Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(8)2021 Apr 18.
Article in English | MEDLINE | ID: mdl-33919569

ABSTRACT

Type 2 diabetes (T2D) is a worldwide health problem, ranked as one of the leading causes for severe morbidity and premature mortality in modern society. Management of blood glucose is of major importance in order to limit the severe outcomes of the disease. However, despite the impressive success in the development of new antidiabetic drugs, almost no progress has been achieved with regard to the development of novel insulin-sensitizing agents. As insulin resistance is the most eminent factor in the patho-etiology of T2D, it is not surprising that an alarming number of patients still fail to meet glycemic goals. Owing to its wealth of chemical structures, the plant kingdom is considered as an inventory of compounds exerting various bioactivities, which might be used as a basis for the development of novel medications for various pathologies. Antidiabetic activity is found in over 400 plant species, and is attributable to varying mechanisms of action. Nevertheless, relatively limited evidence exists regarding phytochemicals directly activating insulin signaling, which is the focus of this review. Here, we will list plants and phytochemicals that have been found to improve insulin sensitivity by activation of the insulin signaling cascade, and will describe the active constituents and their mechanism of action.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Receptor, Insulin/metabolism , Animals , Diabetes Mellitus, Type 2/genetics , Humans , Phytochemicals/metabolism , Receptor, Insulin/genetics , Signal Transduction
2.
Curr Dev Nutr ; 3(4): nzy097, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30993256

ABSTRACT

BACKGROUND: Oxidative stress contributes to the pathologic process leading to the development, progression, and complications of type 1 diabetes (T1D). OBJECTIVE: The aim of this study was to investigate the effect of the antioxidant N-acetyl-l-cysteine (NAC), supplemented during early life or adulthood on the development of T1D. METHODS: NAC was administered to nonobese diabetic (NOD) female mice during pregnancy and lactation, and the development of diabetes was followed in offspring. In an additional set of experiments, offspring of untreated mice were given NAC during adulthood, and the development of T1D was followed. Morbidity rate, insulitis and serum cytokines were measured in the 2 sets of experiments. In addition, markers of oxidative stress, glutathione, lipid peroxidation, total antioxidant capacity and activity of antioxidant enzymes, were followed. RESULTS: Morbidity rate was reduced in both treatment protocols. A decrease in interferon γ, tumor necrosis factor α, interleukin 1α, and other type 1 diabetes-associated proinflammatory cytokines was found in mice supplemented with NAC in adulthood or during early life compared with control NOD mice. The severity of insulitis was higher in control NOD mice than in treated groups. NAC administration significantly reduced oxidative stress, as determined by reduced lipid peroxidation and increased total antioxidant capacity in serum and pancreas of mice treated in early life or in adulthood and increased pancreatic glutathione when administrated in adulthood. The activity of antioxidant enzymes was not affected in mice given NAC in adulthood, whereas an increase in the activity of superoxide dismutase and catalase was demonstrated in the pancreas of their offspring. CONCLUSION: NAC decreased morbidity of NOD mice by attenuating the immune response, presumably by eliminating oxidative stress, and might be beneficial in reducing morbidity rates of T1D in high-risk individuals.

4.
Biochim Biophys Acta Mol Cell Res ; 1865(4): 551-559, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29317261

ABSTRACT

Insulin binding to its cell surface receptor (IR) activates a cascade of events leading to its biological effects. The Insulin-IR complex is rapidly internalized and then is either recycled back to the plasma membrane or sent to lysosomes for degradation. Although most of the receptor is recycled or degraded, a small amount may escape this pathway and migrate to the nucleus of the cell where it might be important in promulgation of receptor signals. In this study we explored the mechanism by which insulin induces IR translocation to the cell nucleus. Experiments were performed cultured L6 myoblasts, AML liver cells and 3T3-L1 adipocytes. Insulin treatment induced a rapid increase in nuclear IR protein levels within 2 to 5 min. Treatment with WGA, an inhibitor of nuclear import, reduced insulin-induced increases nuclear IR protein; IR was, however, translocated to a perinuclear location. Bioinformatics tools predicted a potential nuclear localization sequence (NLS) on IR. Immunofluorescence staining showed that a point mutation on the predicted NLS blocked insulin-induced IR nuclear translocation. In addition, blockade of nuclear IR activation in isolated nuclei by an IR blocking antibody abrogated insulin-induced increases in IR tyrosine phosphorylation and nuclear PKCδ levels. Furthermore, over expression of mutated IR reduced insulin-induced glucose uptake and PKB phosphorylation. When added to isolated nuclei, insulin induced IR phosphorylation but had no effect on nuclear IR protein levels. These results raise questions regarding the possible role of nuclear IR in IR signaling and insulin resistance.


