Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
Add more filters










Publication year range
1.
J Endocrinol ; 230(3): 291-307, 2016 09.
Article in English | MEDLINE | ID: mdl-27411561

ABSTRACT

Insulin resistance results from impaired insulin signaling in target tissues that leads to increased levels of insulin required to control plasma glucose levels. The cycle of hyperglycemia and hyperinsulinemia eventually leads to pancreatic cell deterioration and death by a mechanism that is yet unclear. Insulin induces ROS formation in several cell types. Furthermore, death of pancreatic cells induced by oxidative stress could be potentiated by insulin. Here, we investigated the mechanism underlying this phenomenon. Experiments were done on pancreatic cell lines (Min-6, RINm, INS-1), isolated mouse and human islets, and on cell lines derived from nonpancreatic sources. Insulin (100nM) for 24h selectively increased the production of ROS in pancreatic cells and isolated pancreatic islets, but only slightly affected the expression of antioxidant enzymes. This was accompanied by a time- and dose-dependent decrease in cellular reducing power of pancreatic cells induced by insulin and altered expression of several ER stress response elements including a significant increase in Trb3 and a slight increase in iNos The effect on iNos did not increase NO levels. Insulin also potentiated the decrease in cellular reducing power induced by H2O2 but not cytokines. Insulin decreased the expression of MCL-1, an antiapoptotic protein of the BCL family, and induced a modest yet significant increase in caspase 3/7 activity. In accord with these findings, inhibition of caspase activity eliminated the ability of insulin to increase cell death. We conclude that prolonged elevated levels of insulin may prime apoptosis and cell death-inducing mechanisms as a result of oxidative stress in pancreatic cells.


Subject(s)
Apoptosis/drug effects , Hyperinsulinism/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Oxidative Stress/drug effects , Animals , Cell Line , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Humans , Hydrogen Peroxide/metabolism , Hyperinsulinism/chemically induced , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Nitrogen Oxides/metabolism , Signal Transduction/drug effects
2.
Apoptosis ; 15(10): 1165-76, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20544287

ABSTRACT

Insulin resistance results, in part, from impaired insulin signaling in insulin target tissues. Consequently, increased levels of insulin are necessary to control plasma glucose levels. The effects of elevated insulin levels on pancreatic beta (ß) cell function, however, are unclear. In this study, we investigated the possibility that insulin may influence survival of pancreatic ß cells. Studies were conducted on RINm, RINm5F and Min-6 pancreatic ß-cells. Cell death was induced by treatment with H(2)O(2), and was estimated by measurements of LDH levels, viability assay (Cell-Titer Blue), propidium iodide staining and FACS analysis, and mitochondrial membrane potential (JC-1). In addition, levels of cleaved caspase-3 and caspase activity were determined. Treatment with H(2)O(2) increased cell death; this effect was increased by simultaneous treatment of cells with insulin. Insulin treatment alone caused a slight increase in cell death. Inhibition of caspase-3 reduced the effect of insulin to increase H(2)O(2)-induced cell death. Insulin increased ROS production by pancreatic ß cells and increased the effect of H(2)O(2). These effects were increased by inhibition of IR signaling, indicative of an effect independent of the IR cascade. We conclude that elevated levels of insulin may act to exacerbate cell death induced by H(2)O(2) and, perhaps, other inducers of apoptosis.


Subject(s)
Apoptosis , Hydrogen Peroxide/toxicity , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Blotting, Western , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Flow Cytometry , Insulin Resistance , Mice , Oxidative Stress , Rats , Reactive Oxygen Species/metabolism
3.
Mol Cell Endocrinol ; 252(1-2): 82-7, 2006 Jun 27.
Article in English | MEDLINE | ID: mdl-16697522

ABSTRACT

Melatonin induces nuclear exclusion of the androgen receptor (AR) via activation of protein kinase C (PKC). The specific members of the PKC superfamily involved in AR nuclear exclusion were investigated in prostate cancer PC3 cells stably transfected with the wild-type androgen receptor (PC3-AR). PKCalpha was essentially cytoplasmic whereas PKCbeta and PKCepsilon were essentially membranal, suggesting their constitutive activity in the PC3-AR cells. Melatonin treatment induced membrane association of PKCalpha in a time and dose dependent manner. The PKCalpha and PKCbeta1 specific inhibitor GO6976 and the PKCbeta isoform-specific inhibitor hispidin had no effects on AR localization under basal conditions. However, GO6976 but not hispidin negated the melatonin-mediated nuclear exclusion of the AR. These data indicate that PKCalpha activation is a critical step in AR nuclear exclusion by melatonin. They also imply that PKCalpha-activation is a potentially effective way to control of the AR activity in prostate cancer cells.


