Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Cancer Res Clin Oncol ; 133(11): 793-808, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17661083

ABSTRACT

The protein kinase C (PKC) family represents a large group of phospholipid dependent enzymes catalyzing the covalent transfer of phosphate from ATP to serine and threonine residues of proteins. Phosphorylation of the substrate proteins induces a conformational change resulting in modification of their functional properties. The PKC family consists of at least ten members, divided into three subgroups: classical PKCs (alpha, betaI, betaII, gamma), novel PKCs (delta, epsilon, eta, theta), and atypical PKCs (zeta, iota/lambda). The specific cofactor requirements, tissue distribution, and cellular compartmentalization suggest differential functions and fine tuning of specific signaling cascades for each isoform. Thus, specific stimuli can lead to differential responses via isoform specific PKC signaling regulated by their expression, localization, and phosphorylation status in particular biological settings. PKC isoforms are activated by a variety of extracellular signals and, in turn, modify the activities of cellular proteins including receptors, enzymes, cytoskeletal proteins, and transcription factors. Accordingly, the PKC family plays a central role in cellular signal processing. Accumulating data suggest that various PKC isoforms participate in the regulation of cell proliferation, differentiation, survival and death. These findings have enabled identification of abnormalities in PKC isoform function, as they occur in several cancers. Specifically, the initiation of squamous cell carcinoma formation and progression to the malignant phenotype was found to be associated with distinct changes in PKC expression, activation, distribution, and phosphorylation. These studies were recently further extended to transgenic and knockout animals, which allowed a more direct analysis of individual PKC functions. Accordingly, this review is focused on the involvement of PKC in physiology and pathology of the skin.


Subject(s)
Protein Kinase C/physiology , Signal Transduction/physiology , Skin Neoplasms/enzymology , Skin/enzymology , Animals , Epithelium/enzymology , Humans , Isoenzymes
2.
Exp Neurol ; 194(1): 97-105, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15899247

ABSTRACT

The neurotrophic factor glial cell line-derived neurotrophic factor (GDNF) may have therapeutic potential for preventing and treating cocaine addiction. Previously, we found that transplantation of a GDNF-expressing astrocyte cell line into the striatum and nucleus accumbens attenuates cocaine-seeking behavior in Sprague-Dawley rats. However, as a potential treatment for humans, cell transplantation presents several technical and ethical complications. Nanoparticulate systems are a safe and effective method for introducing exogenous compounds into the brain. Therefore, we examined the effect of GDNF-conjugated nanoparticles microinjected into the striatum and nucleus accumbens on cocaine self-administration in rats. GDNF-conjugated nanoparticles blocked the acquisition of cocaine self-administration compared to control treatments. Furthermore, a cocaine dose response demonstrated that decreased lever response in rats that received GDNF-conjugated nanoparticles persisted after substitution with different cocaine doses. This effect is not due to a non-specific disruption of locomotor or operant behavior, as seen following a water operant task. The current study is one of the first demonstrations that drug-conjugated nanoparticles may be effective in treating brain disorders. These findings suggest that GDNF-conjugated nanoparticles may serve as a novel potential treatment for drug addiction.


Subject(s)
Brain/drug effects , Cocaine-Related Disorders/drug therapy , Cocaine/antagonists & inhibitors , Ferric Compounds/administration & dosage , Nanostructures , Nerve Growth Factors/administration & dosage , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/metabolism , Brain/physiopathology , Cocaine/adverse effects , Cocaine-Related Disorders/physiopathology , Cocaine-Related Disorders/prevention & control , Conditioning, Operant/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Routes , Glial Cell Line-Derived Neurotrophic Factor , Male , Microinjections/methods , Nanostructures/chemistry , Nanotechnology/methods , Nerve Growth Factors/chemistry , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiopathology , Rats , Rats, Sprague-Dawley , Self Administration , Treatment Outcome
3.
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
4.
Cancer Res ; 61(11): 4591-8, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11389095