Subject(s)
Cell Nucleus/metabolism , Insulin/pharmacology , Nuclear Localization Signals/metabolism , Receptor, Insulin/metabolism , 3T3-L1 Cells , Active Transport, Cell Nucleus/drug effects , Amino Acid Sequence , Animals , Cell Nucleus/drug effects , Glucose/metabolism , Humans , Mice , Mutant Proteins/metabolism , Nuclear Localization Signals/chemistry , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/chemistry
5.
Am J Transl Res ; 8(9): 3744-3756, 2016.
Article in English | MEDLINE | ID: mdl-27725855

ABSTRACT

Oxidative stress is associated with different pathological conditions, including glucose intolerance and type 2 diabetes (T2D), however studies had failed to prove the benefits of antioxidants in T2D. AIM: On the assumption that the failure to demonstrate such anti-diabetic effects is a result of sub-optimal or excessive antioxidant dosage, we aimed to clarify the dose-response effect of the antioxidant N-Acetyl-L-Cysteine (NAC) on the progression of T2D in-vivo. METHODS: Experiments were conducted on KK-Ay mice and HFD-fed mice given NAC at different concentrations (200-1800 and 60-600 mg/kg/day, respectively). Glucose and insulin tolerance tests were performed and plasma insulin and lipid peroxidation were measured. Insulin signaling pathway was followed in muscle and liver. Hepatic TG accumulation and mRNA expression of genes involved in glucose metabolism were measured. RESULTS: While 600-1800 mg/kg/day NAC all improved glucose tolerance in KK-Ay mice, only the 1200 mg/kg/day treatment increased insulin sensitivity. Hepatic function was not affected, however; microsteatosis rather than macrosteatosis was observed in NAC-treated mice compared to control. Glucose tolerance was improved in NAC-treated HFD-fed mice as well; the best results obtained with a dose of 400 mg NAC/kg/day. This was followed by lower weight gain and hepatic TG. Plasma lipid peroxidation was not correlated with the glucose-lowering effects of NAC in either model. CONCLUSION: Identification of the optimal dose of NAC and the population that would benefit the most from such intervention is essential in order to apply preventive and/or therapeutic use of NAC and similar agents in the future.

6.
J Ethnopharmacol ; 155(1): 362-72, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24882728

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sarcopoterium spinosum is an abundant plant in Israel, used by Bedouin medicinal practitioners for the treatment of diabetes. In our previous study we validated the anti-diabetic activity of Sarcopoterium spinosum. The aim of this study was to further clarify its mechanism of action. MATERIALS AND METHODS: In-vivo studies were performed on KK-a/y mice given the extract for 6 weeks. Insulin tolerance test was performed, and relative pancreatic islets area was measured. Mechanisms of action were investigated in L6 myotubes using protein array, Western blot analysis and confocal microscopy. Glucose uptake assays were performed in 3T3-L1 adipocytes. RESULTS: Sarcopoterium spinosum extract reduced fasting blood glucose and improved insulin sensitivity in treated mice. Hypertrophic islets were detected in diabetic, but not in Sarcopoterium spinosum-treated mice. Sarcopoterium spinosum phosphorylated PTEN on ser380 and thr382/383, which are known inhibitory sites. PKB was not phosphorylated by Sarcopoterium spinosum, however, translocation of PKB from cytoplasm to the membrane and nucleus was detected. Target proteins of PKB were regulated by Sarcopoterium spinosum; GSK3ß was phosphorylated and cytosolic localization of FoxO was increased. Glucose uptake was increased in a PI3K and AMPK-independent mechanism. CONCLUSIONS: We suggest that Sarcopoterium spinosum inhibited PTEN and activated PKB by a mechanism which is independent of ser473 and thr308 phosphorylation. Other post translation modifications might be involved and should be analyzed further in order to understand this unique PKB activation. Identifying the active molecules in the extract, may lead to the development of new agents for the treatment of insulin resistance.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Insulin/metabolism , Plant Extracts/pharmacology , Rosaceae/chemistry , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Glucose/metabolism , Insulin Resistance , Israel , Male , Medicine, Traditional , Mice , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , PTEN Phosphohydrolase/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism
7.
J Cell Biochem ; 113(6): 1966-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22253064

