrate 1 (IRS-1) were assessed in whole cell homogenates. With both methods to induce insulin resistance, IR/IRS-1 Tyr(P) levels were virtually indistinguishable from those in control cells. Insulin-stimulated phosphorylation of Akt on Ser(473) was not impaired in insulin-resistant cells. Furthermore, the relative Tyr(P) content of the PDGF receptor was comparable in high glucose/insulin- or glucosamine-treated 3T3-L1 adipocytes upon subsequent challenge with PDGF. Finally, the relative amounts of glutamine:fructose-6-phosphate amidotransferase and O-linked N-acetylglucosamine transferase, two important hexosamine pathway enzymes, were similar in both treatments when compared with controls. Thus, 3T3-L1 adipocytes can be used as a model system for studying insulin resistance induced by increased influx of glucose. Under appropriate experimental conditions, glucosamine treatment can mimic the effects of increased glucose flux without impairment of tyrosine phosphorylation-based signaling.
Collapse
Affiliation(s)
- S A Ross
- Cardiovascular and Metabolic Diseases Research, Biochemistry and Molecular Biology, G. D. Searle and Company, 800 N. Lindbergh Boulevard, St. Louis, Missouri 63167, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Egawa K, Nakashima N, Sharma PM, Maegawa H, Nagai Y, Kashiwagi A, Kikkawa R, Olefsky JM. Persistent activation of phosphatidylinositol 3-kinase causes insulin resistance due to accelerated insulin-induced insulin receptor substrate-1 degradation in 3T3-L1 adipocytes. Endocrinology 2000; 141:1930-5. [PMID: 10830273 DOI: 10.1210/endo.141.6.7516] [Citation(s) in RCA: 48] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recently, we have reported that the overexpression of a membrane-targeted phosphatidylinositol (PI) 3-kinase (p110CAAX) stimulated p70S6 kinase, Akt, glucose transport, and Ras activation in the absence of insulin but inhibited insulin-stimulated glycogen synthase activation and MAP kinase phosphorylation in 3T3-L1 adipocytes. To investigate the mechanism of p110CAAX-induced cellular insulin resistance, we have now studied the effect of p110CAAX on insulin receptor substrate (IRS)-1 protein. Overexpression of p110CAAX alone decreased IRS-1 protein levels to 63+/-10% of control values. Insulin treatment led to an IRS-1 gel mobility shift (most likely caused by serine/threonine phosphorylation), with subsequent IRS-1 degradation. Moreover, insulin-induced IRS-1 degradation was enhanced by expression of p110CAAX (61+/-16% vs. 13+/-15% at 20 min, and 80+/-8% vs. 41+/-12% at 60 min, after insulin stimulation with or without p110CAAX expression, respectively). In accordance with the decreased IRS-1 protein, the insulin-stimulated association between IRS-1 and the p85 subunit of PI 3-kinase was also decreased in the p110CAAX-expressing cells, and IRS-1-associated PI 3-kinase activity was decreased despite the fact that total PI 3-kinase activity was increased. Five hours of wortmannin pretreatment inhibited both serine/threonine phosphorylation and degradation of IRS-1 protein. These results indicate that insulin treatment leads to serine/threonine phosphorylation of IRS-1, with subsequent IRS-1 degradation, through a PI 3-kinase-sensitive mechanism. Consistent with this, activated PI 3-kinase phosphorylates IRS-1 on serine/threonine residues, leading to IRS- 1 degradation. The similar finding was observed in IRS-2 as well as IRS-1. These results may also explain the cellular insulin-resistant state induced by chronic p110CAAX expression.