Subject(s)
Melatonin/physiology , Signal Transduction/physiology , Cell Line, Tumor , Cell Membrane/enzymology , Cytosol/enzymology , Humans , Kinetics , Male , Prostatic Neoplasms , Protein Kinase C-alpha/physiology
4.
Life Sci ; 77(23): 2977-85, 2005 Oct 21.
Article in English | MEDLINE | ID: mdl-16043194

ABSTRACT

INTRODUCTION: Tumor necrosis factor-alpha (TNFalpha) is a major mediator of insulin resistance. On the other hand, it has been suggested that TNFalpha may facilitate glucose uptake through GLUT 1 expression. We recently found that physical exercise prevented the progression to type 2 diabetes mellitus in diabetes prone Psammomys obesus (sand rat). AIM: The aim of the present study was to characterize the influence of physical exercise on the expression of TNFalpha, its receptor R1 and GLUT 1 in muscle tissue of this animal model. METHODS: Animals were assigned for 4 weeks to four groups: high-energy diet (HC), high-energy diet and exercise (HE), low-energy diet (LC), low-energy diet and exercise (LE). TNFalpha, R1 and GLUT 1 expression were analyzed using Western blot technique. RESULTS: None of the animals in the HE group became diabetic while all the animals in the HC group became diabetic. TNFalpha, its receptor (R1) and GLUT 1 expressions were significantly higher in the two exercising groups (LE and HE) and significantly lower in the HC group compared to the control LC group. CONCLUSIONS: Physical exercise augments the expression of TNFalpha, its receptor R1 and the glucose transporter GLUT 1 in muscle tissue. We suggest that this mechanism may improve glucose uptake through pathways parallel and unrelated to insulin signaling that may include MAPK and/or NO. These biochemical processes contribute to the beneficial effects of physical exercise on the prevention of type 2 diabetes mellitus.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Tumor Necrosis Factor-alpha/metabolism , Animals , Diabetes Mellitus, Type 2/prevention & control , Diet, Reducing , Disease Models, Animal , Energy Intake , Gerbillinae , Glucose Transporter Type 1 , Male , Muscle Proteins/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism
5.
Mol Cell Biol ; 21(22): 7852-61, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11604519

ABSTRACT

Insulin stimulates glucose uptake into skeletal muscle tissue mainly through the translocation of glucose transporter 4 (GLUT4) to the plasma membrane. The precise mechanism involved in this process is presently unknown. In the cascade of events leading to insulin-induced glucose transport, insulin activates specific protein kinase C (PKC) isoforms. In this study we investigated the roles of PKC zeta in insulin-stimulated glucose uptake and GLUT4 translocation in primary cultures of rat skeletal muscle. We found that insulin initially caused PKC zeta to associate specifically with the GLUT4 compartments and that PKC zeta together with the GLUT4 compartments were then translocated to the plasma membrane as a complex. PKC zeta and GLUT4 recycled independently of one another. To further establish the importance of PKC zeta in glucose transport, we used adenovirus constructs containing wild-type or kinase-inactive, dominant-negative PKC zeta (DNPKC zeta) cDNA to overexpress this isoform in skeletal muscle myotube cultures. We found that overexpression of PKC zeta was associated with a marked increase in the activity of this isoform. The overexpressed, active PKC zeta coprecipitated with the GLUT4 compartments. Moreover, overexpression of PKC zeta caused GLUT4 translocation to the plasma membrane and increased glucose uptake in the absence of insulin. Finally, either insulin or overexpression of PKC zeta induced serine phosphorylation of the GLUT4-compartment-associated vesicle-associated membrane protein 2. Furthermore, DNPKC zeta disrupted the GLUT4 compartment integrity and abrogated insulin-induced GLUT4 translocation and glucose uptake. These results demonstrate that PKC zeta regulates insulin-stimulated GLUT4 translocation and glucose transport through the unique colocalization of this isoform with the GLUT4 compartments.


Subject(s)
Glucose/metabolism , Membrane Proteins/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Muscle, Skeletal/metabolism , Protein Kinase C/metabolism , Serine/metabolism , Animals , Biological Transport , Cell Fractionation , Cells, Cultured , Enzyme Activation , Gene Expression , Glucose Transporter Type 4 , Intracellular Membranes/metabolism , Muscle, Skeletal/cytology , Phosphorylation , Protein Kinase C/genetics , R-SNARE Proteins , Rats
6.
Am J Clin Pathol ; 116(3): 377-85, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11554166

ABSTRACT

We determined the pattern of protein kinase C (PKC) isoform expression in human cell lines by Western blotting and immunofluorescent staining techniques. In addition, we examined PKC isoform expression in tissue samples of transitional cell carcinoma (TCC) of the bladder. PKC delta, PKC beta II, and PKC eta were found primarily in the RT4 cell line (low-grade tumor), and PKC zeta was expressed most strongly in the SUP cell line (invasive tumor). In tissue samples of urinary bladder cancer, PKC isoenzymes were expressed differentially as a function of tumor stage and grade; expression of PKC beta II and PKC delta was high in normal tissue and in low-grade tumors and decreased with increasing stage and grade of TCC. The opposite pattern was seen with PKC zeta. The differences in expression of specific isoenzymes as related to levels of malignancy of the cell lines and tissue samples suggest that the PKC family has an important role in normal and neoplastic urothelium.