ABSTRACT

Whereas oxidative stress is linked to cellular damage, reactive oxygen species (ROS) are also believed to be involved in the propagation of signaling pathways. Studies on the role of ROS in pancreatic beta-cell physiology, in contrast to pathophysiology, have not yet been reported. In this study we investigate the importance of maintaining cellular redox state on pancreatic beta-cell function and viability, and the effects of leptin and adiponectin on this balance. Experiments were conducted on RINm and MIN6 pancreatic beta-cells. Leptin (1-100 ng/ml) and adiponectin (1-100 nM) increased ROS accumulation, as was determined by DCFDA fluorescence. Using specific inhibitors, we found that the increase in ROS levels was mediated by NADPH oxidase (Nox), but not by AMP kinase (AMPK) or phosphatidyl inositol 3 kinase (PI3K). Leptin and adiponectin increased beta-cell number as detected by the XTT method, but did not affect apoptosis, indicating that the increased cell number results from increased proliferation. The adipokines-induced increase in viability is ROS dependent as this effect was abolished by N-acetyl-L-cysteine (NAC) or PEG-catalase. In addition, insulin secretion was found to be regulated by alterations in redox state, but not by adipokines. Finally, the effects of the various treatments on activity and mRNA expression of several antioxidant enzymes were determined. Both leptin and adiponectin reduced mRNA levels of superoxide dismutase (SOD)1. Adiponectin also decreased SOD activity and increased catalase and glutathione peroxidase (GPx) activities in the presence of H2O2. The results of this study show that leptin and adiponectin, by inducing a physiological increase in ROS levels, may be positive regulators of beta-cell mass.


Subject(s)
Adiponectin/pharmacology , Insulin-Secreting Cells/metabolism , Leptin/pharmacology , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Adenylate Kinase/metabolism , Adiponectin/metabolism , Animals , Apoptosis/drug effects , Catalase/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Glutathione Peroxidase/biosynthesis , Glutathione Peroxidase/genetics , Insulin/metabolism , Insulin Secretion , Leptin/metabolism , Mice , NADPH Oxidases/metabolism , Oxidation-Reduction/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Polyethylene Glycols/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reactive Oxygen Species , Signal Transduction/drug effects , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Superoxide Dismutase-1
8.
J Cell Biochem ; 113(6): 2064-76, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22271577

ABSTRACT

The liver is a major insulin-responsive tissue responsible for glucose regulation. One important mechanism in this phenomenon is insulin-induced glycogen synthesis. Studies in our laboratory have shown that protein kinase Cs delta (PKCδ) and alpha (α) have important roles in insulin-induced glucose transport in skeletal muscle, and that their expression and activity are regulated by insulin. Their importance in glucose regulation in liver cells is unclear. In this study we investigated the possibility that these isoforms are involved in the mediation of insulin-induced glycogen synthesis in hepatocytes. Studies were done on rat hepatocytes in primary culture and on the AML-12 (alpha mouse liver) cell line. Insulin increased activity and tyrosine phosphorylation of PKCδ within 5 min. In contrast, activity and tyrosine phosphorylation of PKCα were not increased by insulin. PKCδ was constitutively associated with IR, and this was increased by insulin stimulation. Suppression of PKCδ expression by transfection with RNAi, or overexpression of kinase dead (dominant negative) PKCδ reduced both the insulin-induced activation of PKB/Akt and the phosphorylation of glycogen synthase kinase 3 (GSK3) and reduced significantly insulin-induced glucose uptake. In addition, treatment of primary rat hepatocytes with rottlerin abrogated insulin-induced increase in glycogen synthesis. Neither overexpression nor inhibition of PKCα appeared to alter activation of PKB, phosphorylation of GSK3 or glucose uptake in response to insulin. We conclude that PKCδ, but not PKCα, plays an essential role in insulin-induced glucose uptake and glycogenesis in hepatocytes.


Subject(s)
Glucose/metabolism , Hepatocytes/metabolism , Insulin/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Cells, Cultured , Glycogen/biosynthesis , Glycogen Synthase Kinase 3/metabolism , Hepatocytes/drug effects , Liver/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Phosphorylation , Protein Kinase C-alpha/genetics , Protein Kinase C-delta/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering , Rats , Rats, Wistar , Signal Transduction
9.
Biochim Biophys Acta ; 1803(11): 1265-75, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20708645

ABSTRACT

Insulin rapidly upregulates protein levels of PKCδ in classical insulin target tissues skeletal muscle and liver. Insulin induces both a rapid increase in de novo synthesis of PKCδ protein. In this study we examined the possibility that insulin may also inhibit degradation of PKCδ. Experiments were performed on L6 skeletal muscle myoblasts or myotubes in culture. Phorbol ester (PMA)- and insulin-induced degradation of PKCδ were abrogated by proteasome inhibition. Both PMA and insulin induced ubiquitination of PKCδ, but not of that PKCα or PKCε and increased proteasome activity within 5 min. We examined the role of tyrosine phosphorylation of PKCδ in targeting PKCδ for degradation by the ubiquitin-proteasome pathway. Transfection of cells with PKCδY(311)F, which is not phosphorylated, resulted in abolition of insulin-induced ubiquitination of PKCδ and increase in proteasome activity. We conclude that insulin induces degradation of PKCδ via the ubiquitin-proteasome system, and that this effect requires phosphorylation on specific tyrosine residues for targeting PKCδ for degradation by the ubiquitin-proteasome pathway. These studies provide additional evidence for unique effects of insulin on regulation of PKCδ protein levels.