ABSTRACT

In mammalian epidermis, expression of the alpha6beta4 integrin is restricted to the hemidesmosome complexes, which connect the proliferative basal cell layer with the underlying basement membrane. Keratinocyte differentiation is associated with down-regulation of alpha6beta4 expression and detachment of keratinocytes from the basement membrane. Neoplastic keratinocytes delay maturation, proliferate suprabasally, and retain the expression of the alpha6beta4 integrin in suprabasal cells disassociated from the hemidesmosomes. We now show that the alpha6beta4 integrin is a substrate for serine phosphorylation by protein kinase C in keratinocytes. Furthermore, protein kinase C-mediated phosphorylation of alpha6beta4 is associated with redistribution of this integrin from the hemidesmosome to the cytosol. Specifically, in vitro kinase assays identified the protein kinase Cdelta as the primary isoform phosphorylating alpha6 and beta4 integrin subunits. Using recombinant protein kinase C adenoviruses, overexpression of protein kinase Cdelta but not protein kinase Calpha in primary keratinocytes increased beta4 serine phosphorylation, decreased alpha6beta4 localization to the hemidesmosome complexes, and reduced keratinocyte attachment. Taken together, these results establish a link between protein kinase Cdelta-mediated serine phosphorylation of alpha6beta4 integrin and its effects on alpha6beta4 subcellular localization and keratinocyte attachment to the laminin underlying matrix.


Subject(s)
Antigens, Surface/metabolism , Hemidesmosomes/metabolism , Integrins/metabolism , Isoenzymes/metabolism , Keratinocytes/metabolism , Protein Kinase C/metabolism , Animals , Antigens, Surface/physiology , Cell Adhesion/physiology , Enzyme Activation , Hemidesmosomes/physiology , Homeostasis/physiology , Integrin alpha6beta4 , Integrins/physiology , Isoenzymes/physiology , Keratinocytes/cytology , Keratinocytes/enzymology , Mice , Mice, Inbred BALB C , Phosphorylation , Protein Kinase C/physiology , Protein Kinase C-delta
5.
Diabetes ; 50(7): 1627-35, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11423485

ABSTRACT

Altered skin wound healing is a common cause of morbidity and mortality among diabetic patients. However, the molecular mechanisms whereby diabetes alters skin physiology have not been elucidated. In this study, we investigated the relative roles of hyperglycemia, insulin, and IGF-I, all of which are abnormal in diabetes, in primary murine skin keratinocytes. These cells proliferate and differentiate in vitro in a manner similar to skin in vivo. It was found that in the presence of high glucose (20 mmol/l), the glucose transport rate of primary proliferating or differentiating keratinocytes was downregulated, whereas at 2 mmol/l glucose, the transport rate was increased. These changes were associated with changes in the GLUT1 expression and with changes in the affinity constant (K(m)) of the transport. Exposure to high glucose was associated with changes in cellular morphology, as well as with decreased proliferation and enhancement of Ca(2+)-induced differentiation of keratinocytes. Furthermore, in the presence of high glucose, ligand-induced IGF-I receptor but not insulin receptor (IR) autophosphorylation was decreased. Consequently, in high glucose, the effects of IGF-I on glucose uptake and keratinocyte proliferation were inhibited. Interestingly, lack of IR expression in IR-null keratinocytes abolished insulin-induced glucose uptake and partially decreased insulin- and IGF-I-induced proliferation, demonstrating the direct involvement of the IR in these processes. Our results demonstrate that hyperglycemia and impaired insulin signaling might be directly involved in the development of chronic complications of diabetes by impairing glucose utilization of skin keratinocytes as well as skin proliferation and differentiation.


Subject(s)
Glucose/pharmacology , Keratinocytes/drug effects , Animals , Biological Transport, Active , Calcium/pharmacology , Cell Differentiation/drug effects , Cell Division/drug effects , Cells, Cultured , Diabetes Complications , Down-Regulation , Female , Glucose/administration & dosage , Glucose/pharmacokinetics , Glucose Transporter Type 1 , Homeostasis , Insulin-Like Growth Factor I/pharmacology , Keratinocytes/metabolism , Kinetics , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Monosaccharide Transport Proteins/biosynthesis , Phosphorylation , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Receptor, Insulin/physiology , Wound Healing
6.
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
7.
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
8.
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
9.
Endocrinology ; 142(3): 1234-41, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181540

ABSTRACT

Impaired wound healing of skin is one of the most serious complications of diabetes. However, the pathogenesis of this process is not known, and it is unclear whether impaired insulin signaling could directly affect skin physiology. To elucidate the role of insulin in skin, we studied skin insulin receptor (IR) null mice. The morphology of the skin of newborn IR null mice was normal; however, these mice exhibited decreased proliferation of skin keratinocytes and changes in expression of skin differentiation markers. Due to the short life span of the IR null mice, further characterization was performed in cultured skin keratinocytes that can be induced to differentiate in vitro, closely following the maturation pattern of epidermis in vivo. It was found that despite a compensatory increase in the insulin-like growth factor I receptor autophosphorylation, differentiation of cultured IR null keratinocytes was markedly impaired. In vitro proliferation was not affected as much. Furthermore, although the basal glucose transport system of the null mice was not defective, the insulin-induced increase in glucose transport was abrogated. These results suggest that insulin regulates, via the IR, the differentiation and glucose transport of skin keratinocytes, whereas proliferation is affected by the diabetes milieu of IR knockout mice.