Subject(s)
Carcinoma, Transitional Cell/enzymology , Protein Kinase C/metabolism , Urinary Bladder Neoplasms/enzymology , Blotting, Western , Carcinoma, Transitional Cell/pathology , Fluorescent Antibody Technique , Humans , Isoenzymes/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured/drug effects , Urinary Bladder Neoplasms/pathology
7.
Am J Physiol Endocrinol Metab ; 281(1): E16-24, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11404219

ABSTRACT

We have studied the role of the insulin receptor (IR) in metabolic and growth-promoting effects of insulin on primary cultures of skeletal muscle derived from the limb muscle of IR null mice. Cultures of IR null skeletal muscle displayed normal morphology and spontaneous contractile activity. Expression of muscle-differentiating proteins was slightly reduced in myoblasts and myotubes of the IR null skeletal muscle cells, whereas that of the Na+/K+ pump appeared to be unchanged. Insulin-like growth factor receptor (IGFR) expression was higher in myoblasts from IR knockout (IRKO) than from IR wild-type (IRWT) mice but was essentially unchanged in myotubes. Expression of the GLUT-1 and GLUT-4 transporters appeared to be higher in IRKO than in IRWT myoblasts and was significantly greater in myotubes from IRKO than from IRWT cultures. Consistent with GLUT expression, both basal and insulin or insulin-like growth factor I (IGF-I)-stimulated glucose uptakes were higher in IR null skeletal myotubes than in wild-type skeletal myotubes. Interestingly, autophosphorylation of IGFR induced by insulin and IGF-I was markedly increased in IR null skeletal myotubes. These results indicate that, in the absence of IR, there is a compensatory increase in basal as well as in insulin- and IGF-I-induced glucose transport, the former being mediated via increased activation of the IGF-I receptor.


Subject(s)
Glucose/metabolism , Muscle, Skeletal/metabolism , Receptor, IGF Type 1/biosynthesis , Receptor, IGF Type 2/biosynthesis , Receptor, Insulin/genetics , Animals , Biological Transport, Active/physiology , Blotting, Western , Cell Fractionation , Cells, Cultured , Genotype , Insulin-Like Growth Factor I/pharmacology , Mice , Mice, Knockout , Phosphorylation , Precipitin Tests
8.
J Cell Physiol ; 187(3): 365-73, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11319760

ABSTRACT

We have characterized the physiological and biochemical properties of the Na(+)/K(+) pump and its molecular expression in L8 rat muscle cells. Pump properties were measured by [(3)H]ouabain binding and (86)Rb uptake. Scatchard plot analysis of specific ouabain binding indicated the presence of a single family of binding sites with a B(max) of approximately 135 fmol/ mg P and a K(D) of 3.3 x 10(-8). (86)Rb uptake due to specific pump activity was found to be 20% of the total in L8 cells. The results indicated lower affinity of L8 cells for ouabain and lower activity of the pump than that reported for chick or rat skeletal muscle in primary culture. Both the alpha(1) and beta(1) protein and mRNA isoforms were expressed in myoblasts and in myotubes, while the alpha(2), alpha(3), and beta(2) isoforms were not detectable. We attempted to overcome low physiological expression of the Na(+)/K(+) pump by employing a vector expressing an avian high affinity alpha subunit. This allowed identification of the transfected subunit separate from that endogenously expressed in L8 cells. Successful transfection into L8 myoblasts and myotubes was recognized by anti-avian alpha subunit monoclonal antibodies. Fusion index, Na(+)/K(+) pump activity, and the level of the transmembrane resting potential were all significantly greater in transfected L8 (tL8) cells than in non-tL8. The total amount of alpha subunit (avian and rat) in tL8 cells was greater than that (only rat) in non-tL8 cells. This relatively high abundance of the Na(+)/K(+) pump in transfected cells may indicate that avian and rat alpha subunits hybridize to form functional pump complexes.