Subject(s)
Insulin/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Kinase C-delta/metabolism , Signal Transduction/drug effects , Ubiquitin/metabolism , Amino Acid Substitution , Animals , Blotting, Western , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Leupeptins/pharmacology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation/drug effects , Proteasome Inhibitors , Protein Kinase C-delta/genetics , Tetradecanoylphorbol Acetate/pharmacology , Tyrosine/genetics , Tyrosine/metabolism , Ubiquitination/drug effects
10.
J Ethnopharmacol ; 129(1): 10-7, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20219662

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sarcopoterium spinosum (L.) sp., a common plant in the Mediterranean region, is widely used as an antidiabetic drug by Bedouin healers. However, the antidiabetic properties of Sarcopoterium spinosum had not been fully validated using scientific tools. AIM OF THE STUDY: To determine the effectiveness of Sarcopoterium spinosum extract as an antidiabetic agent in vitro and in vivo. MATERIALS AND METHODS: RINm pancreatic beta-cells, L6 myotubes, 3T3-L1 adipocytes and AML-12 hepatocytes were treated with an aqueous Sarcopoterium spinosum extract (0.001-10mg/ml). The effect of the extract on specific physiological functions, including insulin secretion, pancreatic beta-cell viability, GSK3 beta phosphorylation, lipolysis and glucose uptake was measured. In vivo studies were performed using KK-A(y) mice, given the extract for several weeks. IPGTT was performed, and plasma insulin, FFA, food consumption and body weight were measured. In addition, diabetic KK-A(y) mice were given a single dose of the extract, and IPGTT was performed. RESULTS: Sarcopoterium spinosum extract increased basal and glucose/forskolin-induced insulin secretion in RINm cells, and increased cell viability. The extract inhibited lipolysis in 3T3-L1 adipocytes, and induced glucose uptake in these cells as well as in AML-12 hepatocytes and L6 myotubes. GSK3 beta phosphorylation was also induced in L6 myotubes, suggesting increased glycogen synthesis. Sarcopoterium spinosum extract had a preventive effect on the progression of diabetes in KK-A(y) mice. Catechin and epicatechin were detected in Sarcopoterium spinosum extract using hyphenated LC-MS/MS. CONCLUSIONS: Sarcopoterium spinosum extract has effects that mimic those of insulin and provide the basis for antidiabetic activity of the extract.


Subject(s)
Adipocytes/drug effects , Diabetes Mellitus, Experimental/drug therapy , Glucose/metabolism , Hypoglycemic Agents/therapeutic use , Insulin/metabolism , Plant Extracts/therapeutic use , Rosaceae/chemistry , 3T3-L1 Cells/drug effects , Adipocytes/metabolism , Animals , Biological Transport , Catechin/analysis , Cell Line , Cell Survival/drug effects , Colforsin/pharmacology , Diabetes Mellitus, Experimental/metabolism , Disease Progression , Glucose/pharmacology , Glucose Tolerance Test , Glycogen/biosynthesis , Hepatocytes/drug effects , Hepatocytes/metabolism , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Insulin Secretion , Lipolysis/drug effects , Male , Mice , Mice, Inbred Strains , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Phytotherapy , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Roots
11.
J Mol Endocrinol ; 44(3): 155-69, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19952103