Collapse
Affiliation(s)
- K Egawa
- Department of Medicine, University of California, San Diego, La Jolla 92093-0673, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Laybutt DR, Schmitz-Peiffer C, Saha AK, Ruderman NB, Biden TJ, Kraegen EW. Muscle lipid accumulation and protein kinase C activation in the insulin-resistant chronically glucose-infused rat. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:E1070-6. [PMID: 10600797 DOI: 10.1152/ajpendo.1999.277.6.e1070] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic glucose infusion results in hyperinsulinemia and causes lipid accumulation and insulin resistance in rat muscle. To examine possible mechanisms for the insulin resistance, alterations in malonyl-CoA and long-chain acyl-CoA (LCA-CoA) concentration and the distribution of protein kinase C (PKC) isozymes, putative links between muscle lipids and insulin resistance, were determined. Cannulated rats were infused with glucose (40 mg. kg(-1). min(-1)) for 1 or 4 days. This increased red quadriceps muscle LCA-CoA content (sum of 6 species) by 1.3-fold at 1 day and 1.4-fold at 4 days vs. saline-infused controls (both P < 0.001 vs. control). The concentration of malonyl-CoA was also increased (1.7-fold at 1 day, P < 0.01, and 2.2-fold at 4 days, P < 0.001 vs. control), suggesting an even greater increase in cytosolic LCA-CoA. The ratio of membrane to cytosolic PKC-epsilon was increased twofold in the red gastrocnemius after both 1 and 4 days, suggesting chronic activation. No changes were observed for PKC-alpha, -delta, and -theta. We conclude that LCA-CoAs accumulate in muscle during chronic glucose infusion, consistent with a malonyl-CoA-induced inhibition of fatty acid oxidation (reverse glucose-fatty acid cycle). Accumulation of LCA-CoAs could play a role in the generation of muscle insulin resistance by glucose oversupply, either directly or via chronic activation of PKC-epsilon.
Collapse
Affiliation(s)
- D R Laybutt
- Garvan Institute of Medical Research, Sydney, New South Wales 2010, Australia
| | | | | | | | | | | |
Collapse
|
34
|
Pugliese G, Pricci F, Romeo G, Leto G, Amadio L, Iacobini C, Di Mario U. Autocrine and paracrine mechanisms in the early stages of diabetic nephropathy. J Endocrinol Invest 1999; 22:708-35. [PMID: 10595837 DOI: 10.1007/bf03343635] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- G Pugliese
- Dipartimento di Scienze Cliniche, Endocrinologia III, La Sapienza University, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
35
|
Ciaraldi TP, Carter L, Nikoulina S, Mudaliar S, McClain DA, Henry RR. Glucosamine regulation of glucose metabolism in cultured human skeletal muscle cells: divergent effects on glucose transport/phosphorylation and glycogen synthase in non-diabetic and type 2 diabetic subjects. Endocrinology 1999; 140:3971-80. [PMID: 10465266 DOI: 10.1210/endo.140.9.6974] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Chronic exposure (48 h) to glucosamine resulted in a dose-dependent reduction of basal and insulin-stimulated glucose uptake activities in human skeletal muscle cell cultures from nondiabetic and type 2 diabetic subjects. Insulin responsiveness of uptake was also reduced. There was no change in total membrane expression of either GLUT1, GLUT3, or GLUT4 proteins. While glucosamine treatment had no significant effects on hexokinase activity measured in cell extracts, glucose phosphorylation in intact cells was impaired after treatment. Under conditions where glucose transport and phosphorylation were down regulated, the fractional velocity (FV) of glycogen synthase was increased by glucosamine treatment. Neither the total activity nor protein expression of glycogen synthase were influenced by glucosamine treatment. The stimulation of glycogen synthase by glucosamine was not due totally to soluble mediators. Reflective of the effects on transport/phosphorylation, total glycogen content and net glycogen synthesis were reduced after glucosamine treatment. These effects were similar in nondiabetic and type 2 cells. In summary: 1) Chronic treatment with glucosamine reduces glucose transport/phosphorylation and storage into glycogen in skeletal muscle cells in culture and impairs insulin responsiveness as well. 2) Down-regulation of glucose transport/phosphorylation occurs at a posttranslational level of GLUTs. 3) Glycogen synthase activity increases with glucosamine treatment. 4) Nondiabetic and type 2 muscle cells display equal sensitivity and responsiveness to glucosamine. Increased exposure of skeletal muscle to glucosamine, a substrate/precursor of the hexosamine pathway, alters intracellular glucose metabolism at multiple sites and can contribute to insulin resistance in this tissue.