Subject(s)
Receptor, Insulin/physiology , Skin/cytology , Animals , Biological Transport/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cells, Cultured , Diabetes Complications , Glucose/metabolism , Insulin/physiology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout/genetics , Phosphorylation , Receptor, Insulin/deficiency , Receptor, Insulin/genetics , Receptors, Somatomedin/metabolism , Reference Values , Signal Transduction/physiology , Skin/metabolism , Skin Diseases/etiology
10.
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
11.
Am J Respir Crit Care Med ; 162(6): 2079-86, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11112118

ABSTRACT

Although tuberculosis (TB) screening of immigrants has been conducted for over 50 yr in many industrialized countries, its cost- effectiveness has never been evaluated. We prospectively compared the yield and cost-effectiveness of two immigrant TB screening programs, using close-contact investigation and passive case detection. Study subjects included all immigration applicants undergoing radiographic screening, already arrived immigrants requiring surveillance for inactive TB, and close contacts of active cases resident in Montreal, Quebec, Canada, who were referred from June 1996 to June 1997 to the Montreal Chest Institute (MCI), a referral center specializing in respiratory diseases. For all subjects seen, demographic data, investigations, diagnoses, and therapy were abstracted from administrative data bases and medical charts. Estimated costs of detecting and treating each prevalent active case and preventing future active cases, based on federal and provincial health reimbursement schedules, were compared with the costs for passively diagnosed cases of active TB. Over a period of 1 yr, the three programs detected 27 cases of prevalent active TB and prevented 14 future cases. As compared with passive case detection, close-contact investigation resulted in net savings of $815 for each prevalent active case detected and treated and of $2,186 for each future active case prevented. The incremental cost to treat each case of prevalent active TB was $39,409 for applicant screening and $24,225 for surveillance, and the cost of preventing each case was $33,275 for applicants and $65,126 for surveillance. Close-contact investigation was highly cost effective and resulted in net savings. Immigrant applicant screening and surveillance programs had a significant impact but were much less cost effective, in large part because of substantial operational problems.


Subject(s)
Contact Tracing/economics , Emigration and Immigration , Mass Screening/economics , Tuberculosis, Pulmonary/economics , Cohort Studies , Contact Tracing/methods , Contact Tracing/statistics & numerical data , Cost-Benefit Analysis/economics , Cost-Benefit Analysis/statistics & numerical data , Emigration and Immigration/statistics & numerical data , Humans , Markov Chains , Mass Screening/methods , Mass Screening/statistics & numerical data , Population Surveillance/methods , Prospective Studies , Quebec , Sensitivity and Specificity , Treatment Outcome , Tuberculosis, Pulmonary/diagnosis , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/transmission
12.
J Biol Chem ; 275(37): 28902-10, 2000 Sep 15.
Article in English | MEDLINE | ID: mdl-10878010

ABSTRACT

p94(fer) and p51(ferT) are two tyrosine kinases that share identical SH2 and kinase domains but differ in their N-terminal regions. To further explore the cellular functions of these two highly related tyrosine kinases, their subcellular distribution profiles and in vivo phosphorylation activity were followed using double immunofluorescence assay. When combined with immunoprecipitation analysis, this assay showed that p94(fer) can lead to the tyrosine phosphorylation and activation of Stat3 but not of Stat1 or Stat2. Native p94(fer) exerted this activity when residing in the cytoplasm. However, modified forms of p94(fer), which are constitutively nuclear, could also lead to the phosphorylation of Stat3. Endogenous Stat3 and p94(fer) co-immunoprecipitated with each other, thus proving the interaction of these two proteins in vivo. Unlike p94(fer), p51(ferT) did not induce the phosphorylation of Stat3 but led to the phosphorylation of other nuclear proteins. Replacing the unique 43-amino acid-long N-terminal tail of p51(ferT) with a parallel segment from the N-terminal tail of p94(fer) did not change the subcellular localization of p51(ferT) but enabled it to activate Stat3. Thus the different N-terminal sequences of p94(fer) and p51(ferT) can affect their ability to induce phosphorylation of Stat3 and most probably direct their different cellular functions.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins/physiology , Trans-Activators/metabolism , 3T3 Cells , Animals , CHO Cells , COS Cells , Cricetinae , DNA/metabolism , Enzyme Activation , Mice , Phosphorylation , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/chemistry , STAT3 Transcription Factor , Tyrosine/metabolism
13.
J Invest Dermatol ; 115(1): 24-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10886503