ABSTRACT

Protein kinase C delta (PKCdelta) is induced by insulin to rapidly associate with insulin receptor (IR) and upregulates insulin signaling. We utilized specific JM and CT receptor domains and chimeras of PKCalpha and PKCdelta regulatory and catalytic domains to elucidate which components of PKCdelta are responsible for positive regulatory effects of PKCdelta on IR signaling. Studies were performed on L6 and L8 skeletal muscle myoblasts and myotubes. PKCdelta was preferentially bound to the JM domain of IR, and insulin stimulation increased this binding. Both PKCdelta/alpha and PKCalpha/delta chimeras (regulatory/catalytic) were bound preferentially to the JM but not to the CT domain of IR. Although IR-PKCdelta binding was higher in cells expressing either the PKCdelta/alpha or PKCalpha/delta chimera than in control cells, upregulation of IR signaling was observed only in PKCdelta/alpha cells. Thus, in response to insulin increases in tyrosine phosphorylation of IR and insulin receptor substrate-1, downstream signaling to protein kinase B and glycogen synthase kinase 3 (GSK3) and glucose uptake were greater in cells overexpressing PKCdelta/alpha and the PKCdelta/delta domains than in cells expressing the PKCalpha/delta domains. Basal binding of Src to PKCdelta was higher in both PKCdelta/alpha- and PKCalpha/delta-expressing cells compared to control. Binding of Src to IR was decreased in PKCalpha/delta cells but remained elevated in the PKCdelta/alpha cells in response to insulin. Finally, insulin increased Src activity in PKCdelta/alpha-expressing cells but decreased it in PKCalpha/delta-expressing cells. Thus, the regulatory domain of PKCdelta via interaction with Src appears to determine the role of PKCdelta as a positive regulator of IR signaling in skeletal muscle.


Subject(s)
Protein Kinase C-delta/chemistry , Protein Kinase C-delta/metabolism , Receptor, Insulin/metabolism , Animals , Biological Transport/drug effects , Blotting, Western , Cell Line , Glycogen Synthase Kinase 3/metabolism , Immunoprecipitation , In Vitro Techniques , Insulin/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Binding/genetics , Protein Binding/physiology , Protein Kinase C-delta/genetics , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-akt/metabolism , Rats
12.
Endocrinology ; 149(4): 1718-27, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18162512

ABSTRACT

Protein kinase C (PKC) isoforms are involved in the transduction of a number of signals important for the regulation of cell growth, differentiation, apoptosis, and other cellular functions. PKC proteins reside in the cytoplasm in an inactive state translocate to various membranes to become fully activated in the presence of specific cofactors. Recent evidence indicates that PKC isoforms have an important role in the nucleus. We recently showed that insulin rapidly increases PKCdelta RNA and protein. In this study we initially found that insulin induces an increase in PKCdelta protein in the nuclear fraction. We therefore attempted to elucidate the mechanism of the insulin-induced increase in nuclear PKCdelta. Studies were performed on L6 skeletal myoblasts and myotubes. The increase in nuclear PKCdelta appeared to be unique to insulin because it was not induced by other growth factors or rosiglitazone. Inhibition of transcription or translation blocked the insulin-induced increase in nuclear PKCdelta, whereas inhibition of protein import did not. Inhibition of protein export from the nucleus reduced the insulin-induced increase in PKCdelta in the cytoplasm and increased it in the nucleus. The increase in nuclear PKCdelta induced by insulin was reduced but not abrogated by treatment of isolated nuclei by trypsin digestion. Finally, we showed that insulin induced incorporation of (35)S-methionine into nuclear PKCdelta protein; this effect was not blocked by inhibition of nuclear import. Thus, these results suggest that insulin may induce nuclear-associated, or possibly nuclear, translation of PKCdelta protein.


Subject(s)
Cell Nucleus/metabolism , Insulin/pharmacology , Muscle, Skeletal/drug effects , Protein Kinase C-delta/biosynthesis , Active Transport, Cell Nucleus , Animals , Cells, Cultured , Methionine/metabolism , Muscle, Skeletal/metabolism , Protein Kinase C-delta/analysis , Rats
13.
Endocrinology ; 149(2): 605-14, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18006633

ABSTRACT

Whereas positive regulatory events triggered by insulin binding to insulin receptor (IR) have been well documented, the mechanism by which the activated IR is returned to the basal status is not completely understood. Recently studies focused on the involvement of protein tyrosine phosphatases (PTPs) and how they might influence IR signaling. In this study, we examined the possibility that cytosolic PTPepsilon (cytPTPepsilon) is involved in IR signaling. Studies were performed on L6 skeletal muscle cells. cytPTPepsilon was overexpressed by using pBABE retroviral expression vectors. In addition, we inhibited cytPTPepsilon by RNA silencing. We found that insulin induced rapid association of cytPTPepsilon with IR. Interestingly, this association appeared to occur in the plasma membrane and on stimulation with insulin the two proteins internalized together. Moreover, it appeared that almost all internalized IR was associated with cytPTPepsilon. We found that knockdown of cytPTPepsilon by RNA silencing increased insulin-induced tyrosine phosphorylation of IR and IR substrate (IRS)-1 as well as phosphorylation of protein kinase B and glycogen synthase kinase-3 and insulin-induced stimulation of glucose uptake. Moreover, overexpression of wild-type cytPTPepsilon reduced insulin-induced tyrosine phosphorylation of IR, IRS-1, and phosphorylation of protein kinase B and glycogen synthase kinase-3 and insulin-induced stimulation of glucose uptake. Finally, insulin-induced tyrosine phosphorylation of IR and IRS-1 was greater in skeletal muscle from mice lacking the cytPTPepsilon gene than that from wild-type control animals. We conclude that cytPTPepsilon serves as another major candidate negative regulator of IR signaling in skeletal muscle.