Subject(s)
Muscle, Skeletal/metabolism , Protein Subunits , Sodium-Potassium-Exchanging ATPase/biosynthesis , Animals , Binding, Competitive/drug effects , Blotting, Western , Cell Division , Cell Line , Chick Embryo , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Gene Expression , Membrane Potentials/physiology , Muscle, Skeletal/cytology , Ouabain/pharmacology , Protein Isoforms/biosynthesis , RNA, Messenger/biosynthesis , Rats , Rubidium/pharmacokinetics , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sodium-Potassium-Exchanging ATPase/genetics , Transfection
9.
Mol Endocrinol ; 15(4): 565-74, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11266508

ABSTRACT

Certain protein kinase C (PKC) isoforms, in particular PKCs beta II, delta, and zeta, are activated by insulin stimulation. In primary cultures of skeletal muscle, PKCs beta II and zeta, but not PKC delta, are activated via a phosphatidylinositol 3-kinase (PI3K)-dependent pathway. The purpose of this study was to investigate the possibility that PKC delta may be activated upstream of PI3K by direct interaction with insulin receptor (IR). Experiments were done on primary cultures of newborn rat skeletal muscle, age 5--6 days in vitro. The time course of insulin-induced activation of PKC delta closely paralleled that of IR. Insulin stimulation caused a selective coprecipitation of PKC delta with IR, and these IR immunoprecipitates from insulin-stimulated cells displayed a striking induction of PKC activity due specifically to PKC delta. To examine the involvement of PKC delta in the IR signaling cascade, we used recombinant adenovirus constructs of wild-type (W.T.) or dominant negative (D.N.) PKC delta. Overexpression of W.T.PKC delta induced PKC delta activity and coassociation of PKC delta and IR without addition of insulin. Overexpression of D.N.PKC delta abrogated insulin- induced coassociation of PKC delta and IR. Insulin-induced tyrosine phosphorylation of IR was greatly attenuated in cells overexpressing W.T.PKC delta, whereas in myotubes overexpressing D.N.PKC delta, tyrosine phosphorylation occurred without addition of insulin and was sustained longer than that in control myotubes. In control myotubes IR displayed a low level of serine phosphorylation, which was increased by insulin stimulation. In cells overexpressing W.T.PKC delta, serine phosphorylation was strikingly high under basal conditions and did not increase after insulin stimulation. In contrast, in cells overexpressing D.N.PKC delta, the level of serine phosphorylation was lower than that in nonoverexpressing cells and did not change notably after addition of insulin. Overexpression of W.T.PKC delta caused IR to localize mainly in the internal membrane fractions, and blockade of PKC delta abrogated insulin-induced IR internalization. We conclude that PKC delta is involved in regulation of IR activity and routing, and this regulation may be important in subsequent steps in the IR signaling cascade.


Subject(s)
Insulin/metabolism , Isoenzymes/metabolism , Muscle, Skeletal/metabolism , Protein Kinase C/metabolism , Receptor, Insulin/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/drug effects , Isoenzymes/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , Phosphorylation , Precipitin Tests , Protein Kinase C/drug effects , Protein Kinase C/genetics , Protein Kinase C-delta , Rats , Receptor, Insulin/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Serine/metabolism , Tyrosine/metabolism
10.
J Biol Chem ; 276(17): 14459-65, 2001 Apr 27.
Article in English | MEDLINE | ID: mdl-11278339

ABSTRACT

Incubation of rat hepatoma Fao cells with insulin leads to a transient rise in Tyr phosphorylation of insulin receptor substrate (IRS) proteins. This is followed by elevation in their P-Ser/Thr content, and their dissociation from the insulin receptor (IR). Wortmannin, a phosphatidylinositol 3-kinase (PI3K) inhibitor, abolished the increase in the P-Ser/Thr content of IRS-1, its dissociation from the IR, and the decrease in its P-Tyr content following 60 min of insulin treatment, indicating that the Ser kinases that negatively regulate IRS-1 function are downstream effectors of PI3K. PKCzeta fulfills this criterion, being an insulin-activated downstream effector of PI3K. Overexpression of PKCzeta in Fao cells, by infection of the cells with adenovirus-based PKCzeta construct, had no effect on its own, but it accelerated the rate of insulin-stimulated dissociation of IR.IRS-1 complexes and the rate of Tyr dephosphorylation of IRS-1. The insulin-stimulated negative regulatory role of PKCzeta was specific and could not be mimic by infecting Fao cells with adenoviral constructs encoding for PKC alpha, delta, or eta. Because the reduction in P-Tyr content of IRS-1 was accompanied by a reduced association of IRS-1 with p85, the regulatory subunit of PI3K, it suggests that this negative regulatory process induced by PKCzeta, has a built-in attenuation signal. Hence, insulin triggers a sequential cascade in which PI3K-mediated activation of PKCzeta inhibits IRS-1 functions, reduces complex formation between IRS-1 and PI3K, and inhibits further activation of PKCzeta itself. These findings implicate PKCzeta as a key element in a multistep negative feedback control mechanism of IRS-1 functions.