ABSTRACT

The insulin receptor and the insulin-like growth factor-1 receptor are widely expressed tyrosine kinases that mediate insulin and insulin-like growth factor-1 signaling. Both receptors are expressed in many cells in which insulin stimulation does not result in an increase in glucose transport, and the distinct role of the insulin receptor in these tissues, is not known. We have studied the regulation of insulin receptor and insulin-like growth factor-1 receptor in the differentiation of cultured murine keratinocytes. Both receptors are expressed in skin keratinocytes and their expression was unchanged in all stages of calcium-induced differentiation. Insulin binding to skin keratinocytes, however, increased during calcium-induced differentiation, whereas insulin-like growth factor-1 binding decreased. Ligand-induced autophosphorylation was also changed during differentiation. In proliferating keratinocytes both receptors became phosphorylated upon ligand binding, insulin-like growth factor-1 receptor to a greater extent. Terminal differentiation resulted in a decrease in insulin receptor autophosphorylation, whereas insulin-like growth factor-1 receptor autophosphorylation was abolished. There was no change in the cellular localization of the proteins, their intrinsic activity, or their internal structure. Finally, due to the change in the receptor's activity during keratinocyte differentiation, the role of insulin and insulin-like growth factor-1 in the differentiation process was examined. The expected increase in the expression of keratins 1 and 10 during calcium-induced differentiation was facilitated in the presence of insulin, whereas this induction was inhibited in the presence of insulin-like growth factor-1. In conclusion, these results demonstrate that insulin and insulin-like growth factor-1 signaling pathways are differentially involved in skin differentiation, suggesting that abnormal insulin signaling, as occurs in diabetes, may lead to skin pathology.


Subject(s)
Keratinocytes/chemistry , Receptor, IGF Type 1/physiology , Receptor, Insulin/physiology , Animals , Animals, Newborn , Biotinylation , Blotting, Western , Calcium/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Hybridization, Genetic/physiology , Keratinocytes/cytology , Mice , Protein-Tyrosine Kinases , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Skin/cytology
14.
Biol Reprod ; 62(6): 1564-70, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10819756

ABSTRACT

Protein kinase C (PKC), the major cell target for tumor-promoting phorbol esters, plays a central role in signal transduction pathways. In many biological systems where Ca(2+) serves as a second messenger, regulatory control is mediated by PKC. The activation of PKC depends on its binding to RACK1 receptor, which is an intracellular protein anchor for activated PKC. We demonstrate that the conventional PKC (cPKC) isoforms, PKC-alpha, PKC-betaI, and PKC-betaII, as well as RACK1, are expressed in mouse oocytes (germinal vesicle [GV]) and mature eggs (metaphase II [MII]). In GV oocytes, PKC-alpha, PKC-betaII, and RACK1 were uniformly distributed in the cytoplasm, while PKC-betaI was localized in the cytoplasm and in the plasma membrane as well. Treatment of GV oocytes with the biologically active phorbol ester, 12-o-tetradecanoyl phorbol-13-acetate (TPA), resulted in a rapid translocation of the cytosolic PKC-alpha, but not PKC-betaI, PKC-betaII, or RACK1, to the plasma membrane. This was associated with inhibition of GV breakdown. In MII eggs (17 h post-hCG), PKC-alpha was uniformly distributed in the cytoplasm while PKC-betaI and -betaII were distributed in the cytoplasm and in the plasma membrane as well. Treatment with TPA resulted in a rapid translocation of PKC-alpha from the cytoplasm to the plasma membrane and a significant decrease of PKC-betaI throughout the cytoplasm, while it also remained in the cell periphery. No change in the distribution of PKC-betaII or RACK1 was observed. TPA also induced pronucleus formation. Physiological activation of MII eggs by sperm induced cortical granule exocytosis associated with significant translocation of PKC-alpha and -betaI, but not -betaII, to the plasma membrane. Overall, these results suggest a possible involvement of cPKC isoforms in the mechanism of mouse oocyte maturation and egg activation.