Collapse
Affiliation(s)
- T P Ciaraldi
- Department of Medicine, University of California, San Diego, La Jolla 92093, USA
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Impaired function of the hormone insulin (insulin resistance) is a major feature of type 2 diabetes, a condition that is expected to afflict over 200 million people by early next century. Intensive investigation has failed to find a genetic basis for insulin resistance in the majority of cases. In this brief review the evidence that insulin resistance may be caused by excess nutrient supply will be presented. Both excess glucose and excess fat can cause insulin resistance in muscle and fat tissue, while excess fat can cause impaired suppression of endogenous glucose production. Each nutrient may impair insulin action by several mechanisms, at least one of which may be common to both.
Collapse
Affiliation(s)
- J Proietto
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
37
|
Ruderman NB, Dean D. Malonyl CoA, long chain fatty acyl CoA and insulin resistance in skeletal muscle. J Basic Clin Physiol Pharmacol 1999; 9:295-308. [PMID: 10212840 DOI: 10.1515/jbcpp.1998.9.2-4.295] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Malonyl CoA is an inhibitor of carnitine palmitoyl transferase 1 (CPT1), the enzyme that regulates the transfer of long chain fatty acyl CoA into mitochondria. By virtue of this effect, it is thought to play a key role in regulating fatty acid oxidation. Thus, when the supply of glucose to muscle is increased, malonyl CoA levels increase in keeping with a decreased need for fatty acid oxidation, and fatty acids are preferentially esterified to form diaglycerol and triglycerides. In contrast, during exercise, when the need for fatty acid oxidation is increased, malonyl CoA levels fall. Changes in glucose supply regulate malonyl CoA by modulating the concentration of cytosolic citrate, an allosteric activator of acetyl CoA carboxylase (ACC), the rate-limiting enzyme for malonyl CoA formation and a precursor of its substrate cytosolic acetyl CoA. Conversely, exercise lowers the concentration of malonyl CoA, by activating an AMP-activated protein kinase, which phosphorylates and inhibits ACC. A number of reports have linked sustained increases in the concentration of malonyl CoA in muscle to insulin resistance. In this paper, we review these reports, as well as the notion that changes in malonyl CoA contribute to the increases in long chain fatty acyl CoA, (LCFA CoA), diacylglycerol and triglyceride content and changes in protein kinase C activity and distribution observed in insulin-resistant muscle. We also review the implications of the malonyl CoA/LCFA CoA hypothesis to two other proposed mechanisms for insulin resistance, the glucose-fatty acid cycle and the hexosamine theory.
Collapse
Affiliation(s)
- N B Ruderman
- Diabetes and Metabolism Unit, Boston University Medical Center, MA 02118, USA
| | | |
Collapse
|
38
|
Gustafson TA, Moodie SA, Lavan BE. The insulin receptor and metabolic signaling. Rev Physiol Biochem Pharmacol 1999; 137:71-190. [PMID: 10207305 DOI: 10.1007/3-540-65362-7_5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- T A Gustafson
- Metabolex, Inc., Section of Signal Transduction, Hayward, CA 94545, USA
| | | | | |
Collapse
|
39
|
Ruderman NB, Saha AK, Vavvas D, Witters LA. Malonyl-CoA, fuel sensing, and insulin resistance. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 276:E1-E18. [PMID: 9886945 DOI: 10.1152/ajpendo.1999.276.1.e1] [Citation(s) in RCA: 255] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Malonyl-CoA is an allosteric inhibitor of carnitine palmitoyltransferase (CPT) I, the enzyme that controls the transfer of long-chain fatty acyl (LCFA)-CoAs into the mitochondria where they are oxidized. In rat skeletal muscle, the formation of malonyl-CoA is regulated acutely (in minutes) by changes in the activity of the beta-isoform of acetyl-CoA carboxylase (ACCbeta). This can occur by at least two mechanisms: one involving cytosolic citrate, an allosteric activator of ACCbeta and a precursor of its substrate cytosolic acetyl-CoA, and the other involving changes in ACCbeta phosphorylation. Increases in cytosolic citrate leading to an increase in the concentration of malonyl-CoA occur when muscle is presented with insulin and glucose, or when it is made inactive by denervation, in keeping with a diminished need for fatty acid oxidation in these situations. Conversely, during exercise, when the need of the muscle cell for fatty acid oxidation is increased, decreases in the ATP/AMP and/or creatine phosphate-to-creatine ratios activate an isoform of an AMP-activated protein kinase (AMPK), which phosphorylates ACCbeta and inhibits both its basal activity and activation by citrate. The central role of cytosolic citrate links this malonyl-CoA regulatory mechanism to the glucose-fatty acid cycle concept of Randle et al. (P. J. Randle, P. B. Garland. C. N. Hales, and E. A. Newsholme. Lancet 1: 785-789, 1963) and to a mechanism by which glucose might autoregulate its own use. A similar citrate-mediated malonyl-CoA regulatory mechanism appears to exist in other tissues, including the pancreatic beta-cell, the heart, and probably the central nervous system. It is our hypothesis that by altering the cytosolic concentrations of LCFA-CoA and diacylglycerol, and secondarily the activity of one or more protein kinase C isoforms, changes in malonyl-CoA provide a link between fuel metabolism and signal transduction in these cells. It is also our hypothesis that dysregulation of the malonyl-CoA regulatory mechanism, if it leads to sustained increases in the concentrations of malonyl-CoA and cytosolic LCFA-CoA, could play a key role in the pathogenesis of insulin resistance in muscle. That it may contribute to abnormalities associated with the insulin resistance syndrome in other tissues and the development of obesity has also been suggested. Studies are clearly needed to test these hypotheses and to explore the notion that exercise and some pharmacological agents that increase insulin sensitivity act via effects on malonyl-CoA and/or cytosolic LCFA-CoA.
Collapse
Affiliation(s)
- N B Ruderman
- Diabetes Unit, Section of Endocrinology and Departments of Medicine and Physiology, Boston University Medical Center, Boston, Massachusetts 02118, USA
| | | | | | | |
Collapse
|
40
|
Morris MJ, Tortelli CF, Filippis A, Proietto J. Reduced BAT function as a mechanism for obesity in the hypophagic, neuropeptide Y deficient monosodium glutamate-treated rat. REGULATORY PEPTIDES 1998; 75-76:441-7. [PMID: 9802441 DOI: 10.1016/s0167-0115(98)00100-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Neuropeptide Y (NPY) exerts effects on food intake at the level of the paraventricular nucleus (PVN), which receives a dense projection from the arcuate nucleus. Monosodium glutamate (MSG) has been shown to induce hyperadiposity despite hypophagia associated with chemical ablation of the arcuate nucleus. We investigated the mechanism for the excess fat accumulation by studying the time course of changes in brain NPY content, food intake, leptin levels and BAT GLUT4 content after neonatal MSG treatment. Male rat pups were injected with MSG or saline vehicle on days 2, 4, and 6 and examined at 30 and 90 days. Plasma leptin, body mass, length, adipose tissue mass and brown fat GLUT4 were measured and brains dissected for measurement of NPY content. By 30 days, NPY concentrations were reduced in the arcuate nucleus and anterior hypothalamus, and animals tended to be hypophagic. Peripheral adipose tissue levels were less than controls, in line with their low leptin concentrations. At 90 days, MSG treatment was associated with marked reductions in NPY concentrations in several hypothalamic areas, including the PVN and arcuate nucleus, along with increased adiposity and plasma leptin. Animals also displayed marked hypophagia. Levels of GLUT4 transporter were reduced in brown adipose tissue at both ages. The early decrease in brown fat GLUT4 suggests an impairment of the hypothalamic sympathetic input to brown fat which disrupts thermogenesis, contributing to the development of adiposity in the presence of hypophagia.
Collapse
Affiliation(s)
- M J Morris
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | |
Collapse
|
41
|
Filippis A, Clark S, Proietto J. Possible role for gp160 in constitutive but not insulin-stimulated GLUT4 trafficking: dissociation of gp160 and GLUT4 localization. Biochem J 1998; 330 ( Pt 1):405-11. [PMID: 9461537 PMCID: PMC1219154 DOI: 10.1042/bj3300405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
GLUT4-containing vesicles are constantly cycling in both basal and insulin-stimulated states. Our previous studies have shown that basal cycling of GLUT4 is impaired under conditions of high glucose or glucosamine and, as a consequence, GLUT4 is retained intracellularly in low-density microsomes [Filippis A., Clark, S., and Proietto, J. (1997) Biochem. J. 324, 981-985]. In addition to GLUT4 itself, a major protein component of GLUT4-containing vesicles is a glycoprotein of Mr 160000 (gp160). In all studies so far published gp160 has been co-localized with GLUT4 under all conditions. In this study, we show that retention of GLUT4 in low-density microsomes (enriched in Golgi apparatus) is associated with a decrease in gp160 levels in this compartment. A concomitant increase of gp160 in high-density microsomes (enriched in endoplasmic reticulum), demonstrates for the first time a dissociation in the localization of gp160 and GLUT4. Despite the marked decrease in gp160 levels in the GLUT4-containing compartment, insulin-stimulated translocation was normal, while little gp160 appeared in the plasma membrane in response to insulin. The retention of gp160 in the high-density microsomes is apparently not due to a change in the glycosylation state of gp160 as measured by [3H]mannose incorporation. It is concluded that, in rat adipocytes, gp160 is not required for insulin-stimulated translocation, but may be necessary for constitutive trafficking of the GLUT4-containing vesicle.
Collapse
Affiliation(s)
- A Filippis
- The University of Melbourne, Department of Medicine, Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| | | | | |
Collapse
|
42
|
Donnelly R, Qu X. Mechanisms of insulin resistance and new pharmacological approaches to metabolism and diabetic complications. Clin Exp Pharmacol Physiol 1998; 25:79-87. [PMID: 9493493 DOI: 10.1111/j.1440-1681.1998.tb02181.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
1. Resistance to insulin-mediated glucose transport and metabolism has been identified as a primary mechanism in the pathogenesis of non-insulin-dependent diabetes mellitus (NIDDM) and as a target for drug development. The aetiology of insulin resistance is likely to be multifactorial, but the present review focuses on candidate post-receptor mechanisms of insulin resistance, particularly protein kinase C (PKC), and the metabolic and genetic significance of beta3-adrenoceptors (beta3-AR) in adipose tissue. 2. Multiple lines of evidence suggest that isoform-selective activation of PKC phosphorylates and down-regulates one or more substrates involved in glucose transport and metabolism (e.g. glycogen synthase and the insulin receptor) and recent studies have shown increased expression of calcium-independent isozymes (PKC-epsilon and PKC-theta) in the membrane fraction of skeletal muscle in fructose- and fat-fed rat models of insulin resistance. In addition, there is separate evidence that glucose-induced PKC activation plays an important role in the micro- and macrovascular complications of diabetes. 3. New pharmacological approaches to NIDDM and obesity have focused on insulin-sensitizing agents (e.g. troglitazone), beta3-AR agonists, anti-lipolytic drugs (e.g. the adenosine A1 receptor agonist GR79236) and selective inhibitors of PKC isoforms (e.g. the inhibitor of PKC-beta LY333531). Experimental studies with GR79236 show that this drug ameliorates the hypertriglyceridaemia induced by fructose feeding and that the reduction in fatty acid levels is associated with secondary improvements in glucose tolerance. 4. Recent insights into the pathogenesis of NIDDM and its associated complications have been used to develop a range of new therapeutic agents that are currently showing promise in clinical and preclinical development.
Collapse
Affiliation(s)
- R Donnelly
- Department of Pharmacology, University of Sydney, New South Wales, Australia.
| | | |
Collapse
|