Subject(s)
Gene Expression Regulation , Insulin/metabolism , Phosphoproteins/metabolism , Protein Kinase C/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Androstadienes/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Humans , Insulin/physiology , Insulin Receptor Substrate Proteins , Liver Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Precipitin Tests , Protein Isoforms , Rats , Receptor, Insulin/metabolism , Recombinant Proteins/metabolism , Serine/chemistry , Threonine/chemistry , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tyrosine/metabolism , Wortmannin
11.
Diabetes ; 50(2): 255-64, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11272134

ABSTRACT

Insulin and insulin-like growth factor-1 (IGF-1) are members of the family of the insulin family of growth factors, which activate similar cellular downstream pathways. In this study, we analyzed the effects of insulin and IGF-1 on the proliferation of murine skin keratinocytes in an attempt to determine whether these hormones trigger the same signaling pathways. Increasing doses of insulin and IGF-1 promote keratinocyte proliferation in an additive manner. We identified downstream pathways specifically involved in insulin signaling that are known to play a role in skin physiology; these include activation of the Na+/K+ pump and protein kinase C (PKC). Insulin, but not IGF-1, stimulated Na+/K+ pump activity. Furthermore, ouabain, a specific Na+/K+ pump inhibitor, abolished the proliferative effect of insulin but not that of IGF-1. Insulin and IGF-1 also differentially regulated PKC activation. Insulin, but not IGF-1, specifically activated and translocated the PKCB isoform to the membrane fraction. There was no effect on PKC isoforms alpha, eta, epsilon, and zeta, which are expressed in skin. PKC8 overexpression increased keratinocyte proliferation and Na+/K+ pump activity to a degree similar to that induced by insulin but had no affect on IGF-1-induced proliferation. Furthermore, a dominant negative form of PKCdelta abolished the effects of insulin on both proliferation and Na+/K+ pump activity but did not abrogate induction of keratinocyte proliferation induced by other growth factors. These data indicate that though insulin or IGF-1 stimulation induce keratinocyte proliferation, only insulin action is specifically mediated via PKC8 and involves activation of the Na+/K+ pump.


Subject(s)
Insulin-Like Growth Factor I/physiology , Insulin/physiology , Isoenzymes/metabolism , Keratinocytes/cytology , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Biological Transport/drug effects , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured , Enzyme Activation , Genes, Dominant , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Isoenzymes/genetics , Mice , Mice, Inbred BALB C , Protein Kinase C/genetics , Protein Kinase C-delta , Rubidium/pharmacokinetics , Sodium-Potassium-Exchanging ATPase/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism
12.
Eur J Med Chem ; 36(1): 63-74, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11231050

ABSTRACT

The synthesis and biological activities of acidic, basic and neutral types of butyric acid (BA) prodrugs possessing increased aqueous solubility are described. The compounds are butyroyloxyalkyl derivatives of carboxylic acids, which possess functionalities suitable for aqueous solubilization. The anticancer activity of the prodrugs in vitro was evaluated by examining their effect on the growth of human colon, breast and pancreatic carcinoma cell lines, and their solubility in aqueous media was determined. The most promising compounds, with respect to activity and solubility, were found to be the butyroyloxymethyl esters of glutaric 2a and nicotinic acids 4a and phosphoric acid as its diethyl ester 10a, which displayed IC(50) values of 100 microM or lower. These prodrugs are expected to release formaldehyde upon metabolic hydrolysis. The corresponding butyroyloxyethyl esters (2b, 4b and 10b) that release acetaldehyde upon metabolism were significantly less potent. A similar correlation was observed for growth inhibition of the human prostate carcinoma cell lines PC-3 and LnCap and for induction of differentiation and apoptosis in the human myeloid leukemia cell line HL-60. The higher biological activity of the formaldehyde-releasing prodrugs 2a and 10a was further confirmed when induction of hemoglobin (Hb) synthesis in the human erythroleukemic cell line K562 was measured. Moreover, a therapeutic index (IC(50)/ED(50)) of ca. 5 was observed. The acute i.p. toxicity LD(50) in mice for 2a, 2b, 10a and 10b was similar and in the range of 400-600 mg kg(-1). The results obtained support the potential use of the butyric acid prodrugs for the treatment of neoplastic diseases and beta-globin disorders.