Subject(s)
Isoenzymes/analysis , Oocytes/enzymology , Oocytes/physiology , Protein Kinase C/analysis , Animals , Biological Transport/drug effects , Cell Membrane/enzymology , Cytoplasm/enzymology , Exocytosis , Female , Isoenzymes/metabolism , Male , Mice , Mice, Inbred BALB C , Oocytes/ultrastructure , Peptides/analysis , Peptides/metabolism , Protein Kinase C/metabolism , Receptors for Activated C Kinase , Sperm-Ovum Interactions , Tetradecanoylphorbol Acetate/pharmacology
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.
Cell Growth Differ ; 10(2): 113-29, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10074905

ABSTRACT

p94fer and p51ferT are two tyrosine kinases that are encoded by differentially spliced transcripts of the FER locus in the mouse. The two tyrosine kinases share identical SH2 and kinase domains but differ in their NH2-terminal amino acid sequence. Unlike p94fer, the presence of which has been demonstrated in most mammalian cell lines analyzed, the expression of p51ferT is restricted to meiotic cells. Here, we show that the two related tyrosine kinases also differ in their subcellular localization profiles. Although p51ferT accumulates constitutively in the cell nucleus, p94fer is cytoplasmic in quiescent cells and enters the nucleus concomitantly with the onset of S phase. The nuclear translocation of the FER proteins is driven by a nuclear localization signal (NLS), which is located within the kinase domain of these enzymes. The functioning of that NLS depends on the integrity of the kinase domain but was not affected by inactivation of the kinase activity. The NH2 terminus of p94fer dictated the cell cycle-dependent functioning of the NLS of FER kinase. This process was governed by coiled-coil forming sequences that are present in the NH2 terminus of the kinase. The regulatory effect of the p94fer NH2-terminal sequences was not affected by kinase activity but was perturbed by mutations in the kinase domain ATP binding site. Ectopic expression of the constitutively nuclear p51ferT in CHO cells interfered with S-phase progression in these cells. This was not seen in p94fer-overexpressing cells. The FER tyrosine kinases seem, thus, to be regulated by novel mechanisms that direct their different subcellular distribution profiles and may, consequently, control their cellular functioning.


Subject(s)
Adenosine Triphosphate/metabolism , Cell Cycle/physiology , Cell Nucleus/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Antibodies, Monoclonal , Aphidicolin/pharmacology , Blotting, Western , Bromodeoxyuridine/metabolism , CHO Cells , COS Cells , Cell Count , Cell Division , Cricetinae , Cytoplasm/metabolism , DNA/metabolism , Fibroblasts/metabolism , Flow Cytometry , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Protein Binding , Time Factors
18.
Vaccine ; 17(3): 201-4, 1999 Jan 21.
Article in English | MEDLINE | ID: mdl-9987155

ABSTRACT

Administration of vaccinia immune globulin (VIG), derived from vaccinated healthy adult volunteers, is the treatment-of-choice for patients suffering from severe complications following smallpox vaccination. The present study was aimed to determine the time interval after vaccination, at which the highest titer of neutralizing antibodies is obtained. Ninety-nine 18-year-old soldiers, immunized with vaccinia virus at birth, participated in the study, 87 of whom had detectable antibodies against vaccinia virus prior to re-vaccination. Their initial average neutralizing antibodies titer (NT50) was 27. Fourteen days after re-vaccination the titer reached 152 and then dropped to 136, 119, 110 and 87 at 21, 30, 45 and 60 d, respectively. The titers of vaccinia antibodies induced in vaccinees without detectable antibodies at the start of the study, were significantly lower and the titers observed after re-vaccination were: 62, 56, 66, 38 and 34, at 14, 21, 30, 45 and 60 d, respectively. In an additional study, 65 volunteers vaccinated at birth and again at the age of 8 years old were re-vaccinated. Fourteen days later their NT50 was higher than those vaccinated only at birth. It can be concluded that bleeding of vaccinees 14 d following re-vaccination is the preferable time for the preparation of VIG.