Subject(s)
Muscle, Skeletal/enzymology , Receptor, Insulin/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Signal Transduction/physiology , Animals , Cell Line , Cell Membrane/enzymology , Cytosol/enzymology , Gene Expression Regulation, Enzymologic , Glucose/metabolism , Insulin/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/cytology , Phosphorylation , RNA Interference , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Tyrosine/metabolism , Up-Regulation/physiology , src-Family Kinases/metabolism
14.
Cell Signal ; 19(3): 556-62, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17046201

ABSTRACT

SP-1, a ubiquitous transcription factor involved in regulation of target genes participating in specific signaling pathways, is utilized by insulin for induction of gene transcription. Transcriptional activation generally occurs only after several (14-24) hours. A major element rapidly activated by insulin in skeletal muscle is PKCdelta, which plays a positive regulatory role in insulin signaling. We recently reported that insulin stimulation of skeletal muscle increases PKCdelta RNA expression and PKCdelta protein levels within 5 min. These effects were blocked by inhibitors of either translation or transcription. In this study, we investigated the possibility that SP-1 may participate in this unusually rapid effect. Studies were performed on myoblasts and myotubes of the L6 skeletal muscle cell line. Insulin rapidly increased SP-1 levels and stimulated SP-1 phosphorylation in the nuclear fraction of L6 myotubes. The increase in nuclear SP-1 was blocked by inhibition of nuclear import. Inhibition of SP-1, either pharmacologically or by suppression of SP-1 by RNAi, nearly completely abrogated insulin-induced increase in PKCdelta promoter activity. Insulin induced a rapid association of SP-1 with the PKCalpha promoter. In addition, SP-1 inhibition blocked insulin-induced increases in both PKCdelta RNA expression and PKCdelta protein levels. We conclude that insulin rapidly stimulates SP-1, which mediates the ability of this hormone to induce the rapid transcription of a major target gene utilized in the insulin signaling cascade.


Subject(s)
Insulin/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Protein Kinase C-delta/metabolism , Sp1 Transcription Factor/metabolism , Animals , Cell Line , Humans , Hypoglycemic Agents/pharmacology , Insulin/genetics , Kinetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , Myoblasts/drug effects , Myoblasts/enzymology , Promoter Regions, Genetic , Protein Kinase C-delta/genetics , RNA Interference , Rats , Recombinant Proteins/pharmacology
15.
Biochem Biophys Res Commun ; 352(1): 78-83, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17109817

ABSTRACT

Protein kinase C delta (PKCdelta) is a key molecule in insulin signaling essential for insulin-induced glucose transport in skeletal muscle. Recent studies in our laboratory have shown that insulin rapidly stimulates PKCdelta activity and increases PKCdelta protein and RNA levels, and that the SP-1 transcription factor is involved in insulin-induced transcription of the PKCdelta gene. Activation of SP-1 involves serine phosphorylation and translocation to the nucleus. In this study we examined the possibility that PKCalpha might be involved in serine phosphorylation and activation of SP-1. We found that insulin rapidly phosphorylates and translocates SP-1. In the cytoplasm, SP-1 was constitutively associated with PKCalpha, and insulin stimulation caused these proteins to dissociate. In contrast, in the nucleus insulin induced an increase in association between PKCalpha and SP-1. PKCalpha inhibition blocked insulin-induced serine phosphorylation of SP-1 and its association with PKCalpha in the nucleus. Inhibition of PKCalpha also reduced the insulin-induced increase in PKCdelta RNA and protein in the cytoplasmic and nuclear fractions. We also attempted to determine if another transcription factor might be involved in regulation of PKCdelta expression. We earlier showed that insulin did not affect nuclear NFkappaB levels. Inhibition of NFkappaB, however, increased insulin-induced increase in PKCdelta RNA and protein in the cytoplasmic and nuclear fractions. Surprisingly, this inhibition reduced the insulin-induced increase in cytoplasmic and nuclear PKCalpha RNA and protein. Inhibition of PKCdelta reduced IkappaBalpha phosphorylation as well as NFkappaB activation. Thus, PKCalpha regulates insulin-induced PKCdelta expression levels and this regulation involves activation of SP-1 and NFkappaB.


Subject(s)
Insulin/pharmacology , NF-kappa B/metabolism , Protein Kinase C-alpha/metabolism , Protein Kinase C-delta/metabolism , Sp1 Transcription Factor/metabolism , Transcription, Genetic/genetics , Animals , Cell Line , Gene Expression Regulation/drug effects , Phosphorylation , Phosphoserine/metabolism , Protein Binding , Protein Kinase C-delta/genetics , Rats
16.
Biochem Biophys Res Commun ; 349(3): 954-62, 2006 Oct 27.
Article in English | MEDLINE | ID: mdl-16962999

ABSTRACT

PKCdelta has been shown to be activated by insulin and to interact with insulin receptor and IRS. PKB(Akt) plays an important role in glucose transport and glycogen synthesis. In this study, we investigated the possibility that PKCdelta may be involved in insulin-induced activation of PKB. Studies were conducted on primary cultures of rat skeletal muscle. PKB was activated by insulin stimulation within 5min and reached a peak by 15-30min. Insulin also increased the physical association between PKCdelta with PKB and with PDK1. The insulin-induced PKCdelta-PKB association was PI3K dependent. PKB-PKCdelta association was accounted for by the involvement of PDK1. Overexpression of dominant negative PKCdelta abrogated insulin-induced association of PKCdelta with both PKB and PDK1. Blockade of PKCdelta also decreased insulin-induced Thr308 PKB phosphorylation and PKB translocation. Moreover, PKCdelta inhibition reduced insulin-induced GSK3 phosphorylation. The results indicate that insulin-activated PKCdelta interacts with PDK1 to regulate PKB.


Subject(s)
Insulin/pharmacology , Protein Kinase C-delta/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Cells, Cultured , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/enzymology , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Transport , Rats
17.
Mol Genet Metab ; 89(1-2): 32-47, 2006.
Article in English | MEDLINE | ID: mdl-16798038

ABSTRACT

Recent studies implicate specific PKC isoforms in the insulin-signaling cascade. Insulin activates PKCs alpha, betaII, delta and zeta in several cell types. In addition, as will be documented in this review, certain members of the PKC family may also be activated and act upstream of PI3 and MAP kinases. Each of these isoforms has been shown one way or another either to mimic or to modify insulin-stimulated effects in one or all of the insulin-responsive tissues. Moreover, each of the isoforms has been shown to be activated by insulin stimulation or conditions important for effective insulin stimulation. Studies attempting to demonstrate a definitive role for any of the isoforms have been performed on different cells, ranging from appropriate model systems for skeletal muscle, liver and fat, such as primary cultures, and cell lines and even in vivo studies, including transgenic mice with selective deletion of specific PKC isoforms. In addition, studies have been done on certain expression systems such as CHO or HEK293 cells, which are far removed from the tissues themselves and serve mainly as vessels for potential protein-protein interactions. Thus, a clear picture for many of the isoforms remains elusive in spite of over two decades of intensive research. The recent intrusion of transgenic and precise molecular biology technologies into the research armamentarium has opened a wide range of additional possibilities for direct involvement of individual isoforms in the insulin signaling cascade. As we hope to discuss within the context of this review, whereas many of the long sought-after answers to specific questions are not yet clear, major advances have been made in our understanding of precise roles for individual PKC isoforms in mediation of insulin effects. In this review, in which we shall focus our attention on isoforms in the conventional and novel categories, a clear case will be made to show that these isoforms are not only expressed but are importantly involved in regulation of insulin metabolic effects.


Subject(s)
Insulin/metabolism , Protein Kinase C/physiology , Signal Transduction , Animals , Humans , Protein Isoforms/genetics , Protein Isoforms/physiology , Protein Kinase C/genetics
18.
Biochem Biophys Res Commun ; 345(2): 817-24, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16707110

ABSTRACT

Certain PKC isoforms are stimulated by insulin and interact with IR as well as with IRS, but it is still not clear if specific PKC isoforms regulate IR signaling directly or through IRS-1. PKCalpha may regulate IRS activity in response to insulin. We investigated the possibility that PKCalpha may be important in insulin signaling. Studies were conducted on skeletal muscle in adult mice and on L6 skeletal cells. PKCalpha is constitutively associated with IRS-1, and insulin stimulation of PKCalpha causes disassociation of the two proteins within 5 min. Blockade of PKCalpha inhibited insulin-induced disassociation of PKCalpha from IRS1. Selective inhibition of PKCalpha increased the ability of insulin to reduce blood glucose levels. Insulin stimulation activates PKB and increases the association of PKCalpha with PKB. Blockade of PKCalpha increased threonine phosphorylation of PKB. We suggest that PKCalpha regulates insulin signaling in skeletal muscle through its disassociation from IRS-1 and association with PKB.


Subject(s)
Protein Kinase C-alpha/metabolism , Receptor, Insulin/metabolism , Signal Transduction/physiology , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Blood Glucose/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Insulin/pharmacology , Insulin Receptor Substrate Proteins , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase C-alpha/antagonists & inhibitors , Protein Kinase C-alpha/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Time Factors
19.
J Pineal Res ; 40(2): 144-52, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16441551

ABSTRACT

Regulators of G protein signaling (RGS) are proteins that bind specifically to activated Galpha subunits of heterotrimeric G proteins to terminate signaling by both Galpha and Gbetagamma subunits. Signal-induced RGS redistribution may affect their activity in G protein-mediated signaling. We have previously shown that melatonin and the cell permeable cGMP analog 8-bromo cGMP, which lead to protein kinase C (PKC) activation, enhanced cytoplasmic distribution of RGS10 and RGS2 in prostate carcinoma PC3-AR cells. In the present study, we transfected PC3-AR cells with myc-tagged Galphai/Galphaq specific RGS proteins RGS2, RGS4 and RGS10 and examined the effects of melatonin, 8-bromo cGMP and PKC inhibitors on their nuclear-cytoplasmic partitioning. RGS10 and RGS2 were predominantly localized in the nucleus and perinuclear regions whereas RGS4 was mostly cytoplasmic in the PC3-AR cells. Melatonin and the cell permeable cGMP analog 8-bromo cGMP, previously found to activate PKCalpha in the PC3-AR cells, enhanced cytoplasmic localization of RGS10 and RGS2 but induced nuclear accumulation of RGS4. The isozyme specific PKC inhibitor GO6976 (PKCalpha and PKCbeta1) but not hispidin (PKCbeta) negated the effects of melatonin on RGS10, RGS2 and RGS4 localization. These findings indicate that PKCalpha, a downstream effector of the melatonin receptor, differentially affects nuclear/cytoplasmic localization of both Galphai and Galphaq specific RGS proteins. These observations provide further insight into melatonin's ability to fine-tune multiple membrane G-proteins signaling in cells.


Subject(s)
Melatonin/physiology , Protein Kinase C-alpha/physiology , RGS Proteins/metabolism , Animals , Cattle , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Mice , Protein Kinase C-alpha/antagonists & inhibitors , Second Messenger Systems
20.
Cell Signal ; 18(2): 183-93, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16095881

ABSTRACT

Recent studies in our laboratories have shown that Protein Kinase C delta (PKCdelta) is essential for insulin-induced glucose transport in skeletal muscle, and that insulin rapidly stimulates PKCdelta activity skeletal muscle. The purpose of this study was to examine mechanisms of regulation of PKCdelta protein availability. Studies were done on several models of mammalian skeletal muscle and utilized whole cell lysates of differentiated myotubes. PKCdelta protein levels were determined by Western blotting techniques, and PKCdelta RNA levels were determined by Northern blotting, RT-PCR and Real-Time RT-PCR. Insulin stimulation increased PKCdelta protein levels in whole cell lysates. This effect was not due to an inhibition by insulin of the rate of PKCdelta protein degradation. Insulin also increased 35S-methionine incorporation into PKCdelta within 5-15 min. Pretreatment of cells with transcription or translation inhibitors abrogated the insulin-induced increase in PKCdelta protein levels. We also found that insulin rapidly increased the level of PKCdelta RNA, an effect abolished by inhibitors of transcription. The insulin-induced increase in PKCdelta expression was not reduced by inhibition of either PI3 Kinase or MAP kinase, indicating that these signaling mechanisms are not involved, consistent with insulin activation of PKCdelta. Studies on cells transfected with the PKCdelta promoter demonstrate that insulin activated the promoter within 5 min. This study indicates that the expression of PKCdelta may be regulated in a rapid manner during the course of insulin action in skeletal muscle and raise the possibility that PKCdelta may be an immediate early response gene activated by insulin.


Subject(s)
Insulin/pharmacology , Muscle, Skeletal/enzymology , Protein Kinase C-delta/genetics , Animals , Cells, Cultured , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Kinetics , Mice , Muscle, Skeletal/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Kinase C-delta/biosynthesis , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/biosynthesis , Rats , Transcription, Genetic/drug effects , Transcriptional Activation , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...