Subject(s)
Butyric Acid/chemistry , Butyric Acid/pharmacology , Neoplasms/drug therapy , Prodrugs/chemical synthesis , Prodrugs/pharmacology , Animals , Apoptosis , Cell Division/drug effects , Female , HL-60 Cells/drug effects , Hemoglobins/biosynthesis , Hemoglobins/drug effects , Humans , Lethal Dose 50 , Male , Mice , Mice, Inbred BALB C , Neoplasms/pathology , Prodrugs/chemistry , Solubility , Tumor Cells, Cultured
13.
J Invest Dermatol ; 115(6): 949-54, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11121124

ABSTRACT

Skin is one of the major tissues displaying chronic diabetic complications. We have studied glucose transport following stimulation with insulin and IGF-1 in cultured mouse keratinocytes. In proliferating cells, acute stimulation with insulin and IGF-1 increased glucose uptake. Insulin translocated glucose transporters 1 and 5, whereas IGF-1 translocated glucose transporters 2 and 3. With differentiation, glucose transporter 3 expression increased and the expression of glucose transporters 1, 2, and 5 decreased. No increase in glucose uptake was observed, however, following stimulation with either hormone. These results indicate that insulin and IGF-1 differentially regulate glucose uptake as well as expression and translocation of specific transporters in skin keratinocytes.


Subject(s)
Keratinocytes/chemistry , Monosaccharide Transport Proteins/analysis , Animals , Calcium/pharmacology , Cell Division/drug effects , Cells, Cultured , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/cytology , Mice , Monosaccharide Transport Proteins/drug effects
14.
Appl Immunohistochem Mol Morphol ; 8(2): 166-71, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10937066

ABSTRACT

Protein kinase C (PKC) plays an important role in cellular differentiation and in the malignant process. In an earlier study, it was shown that the expression pattern of PKC isoenzymes is altered in some tumors compared to their corresponding normal tissue. In this study, we evaluated the pattern of PKC isoenzyme immunostaining in bladder transitional cell carcinoma (TCC) of different grades and stages and normal tissue. Twenty-seven TCC samples and six areas of normal bladder mucosa were stained with antibodies specific for the PKC isoenzymes: alpha, beta 1, beta 2, delta, and zeta. The sections were scored for intensity of staining, and the correlation with grade and stage of the tumors was computed. The PKC alpha and beta 2 immunostains were intense in normal urothelium and in all evaluated tumors. PKC beta 1 and delta stains were intense in normal and low-grade and -stage tumors and weak in high-grade and -stage tumors. The opposite trend was found for PKC zeta. PKC isoenzyme expression differs in invasive TCC compared to low-grade, low-stage TCC and normal urothelium. The value of these findings as a marker of tumor aggressiveness should be further assessed.


Subject(s)
Carcinoma, Transitional Cell/enzymology , Protein Kinase C/metabolism , Urinary Bladder Neoplasms/enzymology , Aged , Aged, 80 and over , Carcinoma, Transitional Cell/pathology , Female , Fixatives , Humans , Immunohistochemistry/methods , Isoenzymes/metabolism , Male , Middle Aged , Urinary Bladder Neoplasms/pathology
15.
Mol Endocrinol ; 13(12): 2002-12, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10598577

ABSTRACT

Insulin activates certain protein kinase C (PKC) isoforms that are involved in insulin-induced glucose transport. In this study, we investigated the possibility that activation of PKCdelta by insulin participates in the mediation of insulin effects on glucose transport in skeletal muscle. Studies were performed on primary cultures of rat skeletal myotubes. The role of PKCdelta in insulin-induced glucose uptake was evaluated both by selective pharmacological blockade and by over-expression of wild-type and point-mutated inactive PKCdelta isoforms in skeletal myotubes. We found that insulin induces tyrosine phosphorylation and translocation of PKCdelta to the plasma membrane and increases the activity of this isoform. Insulin-induced effects on translocation and phosphorylation of PKCdelta were blocked by a low concentration of rottlerin, whereas the effects of insulin on other PKC isoforms were not. This selective blockade of PKCdelta by rottlerin also inhibited insulin-induced translocation of glucose transporter 4 (GLUT4), but not glucose transporter 3 (GLUT3), and significantly reduced the stimulation of glucose uptake by insulin. When overexpressed in skeletal muscle, PKCdelta and PKCdelta were both active. Overexpression of PKCdelta induced the translocation of GLUT4 to the plasma membrane and increased basal glucose uptake to levels attained by insulin. Moreover, insulin did not increase glucose uptake further in cells overexpressing PKCdelta. Overexpression of PKCdelta did not affect basal glucose uptake or GLUT4 location. Stimulation of glucose uptake by insulin in cells overexpressing PKCdelta was similar to that in untransfected cells. Transfection of skeletal myotubes with dominant negative mutant PKCdelta did not alter basal glucose uptake but blocked insulin-induced GLUT4 translocation and glucose transport. These results demonstrate that insulin activates PKCdelta and that activated PKCdelta is a major signaling molecule in insulin-induced glucose transport.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Isoenzymes/metabolism , Muscle Proteins , Muscle, Skeletal/enzymology , Nerve Tissue Proteins , Protein Kinase C/metabolism , Acetophenones/pharmacology , Animals , Benzopyrans/pharmacology , Biological Transport/drug effects , Cell Membrane/enzymology , Cells, Cultured , Enzyme Activation , Enzyme Inhibitors/pharmacology , Gene Expression , Glucose Transporter Type 3 , Glucose Transporter Type 4 , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Monosaccharide Transport Proteins/metabolism , Muscle, Skeletal/drug effects , Phosphorylation , Point Mutation , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-delta , Rats , Transfection
16.
Diabetes ; 48(10): 1922-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10512355

ABSTRACT

Several reports indicate that protein kinase C (PKC) plays a role in insulin-induced glucose transport in certain cells. The precise effects of insulin on specific PKC isoforms are as yet unknown. Utilizing primary cultures of rat skeletal muscle, we investigated the possibility that insulin may influence the activation state of PKC isoenzymes by inducing their translocation and tyrosine phosphorylation. This, in turn, may mediate insulin effects on glucose transport. We identified and determined the glucose transporters and PKC isoforms affected by insulin and 12-O-tetradecanoylphorbol-13-acetate (TPA). Insulin and TPA each caused an increase in glucose uptake. Insulin translocated GLUT3 and GLUT4 without affecting GLUT1. In contrast, TPA translocated GLUT1 and GLUT3 without affecting GLUT4. Insulin translocated and tyrosine phosphorylated and activated PKC-beta2 and -zeta; these effects were blocked by phosphatidylinositol 3-kinase (PI3K) inhibitors. TPA translocated and activated PKC-alpha, -beta2, and -delta; these effects were not noticeably affected by PI3K inhibitors. Furthermore, wortmannin significantly inhibited both insulin and TPA effects on GLUT translocation and glucose uptake. Finally, insulin-induced glucose transport was blocked by the specific PKC-beta2 inhibitor LY379196. These results indicate that specific PKC isoenzymes, when tyrosine-phosphorylated, are implicated in insulin-induced glucose transport in primary cultures of skeletal muscle.


Subject(s)
Glucose/metabolism , Insulin/pharmacology , Isoenzymes/metabolism , Muscle, Skeletal/metabolism , Protein Kinase C/metabolism , Tyrosine/metabolism , Animals , Biological Transport, Active/drug effects , Cells, Cultured , Enzyme Inhibitors/pharmacology , Muscle, Skeletal/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Rats , Tetradecanoylphorbol Acetate/pharmacology
17.
J Cell Physiol ; 180(2): 236-44, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10395293

ABSTRACT

Studies from this laboratory have shown that the physiological expression of the Na+/K+ pump in primary cultures of rat skeletal muscle increases with development. The molecular mechanisms underlying these changes are not known. Therefore, we have examined the expression of alpha and beta subunits of the Na+/K+ pump at both the protein and mRNA levels during myogenesis of primary skeletal muscle cell cultures obtained from newborn rats. Protein isoforms were identified by Western blotting techniques with specific monoclonal and polyclonal antibodies and subunit mRNA was studied with specific cDNA probes. Freshly isolated skeletal muscle from newborn rats expressed both alpha1 and alpha2 protein subunits. From day 1 after plating, primary cultures expressed only the alpha1 protein isoform. In contrast, both beta1 and beta2 isoforms were expressed in freshly isolated muscle and in primary cultures, with beta1 expression being stronger in both preparations. Studies on RNA expression showed that mRNA for alpha1, alpha2, beta1, and beta2 isoforms was identified both in freshly isolated muscle and after plating of cells in culture. These findings indicate that the lack of alpha2 protein expression in primary muscle cell cultures reflects a form of posttranscriptional regulation. There did not appear to be a quantitative difference in isoform expression as a function of age or of fusion in spite of developmental increases in Na+/K+ pump activity and its dependence on cell fusion. The lack of expression of the alpha2 subunit isoform suggests that the developmental changes in physiological expression of the Na+/K+ pump in primary cultures of skeletal muscle may be attributable either to the changes in activity of the alpha1 subunit or to differential activities of alphabeta complexes involving either of the beta subunits.


Subject(s)
Isoenzymes/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/enzymology , Sodium-Potassium-Exchanging ATPase/genetics , Animals , Animals, Newborn , Antibodies, Monoclonal , Blotting, Northern , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Chelating Agents/pharmacology , Egtazic Acid/pharmacology , Gene Expression Regulation, Enzymologic , Isoenzymes/analysis , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/enzymology , RNA, Messenger/analysis , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sodium-Potassium-Exchanging ATPase/analysis , Sodium-Potassium-Exchanging ATPase/immunology
18.
J Neurosci Res ; 56(3): 275-83, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10336257

ABSTRACT

We investigated the effects of nerve growth factor (NGF) on expression of K+ channels in cultured skeletal muscle. The channels studied were (1) charybdotoxin (ChTx)-sensitive channels by using a polyclonal antibody raised in rabbits against ChTx, (2) Kv1.5 voltage-sensitive channels, and (3) apamin-sensitive (afterhyperpolarization) channels. Crude homogenates were prepared from cultures made from limb muscles of 1-2-day-old rat pups for identification of ChTx-sensitive and Kv1.5 channels by Western blotting techniques. Apamin-sensitive K+ channels were studied by measurement of specific [125I]-apamin binding by whole cell preparations. ChTx-sensitive channels display a fusion-related increase in expression, and NGF downregulates these channels in both myoblasts and myotubes. Voltage-dependent Kv1.5 channel expression is low in myoblasts and increases dramatically with fusion; NGF induces early expression of these channels and causes expression after fusion to increase even further. NGF downregulates apamin-sensitive channels. NGF increases the rate of fall of the action potential recorded intracellularly from single myotubes with intracellular microelectrodes. The results confirm and extend those of previous studies in showing a functional role for NGF in the regulation of membrane properties of skeletal muscle. Moreover, the findings demonstrate that the different K+ channels in this preparation are regulated in a discoordinate manner. The divergent effects of NGF on expression of different K+ channels, however, do not appear sufficient to explain the NGF-induced increase in the rate of fall of the action potential. The changes during the falling phase may rather be due to increases in channel properties or may result from an increased driving force on the membrane potential secondary to the NGF-induced hyperpolarization.


Subject(s)
Muscle, Skeletal/drug effects , Nerve Growth Factors/pharmacology , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Potassium Channels/physiology , Action Potentials/drug effects , Animals , Animals, Newborn , Apamin/metabolism , Binding Sites , Cell Fusion , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Charybdotoxin/metabolism , Charybdotoxin/pharmacology , Electrophysiology , Gene Expression/drug effects , Ion Channel Gating/drug effects , Kv1.5 Potassium Channel , Molecular Weight , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Rats
19.
J Cardiovasc Pharmacol ; 32(1): 134-8, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9676733

ABSTRACT

The effects of 4-aminopyridine (4-AP) on the transient outward current (I(to)) were investigated in rat ventricular cardiomyocytes at different values of intracellular pH (pHi) and extracellular pH (pHo). The 4-AP was administered either extracellularly (bath application) or intracellularly (diffusion from the intrapipette solution). The 4-AP diminished I(to) given either from inside or outside the cell membrane. The block by extracellularly applied 4-AP (4-APo) of the peak amplitude of I(to) was decreased by external acidification but increased by external alkalinization; conversely, the block by 4-APo was decreased by internal alkalinization but increased by internal acidification. Intracellularly applied 4-AP (3 mM) was more effective at low pHi. Because 4-AP is a tertiary amine and exists in protonated and unprotonated forms, these results are in agreement with the assumption that one major mechanism for 4-AP to block I(to) is to penetrate the cell membrane in its uncharged form and to reach intracellular binding sites in its protonated form.


Subject(s)
4-Aminopyridine/pharmacology , Ion Channels/antagonists & inhibitors , Myocardium/metabolism , Animals , Cell Membrane/drug effects , Cell Membrane/physiology , Extracellular Space/drug effects , Extracellular Space/metabolism , Female , Heart Ventricles/cytology , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hydrogen-Ion Concentration , In Vitro Techniques , Male , Membrane Potentials/drug effects , Myocardium/cytology , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
20.
Isr J Med Sci ; 32(12): 1212-6, 1996 Dec.
Article in English | MEDLINE | ID: mdl-9007156

ABSTRACT

The extensive use of amiodarone as an anti-arrhythmic drug is hampered by numerous side effects and by insufficient knowledge of its cellular action. The use of cell cultures for studying the mechanism of amiodarone action has been questioned, since available information has indicated that the doses employed for the experiments induce cell damage. We have defined conditions to obtain the amiodarone effect on cardiac cells in culture with no detectable damage. Amiodarone, 1 microg/ml, a concentration comparable to serum levels of the drug in acute and chronically treated humans and rats, reduces cell contractions, modifies membrane electrical properties accordingly, increases ATP content, but does not alter cell substructure or change enzyme activities. We strongly support the use of cell cultures for studying the cellular action(s) of amiodarone and offer conditions suitable for such experiments.


Subject(s)
Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Heart/drug effects , Myocardium/cytology , Adenosine Triphosphate/analysis , Animals , Cells, Cultured/drug effects , Drug Evaluation, Preclinical , Electrophysiology , Heart/physiology , Heart Rate/drug effects , Myocardium/chemistry , Myocardium/enzymology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...