Subject(s)
Antibodies, Viral/biosynthesis , Vaccination , Vaccinia virus/immunology , Adolescent , Follow-Up Studies , Humans , Immunization Schedule , Infant, Newborn , Kinetics , Neutralization Tests , Reference Values , Retreatment
19.
Proc Assoc Am Physicians ; 110(4): 333-9, 1998.
Article in English | MEDLINE | ID: mdl-9686681

ABSTRACT

Insulin plays a central role in regulating cellular growth in addition to its classic effects to regulate fuel metabolism. In a previous study, we have identified a patient who was homozygous for a deletion of the insulin receptor gene. In our current investigation, we used cultured skin fibroblasts from this patient as a model system in which to investigate the mechanisms whereby insulin regulates cellular growth in vitro. After cell division, skin fibroblasts from normal individuals migrate on the tissue culture plate and appear to be distributed randomly over the surface of the plate. In contrast, the patient's cells grew in clumps. Furthermore, the patient's fibroblasts exhibited a marked increase in the expression of several integrin subunits, especially the alpha5- and beta1-subunits that comprise the fibronectin receptor. Because the cellular growth pattern was restored to normal when cells were cultivated in the presence of blocking antibodies directed against either alpha5- or beta1-integrin subunits, we infer that increased expression of alpha5beta1-integrin may be the cause of the observed abnormality in the growth of the patient's cells in vitro. Furthermore, insulin stimulation led to downregulation of the levels of the alpha5- and beta1-integrin subunits in normal human fibroblasts but not in the patient's cells that lacked insulin receptors. Taken together, these data suggest that insulin's ability to regulate the expression of cell surface integrins may contribute to the mechanisms whereby insulin regulates cell growth. In light of the important role of integrins in mediating interactions between cells and the basement membrane, we suggest that dysregulation of integrin expression might contribute to the abnormalities in the structure of the basement membranes associated with the chronic microvascular complications of diabetes.


Subject(s)
Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Integrins/physiology , Receptor, Insulin/physiology , Receptors, Vitronectin , Cells, Cultured , Diabetes Mellitus/genetics , Fibroblasts/drug effects , Fibroblasts/pathology , Gene Deletion , Humans , Insulin/pharmacology , Integrins/antagonists & inhibitors , Integrins/biosynthesis , Integrins/deficiency , Integrins/genetics , Receptor, Insulin/genetics
20.
Cancer Res ; 58(7): 1435-43, 1998 Apr 01.
Article in English | MEDLINE | ID: mdl-9537245

ABSTRACT

Retinoic acid (RA) was topically applied to the skin of Sencar mice during the promotion phase of specific tumor induction protocols that produce papillomas at low (12-O-tetradecanoylphorbol-13-acetate promoted, TPA) or high (mezerein-promoted) risk for premalignant progression and malignant conversion. RA consistently reduced the yield of papillomas and carcinomas in both protocols, but the frequency of malignant conversion in papillomas that emerged during RA treatment was not reduced. When TPA was reapplied after cessation of RA treatment, the number of papillomas increased 2-fold, suggesting that RA had not eliminated initiated cells. In vitro, RA prevented the emergence of transformed keratinocytes in an assay that mimics malignant conversion, suggesting that RA can suppress conversion if applied during the stage of premalignant progression. Examination of tumor markers at weeks 14 and 22 of the tumor-induction experiments in vivo indicated that papillomas evolving during RA treatment exhibited a phenotype of high progression risk, even in the TPA-promoted groups. In the majority of these tumors, the alpha6beta4 integrin and retinoid X receptor alpha transcripts were detected suprabasally, indicating an advanced state of premalignant progression. RA-treated tumors also expressed higher levels of transcripts for transforming growth factor (TGF)-beta1 and localized TGF-beta1 peptide in the basal portions of the tumor fronds. Because up-regulated expression of TGF-beta1 suppresses papilloma formation, these studies suggest a mechanism whereby RA can prevent papilloma eruption via a TGF-beta intermediate, but papillomas resistant to RA may have altered TGF-beta signaling and progress to carcinomas at an increased frequency.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Basal Cell/prevention & control , Diterpenes , Papilloma/prevention & control , Precancerous Conditions/drug therapy , Skin Neoplasms/prevention & control , Tretinoin/therapeutic use , Administration, Topical , Animals , Biomarkers, Tumor/analysis , Carcinogens , Carcinoma, Basal Cell/metabolism , Cell Transformation, Neoplastic , Disease Progression , Female , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred SENCAR , Phenotype , Receptors, Retinoic Acid/biosynthesis , Retinoid X Receptors , Risk Factors , Terpenes , Tetradecanoylphorbol Acetate , Transcription Factors/biosynthesis , Transforming Growth Factor beta/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL