Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Commun ; 7: 10686, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26880110

ABSTRACT

Proper regulation of energy storage in adipose tissue is crucial for maintaining insulin sensitivity and molecules contributing to this process have not been fully revealed. Here we show that type II transmembrane protein tenomodulin (TNMD) is upregulated in adipose tissue of insulin-resistant versus insulin-sensitive individuals, who were matched for body mass index (BMI). TNMD expression increases in human preadipocytes during differentiation, whereas silencing TNMD blocks adipogenesis. Upon high-fat diet feeding, transgenic mice overexpressing Tnmd develop increased epididymal white adipose tissue (eWAT) mass, and preadipocytes derived from Tnmd transgenic mice display greater proliferation, consistent with elevated adipogenesis. In Tnmd transgenic mice, lipogenic genes are upregulated in eWAT, as is Ucp1 in brown fat, while liver triglyceride accumulation is attenuated. Despite expanded eWAT, transgenic animals display improved systemic insulin sensitivity, decreased collagen deposition and inflammation in eWAT, and increased insulin stimulation of Akt phosphorylation. Our data suggest that TNMD acts as a protective factor in visceral adipose tissue to alleviate insulin resistance in obesity.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Cell Differentiation/genetics , Insulin Resistance/genetics , Intra-Abdominal Fat/metabolism , Ion Channels/metabolism , Lipogenesis/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/metabolism , Obesity, Morbid/genetics , Adipose Tissue, Brown/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adult , Animals , Blotting, Western , DNA-Binding Proteins/metabolism , Epididymis , Female , Fluorescent Antibody Technique , Glucose Clamp Technique , Humans , Intra-Abdominal Fat/cytology , Intra-Abdominal Fat/pathology , Male , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Middle Aged , Obesity, Morbid/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Uncoupling Protein 1
2.
Indian J Med Res ; 144(5): 741-749, 2016 Nov.
Article in English | MEDLINE | ID: mdl-28361828

ABSTRACT

BACKGROUND & OBJECTIVES: Cervical cancer is a major health problem and a leading cause of death among women in India. Of all the associated risk factors, high-risk human papillomavirus (HPV) infections being the principal aetiologic agent, two HPV vaccines are in use for the control of cervical cancer. The present study was undertaken to explore the knowledge, attitude and practice (KAP) on HPV vaccination among the healthcare providers in India. METHODS: A cross-sectional study was conducted among 590 healthcare professionals from 232 hospitals and 80 PHCs of nine districts of Delhi-NCR (National Capital Region). A total of 590 (526 female, 64 male) healthcare providers were surveyed. RESULTS: Only 47 per cent of respondents recommended young women to get vaccinated against HPV. Majority of respondents (81%) were found to be aware about the existence of vaccines for cervical cancer prevention. District-wise, highest (88.3%) awareness about the existence of vaccines against HPV was reported from Gautam Budh Nagar and lowest (64%) in Faridabad. Although 86 per cent of gynaecologists were aware about the names of HPV vaccines available in the market, only 27 per cent of paramedical staff had this knowledge. There was a significant difference between the respondents from government and private sectors regarding their awareness about HPV vaccines. Lack of awareness about the principal cause, risk factors and symptoms for cervical cancer and HPV vaccination was significantly (P< 0.05) reported in the respondents from paramedical staff category. INTERPRETATION & CONCLUSIONS: The findings reinforce continued medical education of healthcare providers, particularly those from the government sector on HPV vaccination for cervical cancer prevention. Public education is also pertinent for a successful HPV vaccination programme in the country.


Subject(s)
Health Personnel/psychology , Papillomavirus Infections/epidemiology , Papillomavirus Vaccines , Uterine Cervical Neoplasms/epidemiology , Adolescent , Adult , Aged , Female , Health Knowledge, Attitudes, Practice , Humans , India/epidemiology , Male , Middle Aged , Papillomaviridae/pathogenicity , Papillomavirus Infections/psychology , Papillomavirus Infections/virology , Patient Acceptance of Health Care , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Vaccination/trends
3.
J Biol Chem ; 290(24): 15175-84, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25922078

ABSTRACT

The liver is a major site of glucose, fatty acid, and triglyceride (TG) synthesis and serves as a major regulator of whole body nutrient homeostasis. Chronic exposure of humans or rodents to high-calorie diets promotes non-alcoholic fatty liver disease, characterized by neutral lipid accumulation in lipid droplets (LD) of hepatocytes. Here we show that the LD protein hypoxia-inducible gene 2 (Hig2/Hilpda) functions to enhance lipid accumulation in hepatocytes by attenuating TG hydrolysis. Hig2 expression increased in livers of mice on a high-fat diet and during fasting, two states associated with enhanced hepatic TG content. Hig2 expressed in primary mouse hepatocytes localized to LDs and promoted LD TG deposition in the presence of oleate. Conversely, tamoxifen-inducible Hig2 deletion reduced both TG content and LD size in primary hepatocytes from mice harboring floxed alleles of Hig2 and a cre/ERT2 transgene controlled by the ubiquitin C promoter. Hepatic TG was also decreased by liver-specific deletion of Hig2 in mice with floxed Hig2 expressing cre controlled by the albumin promoter. Importantly, we demonstrate that Hig2-deficient hepatocytes exhibit increased TG lipolysis, TG turnover, and fatty acid oxidation as compared with controls. Interestingly, mice with liver-specific Hig2 deletion also display improved glucose tolerance. Taken together, these data indicate that Hig2 plays a major role in promoting lipid sequestration within LDs in mouse hepatocytes through a mechanism that impairs TG degradation.


Subject(s)
Lipolysis/physiology , Liver/metabolism , Neoplasm Proteins/physiology , Triglycerides/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics
4.
Asian Pac J Cancer Prev ; 15(13): 5475-82, 2014.
Article in English | MEDLINE | ID: mdl-25041021

ABSTRACT

Cervical cancer, the second most common malignancy all over the world, is associated with HPV infection. In a developing country like India, lack of early detection and treatment facilities is the main cause for its high burden. Therefore, through our study we e tried to present the current scenario of existing facilities for the detection and treatment of cervical cancer in hospitals and primary health centers (PHCs) of Delhi-NCR region. Data were collected from 312 healthcare facilities including public and private hospitals and PHCs of all nine districts from Delhi-NCR region. Healthcare providers including gynecologists, medical officers, women health care providers and paramedical staff were interviewed, using a questionnaire; the facilities for screening, diagnosing, and treating cervical cancer in each institution were recorded, using a previously designed checklist. Our study has shown that the basic facilities for the detection and treatment of cervical cancer are abhorrently lacking in Public hospitals and PHCs as compared to the Private hospitals in Delhi-NCR region. This study demonstrates that there is an urgent need for more investment in the diagnosis and treatment of cervical cancer facilities in public and rural healthcare facilities of Delhi-NCR region.


Subject(s)
Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/therapy , Developing Countries , Female , Health Facilities , Health Services Needs and Demand , Hospitals , Hospitals, Private , Humans , India , Mass Screening/methods , Rural Population
5.
Mol Cell Biol ; 33(4): 678-87, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23207904

ABSTRACT

Myoblast differentiation into mature myotubes is a critical step in the development and repair of human skeletal muscle. Here we show that small interfering RNA (siRNA)-based silencing of the Ste20-like mitogen-activated protein 4 kinase 4 (Map4k4) in C2C12 myoblasts markedly enhances expression of myogenic differentiation genes, myoblast fusion, and myotube diameter. In contrast, adenovirus-mediated expression of native Map4k4 in C2C12 cells attenuates each of these processes, indicating that Map4k4 is a negative regulator of myogenic differentiation and hypertrophy. Expression of a Map4k4 kinase-inactive mutant enhances myotube formation, suggesting that the kinase activity of Map4k4 is essential for its inhibition of muscle differentiation. Map4k4 regulation of myogenesis is unlikely to be mediated by classic mitogen-activated protein kinase (MAPK) signaling pathways, because no significant difference in phosphorylation of extracellular signal-regulated kinase (ERK), p38, or c-Jun N-terminal kinase (JNK) is observed in Map4k4-silenced cells. Furthermore, silencing of these other MAPKs does not result in a hypertrophic myotube phenotype like that seen with Map4k4 depletion. Uniquely, Map4k4 silencing upregulates the expression of the myogenic regulatory factor Myf5, whose depletion inhibits myogenesis. Furthermore, Myf5 is required for enhancement of myotube formation in Map4k4-silenced cells, while Myf5 overexpression rescues Map4k4-mediated inhibition of myogenic differentiation. These results demonstrate that Map4k4 is a novel suppressor of skeletal muscle differentiation, acting through a Myf5-dependent mechanism.


Subject(s)
Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/cytology , Myoblasts/cytology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation , Cell Line , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Muscle Development , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , RNA Interference , RNA, Small Interfering/genetics , Up-Regulation , NF-kappaB-Inducing Kinase
6.
J Lipid Res ; 52(2): 221-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21097823

ABSTRACT

The lipid droplet-associated fat specific protein 27 (FSP27) suppresses lipolysis and thereby enhances triglyceride accumulation in adipocytes. We and others have recently found FSP27 to be a remarkably short-lived protein (half-life, 15 min) due to its rapid ubiquitination and proteasomal degradation. Thus, we tested the hypothesis that lipolytic agents such as tumor necrosis factor-α (TNF-α) and isoproterenol modulate FSP27 levels to regulate FFA release. Consistent with this concept, we showed that the lipolytic actions of TNF-α, interleukin-1ß (IL-1ß), and IFN-γ are accompanied by marked decreases in FSP27 expression and lipid droplet size in mouse adipocytes. Similar depletion of FSP27 using short interfering RNA (siRNA) mimicked the lipolysis-enhancing effect of TNF-α, while maintaining stable FSP27 levels using expression of hemagglutinin epitope-tagged FSP27 blocked TNF-α-mediated lipolysis. In contrast, we show the robust lipolytic action of isoproterenol is paradoxically associated with increases in FSP27 levels and a delayed degradation rate corresponding to decreased ubiquitination. This catecholamine-mediated increase in FSP27 abundance, probably a feedback mechanism for restraining excessive lipolysis by catecholamines, is mimicked by forskolin or 8-bromo-cAMP treatment and is prevented by the protein kinase A (PKA) inhibitor KT5720 or by PKA depletion using siRNA. Taken together, these data identify the regulation of FSP27 as an important intermediate in the mechanism of lipolysis in adipocytes in response to TNF-α and isoproterenol.


Subject(s)
Isoproterenol/pharmacology , Lipolysis/drug effects , Proteins/metabolism , Tumor Necrosis Factor-alpha/pharmacology , 3T3-L1 Cells , Animals , Mice , Proteins/drug effects , Ubiquitination/drug effects
7.
J Lipid Res ; 51(12): 3455-62, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20810722

ABSTRACT

Cidea, the cell death-inducing DNA fragmentation factor-α-like effector (CIDE) domain-containing protein, is targeted to lipid droplets in mouse adipocytes, where it inhibits triglyceride hydrolysis and promotes lipid storage. In mice, Cidea may prevent lipolysis by binding and shielding lipid droplets from lipase association. Here we demonstrate that human Cidea localizes with lipid droplets in both adipocyte and nonadipocyte cell lines, and we ascribe specific functions to its protein domains. Expression of full-length Cidea in undifferentiated 3T3-L1 cells or COS-1 cells increases total cellular triglyceride and strikingly alters the morphology of lipid droplets by enhancing their size and reducing their number. Remarkably, both lipid droplet binding and increased triglyceride accumulation are also elicited by expression of only the carboxy-terminal 104 amino acids, indicating this small domain directs lipid droplet targeting and triglyceride shielding. However, unlike the full-length protein, expression of the carboxy-terminus causes clustering of small lipid droplets but not the formation of large droplets, identifying a novel function of the N terminus. Furthermore, human Cidea promotes lipid storage via lipolysis inhibition, as the expression of human Cidea in fully differentiated 3T3-L1 adipocytes causes a significant decrease in basal glycerol release. Taken together, these data indicate that the carboxy-terminal domain of Cidea directs lipid droplet targeting, lipid droplet clustering, and triglyceride accumulation, whereas the amino terminal domain is required for Cidea-mediated development of enlarged lipid droplets.


Subject(s)
Adipocytes/metabolism , Apoptosis Regulatory Proteins/analysis , Lipid Metabolism , 3T3-L1 Cells , Adipocytes/cytology , Animals , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Fibroblasts/cytology , Fibroblasts/metabolism , Fluorescent Antibody Technique , Humans , Lipase/metabolism , Lipoproteins/metabolism , Mice , Microscopy, Confocal , Triglycerides/metabolism
8.
J Biol Chem ; 285(36): 27581-9, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20587420

ABSTRACT

Protein kinase B/Akt protein kinases control an array of diverse functions, including cell growth, survival, proliferation, and metabolism. We report here the identification of pleckstrin homology-like domain family B member 1 (PHLDB1) as an insulin-responsive protein that enhances Akt activation. PHLDB1 contains a pleckstrin homology domain, which we show binds phosphatidylinositol PI(3,4)P(2), PI(3,5)P(2), and PI(3,4,5)P(3), as well as a Forkhead-associated domain and coiled coil regions. PHLDB1 expression is increased during adipocyte differentiation, and it is abundant in many mouse tissues. Both endogenous and HA- or GFP-tagged PHLDB1 displayed a cytoplasmic disposition in unstimulated cultured adipocytes but translocated to the plasma membrane in response to insulin. Depletion of PHLDB1 by siRNA inhibited insulin stimulation of Akt phosphorylation but not tyrosine phosphorylation of IRS-1. RNAi-based silencing of PHLDB1 in cultured adipocytes also attenuated insulin-stimulated deoxyglucose transport and Myc-GLUT4-EGFP translocation to the plasma membrane, whereas knockdown of the PHLDB1 isoform PHLDB2 failed to attenuate insulin-stimulated deoxyglucose transport. Furthermore, adenovirus-mediated expression of PHLDB1 in adipocytes enhanced insulin-stimulated Akt and p70 S6 kinase phosphorylation, as well as GLUT4 translocation. These results indicate that PHLDB1 is a novel modulator of Akt protein kinase activation by insulin.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 3T3-L1 Cells , Animals , Blood Proteins/chemistry , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Gene Silencing , Glucose/metabolism , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Mice , Phosphatidylinositol Phosphates/metabolism , Phosphoproteins/chemistry , Phosphorylation/drug effects , Protein Structure, Tertiary , Protein Transport/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Sequence Homology, Amino Acid
9.
J Biol Chem ; 285(9): 6595-603, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20038583

ABSTRACT

The receptor peroxisome proliferator-activated receptor gamma (PPARgamma) is considered a master regulator of adipocyte differentiation and promotes glucose and lipid metabolism in mature adipocytes. We recently identified the yeast Sterile 20 (Ste20) protein kinase ortholog, Map4k4, in an RNA interference-based screen as an inhibitor of PPARgamma expression in cultured adipocytes. Here, we show that RNA interference-mediated silencing of Map4k4 elevates the levels of both PPARgamma1 and PPARgamma2 proteins in 3T3-L1 adipocytes without affecting PPARgamma mRNA levels, suggesting that Map4k4 regulates PPARgamma at a post-transcriptional step. PPARgamma degradation rates are remarkably rapid as measured in the presence of cycloheximide (t(1/2) = 2 h), but silencing Map4k4 had no effect on PPARgamma degradation. However, depletion of Map4k4 significantly enhances [(35)S]methionine/cysteine incorporation into proteins, suggesting that Map4k4 signaling decreases protein translation. We show a function of Map4k4 is to inhibit rapamycin-sensitive mammalian target of rapamycin (mTOR) activity, decreasing 4E-BP1 phosphorylation. In addition, our results show mTOR and 4E-BP1 are required for the increased PPARgamma protein expression upon Map4k4 knockdown. Consistent with this concept, adenovirus-mediated expression of Map4k4 decreased PPARgamma protein levels and mTOR phosphorylation. These data show that Map4k4 negatively regulates PPARgamma post-transcriptionally, by attenuating mTOR signaling and a 4E-BP1-dependent mechanism.


Subject(s)
Adipocytes/metabolism , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , PPAR gamma/antagonists & inhibitors , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/physiology , Signal Transduction , 3T3-L1 Cells , Adaptor Proteins, Signal Transducing , Adipocytes/cytology , Animals , Cell Cycle Proteins , Eukaryotic Initiation Factors , Gene Expression Regulation , Mice , PPAR gamma/biosynthesis , Phosphorylation , Protein Stability , TOR Serine-Threonine Kinases , NF-kappaB-Inducing Kinase
10.
Biotechnol J ; 4(3): 391-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19296443

ABSTRACT

This report describes the immunogenicity and protective efficacy of Escherichia coli-expressed recombinant protective antigen (rPA) in New Zealand White rabbits and Rhesus Macaques against an aerosol challenge with Bacillus anthracis spores (IVRI strain, tox+cap+). A dose-ranging study was performed in which it became evident that the level of anti-PA IgG and toxin-neutralizing antibody titer was directly proportional to the dose of rPA administered. However, the onset time of primary and secondary immune response was not dependent on the dosage. Revaccination of primed animals with the same threshold dose yielded a robust and rapid secondary response. Quantitative differences in peak titers were obtained for both the animal models, in addition to qualitative differences in the immune kinetics. In spite of a weak priming response, the secondary response in rabbits peaked earlier than that in macaques once the booster dose was administered. However, evaluation of the post-challenge quantitative anti-rPA ELISA titer measurements indicated higher titers for non-human primates as compared to the lagomorphs. Importantly, 100% protection was seen for the dosage groups that received > or = 25 microg rPA, following a challenge against a target dose of 1000 LD(50) of aerosolized spores of Bacillus anthracis.


Subject(s)
Anthrax Vaccines/therapeutic use , Anthrax/prevention & control , Antigens, Bacterial/chemistry , Bacillus anthracis/immunology , Bacterial Toxins/chemistry , Spores, Bacterial/immunology , Vaccines, Synthetic/therapeutic use , Aerosols , Animals , Anthrax Vaccines/chemistry , Drug Evaluation, Preclinical , Enzyme-Linked Immunosorbent Assay , Kinetics , Macaca mulatta , Neutralization Tests , Rabbits , Time Factors , Vaccines, Synthetic/chemistry
11.
EMBO Mol Med ; 1(5): 280-7, 2009 Aug.
Article in English | MEDLINE | ID: mdl-20049731

ABSTRACT

Lipodystrophic syndromes are characterized by adipose tissue deficiency. Although rare, they are of considerable interest as they, like obesity, typically lead to ectopic lipid accumulation, dyslipidaemia and insulin resistant diabetes. In this paper we describe a female patient with partial lipodystrophy (affecting limb, femorogluteal and subcutaneous abdominal fat), white adipocytes with multiloculated lipid droplets and insulin-resistant diabetes, who was found to be homozygous for a premature truncation mutation in the lipid droplet protein cell death-inducing Dffa-like effector C (CIDEC) (E186X). The truncation disrupts the highly conserved CIDE-C domain and the mutant protein is mistargeted and fails to increase the lipid droplet size in transfected cells. In mice, Cidec deficiency also reduces fat mass and induces the formation of white adipocytes with multilocular lipid droplets, but in contrast to our patient, Cidec null mice are protected against diet-induced obesity and insulin resistance. In addition to describing a novel autosomal recessive form of familial partial lipodystrophy, these observations also suggest that CIDEC is required for unilocular lipid droplet formation and optimal energy storage in human fat.


Subject(s)
Codon, Nonsense , Diabetes Mellitus/genetics , Insulin Resistance , Lipodystrophy/genetics , Proteins/genetics , 3T3 Cells , Animals , Apoptosis Regulatory Proteins , Base Sequence , Diabetes Mellitus/metabolism , Female , Humans , Lipodystrophy/metabolism , Male , Mice , Molecular Sequence Data , Pedigree , Protein Transport , Proteins/metabolism , Young Adult
12.
Proc Natl Acad Sci U S A ; 105(22): 7833-8, 2008 Jun 03.
Article in English | MEDLINE | ID: mdl-18509062

ABSTRACT

Storage of energy as triglyceride in large adipose-specific lipid droplets is a fundamental need in all mammals. Efficient sequestration of fat in adipocytes also prevents fatty acid overload in skeletal muscle and liver, which can impair insulin signaling. Here we report that the Cide domain-containing protein Cidea, previously thought to be a mitochondrial protein, colocalizes around lipid droplets with perilipin, a regulator of lipolysis. Cidea-GFP greatly enhances lipid droplet size when ectopically expressed in preadipocytes or COS cells. These results explain previous findings showing that depletion of Cidea with RNAi markedly elevates lipolysis in human adipocytes. Like perilipin, Cidea and the related lipid droplet protein Cidec/FSP27 are controlled by peroxisome proliferator-activated receptor gamma (PPARgamma). Treatment of lean or obese mice with the PPARgamma agonist rosiglitazone markedly up-regulates Cidea expression in white adipose tissue (WAT), increasing lipid deposition. Strikingly, in both omental and s.c. WAT from BMI-matched obese humans, expression of Cidea, Cidec/FSP27, and perilipin correlates positively with insulin sensitivity (HOMA-IR index). Thus, Cidea and other lipid droplet proteins define a novel, highly regulated pathway of triglyceride deposition in human WAT. The data support a model whereby failure of this pathway results in ectopic lipid accumulation, insulin resistance, and its associated comorbidities in humans.


Subject(s)
Adipose Tissue, White/metabolism , Apoptosis Regulatory Proteins/metabolism , Insulin Resistance , Triglycerides/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue, White/cytology , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/genetics , Body Mass Index , Carrier Proteins , Humans , Lipolysis , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Obesity/metabolism , PPAR gamma/agonists , PPAR gamma/genetics , PPAR gamma/metabolism , Perilipin-1 , Phosphoproteins/analysis , Phosphoproteins/metabolism , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Messenger/metabolism , Rosiglitazone , Thiazolidinediones/pharmacology
13.
Biochem J ; 411(3): 647-55, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18215134

ABSTRACT

Multiple studies have suggested that the protein kinase Akt/PKB (protein kinase B) is required for insulin-stimulated glucose transport in skeletal muscle and adipose cells. In an attempt to understand links between Akt activation and glucose transport regulation, we applied mass spectrometry-based proteomics and bioinformatics approaches to identify potential Akt substrates containing the phospho-Akt substrate motif RXRXXpS/T. The present study describes the identification of the Rab GAP (GTPase-activating protein)-domain containing protein TBC1D1 [TBC (Tre-2/Bub2/Cdc16) domain family, member 1], which is closely related to TBC1D4 [TBC domain family, member 4, also denoted AS160 (Akt substrate of 160 kDa)], as an Akt substrate that is phosphorylated at Thr(590). RNAi (RNA interference)-mediated silencing of TBC1D1 elevated basal deoxyglucose uptake by approx. 61% in 3T3-L1 mouse embryo adipocytes, while the suppression of TBC1D4 and RapGAP220 under the same conditions had little effect on basal and insulin-stimulated deoxyglucose uptake. Silencing of TBC1D1 strongly increased expression of the GLUT1 glucose transporter but not GLUT4 in cultured adipocytes, whereas the decrease in TBC1D4 had no effect. Remarkably, loss of TBC1D1 in 3T3-L1 adipocytes activated the mTOR (mammalian target of rapamycin)-p70 S6 protein kinase pathway, and the increase in GLUT1 expression in the cells treated with TBC1D1 siRNA (small interfering RNA) was blocked by the mTOR inhibitor rapamycin. Furthermore, overexpression of the mutant TBC1D1-T590A, lacking the putative Akt/PKB phosphorylation site, inhibited insulin stimulation of p70 S6 kinase phosphorylation at Thr(389), a phosphorylation induced by mTOR. Taken together, our data suggest that TBC1D1 may be involved in controlling GLUT1 glucose transporter expression through the mTOR-p70 S6 kinase pathway.


Subject(s)
Gene Expression Regulation , Glucose Transporter Type 1/metabolism , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , 3T3-L1 Cells , Adipocytes , Animals , Antibodies/immunology , Cricetinae , GTPase-Activating Proteins/metabolism , Glucose Transporter Type 1/genetics , Insulin/pharmacology , Mice , Mutation/genetics , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphorylation/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/drug effects , Substrate Specificity , TOR Serine-Threonine Kinases
14.
J Biol Chem ; 282(47): 34213-8, 2007 Nov 23.
Article in English | MEDLINE | ID: mdl-17884815

ABSTRACT

Fat-specific protein (FSP)27/Cidec is most highly expressed in white and brown adipose tissues and increases in abundance by over 50-fold during adipogenesis. However, its function in adipocytes has remained elusive since its discovery over 15 years ago. Here we demonstrate that FSP27/Cidec localizes to lipid droplets in cultured adipocytes and functions to promote lipid accumulation. Ectopically expressed FSP27-GFP surrounds lipid droplets in 3T3-L1 adipocytes and colocalizes with the known lipid droplet protein perilipin. Immunostaining of endogenous FSP27 in 3T3-L1 adipocytes also confirmed its presence on lipid droplets. FSP27-GFP expression also markedly increases lipid droplet size and enhances accumulation of total neutral lipids in 3T3-L1 preadipocytes as well as other cell types such as COS cells. Conversely, RNA interference-based FSP27/Cidec depletion in mature adipocytes significantly stimulates lipolysis and reduces the size of lipid droplets. These data reveal FSP27/Cidec as a novel adipocyte lipid droplet protein that negatively regulates lipolysis and promotes triglyceride accumulation.


Subject(s)
Adipocytes/metabolism , Adipogenesis/physiology , Lipolysis/physiology , Phosphoproteins/metabolism , Proteins/metabolism , Triglycerides/metabolism , 3T3 Cells , Adipocytes/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , COS Cells , Carrier Proteins , Chlorocebus aethiops , Male , Mice , Perilipin-1 , Proteins/genetics , RNA Interference
15.
J Biol Chem ; 282(27): 19302-12, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17500068

ABSTRACT

Tumor necrosis factor alpha (TNFalpha) is a cytokine secreted by macrophages and adipocytes that contributes to the low grade inflammation and insulin resistance observed in obesity. TNFalpha signaling decreases peroxisome proliferator-activated receptor gamma and glucose transporter isoform 4 (GLUT4) expression in adipocytes, impairing insulin action, and this is mediated in part by the yeast Ste20 protein kinase ortholog Map4k4. Here we show that Map4k4 expression is selectively up-regulated by TNFalpha, whereas the expression of the protein kinases JNK1/2, ERK1/2, p38 stress-activated protein kinase, and mitogen-activated protein kinase kinases 4/7 shows little or no response. Furthermore, the cytokines interleukin 1beta (IL-1beta) and IL-6 as well as lipopolysaccharide fail to increase Map4k4 mRNA levels in cultured adipocytes under conditions where TNFalpha elicits a 3-fold effect. Using agonistic and antagonistic antibodies and small interfering RNA (siRNA) against TNFalpha receptor 1 (TNFR1) and TNFalpha receptor 2 (TNFR2), we show that TNFR1, but not TNFR2, mediates the increase in Map4k4 expression. TNFR1, but not TNFR2, also mediates a potent effect of TNFalpha on the phosphorylation of JNK1/2 and p38 stress-activated protein kinase and their downstream transcription factor substrates c-Jun and activating transcription factor 2 (ATF2). siRNA-based depletion of c-Jun and ATF2 attenuated TNFalpha action on Map4k4 mRNA expression. Consistent with this concept, the phosphorylation of ATF2 along with the expression and phosphorylation of c-Jun by TNFalpha signaling was more robust and prolonged compared with that of IL-1beta, which failed to modulate Map4k4. These data reveal that TNFalpha selectively stimulates the expression of a key component of its own signaling pathway, Map4k4, through a TNFR1-dependent mechanism that targets the transcription factors c-Jun and ATF2.


Subject(s)
Activating Transcription Factor 2/metabolism , Mitogen-Activated Protein Kinases/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins c-jun/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , 3T3 Cells , Adipocytes/metabolism , Animals , Glucose Transporter Type 4/metabolism , Inflammation/metabolism , Insulin Resistance , Interleukin-1beta/metabolism , Interleukin-1beta/pharmacology , Interleukin-6/metabolism , Interleukin-6/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , PPAR gamma/metabolism , Phosphorylation , Proto-Oncogene Proteins c-jun/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Receptors, Tumor Necrosis Factor, Type I/agonists , Receptors, Tumor Necrosis Factor, Type II/agonists , Receptors, Tumor Necrosis Factor, Type II/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , NF-kappaB-Inducing Kinase
16.
J Indian Med Assoc ; 103(12): 682-5, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16821665

ABSTRACT

For the global eradication of poliomyelitis to succeed it is necessary to address the problem of type 1 poliovirus infection in those last remaining regions and countries where type 1 disease remains endemic. By general consensus among experts, prevention and eradication of type 1 infection would not be achieved using the currently available trivalent oral poliovirus vaccine (tOPV). Monovalent OPV1 (mOPV1) will be necessary for the purpose. mOPV1 was developed by Panacea Biotec Ltd, a United Nations prequalified producer of tOPV of proven efficacy and safety, by removal from the formulation of the mOPV2 and mOPV3 components with no change whatever in the quality, chemistry and stability of the monovalent product, and the virus content of the mOPV1 vaccine unchanged from the original approved tOPV product. This article summarises the development and regulatory strategy for the World Health Organisation proposal for such development with scrupulous provision being made for the safety of the vaccinees, and for their seroprotective responses.


Subject(s)
Mass Vaccination , National Health Programs , Poliomyelitis/prevention & control , Poliovirus Vaccine, Oral/supply & distribution , Child , Child, Preschool , Endemic Diseases/prevention & control , Humans , India/epidemiology , Poliomyelitis/epidemiology , Poliovirus Vaccine, Oral/administration & dosage , World Health Organization
17.
Mol Cell Biol ; 24(12): 5447-58, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15169906

ABSTRACT

Glucose homeostasis is controlled in part by regulation of glucose uptake into muscle and adipose tissue. Intracellular membrane vesicles containing the GLUT4 glucose transporter move towards the cell cortex in response to insulin and then fuse with the plasma membrane. Here we show that the fusion step is retarded by the inhibition of phosphatidylinositol (PI) 3-kinase. Treatment of insulin-stimulated 3T3-L1 adipocytes with the PI 3-kinase inhibitor LY294002 causes the accumulation of GLUT4-containing vesicles just beneath the cell surface. This accumulation of GLUT4-containing vesicles near the plasma membrane prior to fusion requires an intact cytoskeletal network and the unconventional myosin motor Myo1c. Remarkably, enhanced Myo1c expression under these conditions causes extensive membrane ruffling and overrides the block in membrane fusion caused by LY294002, restoring the display of GLUT4 on the cell exterior. Ultrafast microscopic analysis revealed that insulin treatment leads to the mobilization of GLUT4-containing vesicles to these regions of Myo1c-induced membrane ruffles. Thus, localized membrane remodeling driven by the Myo1c motor appears to facilitate the fusion of exocytic GLUT4-containing vesicles with the adipocyte plasma membrane.


Subject(s)
Membrane Fusion/physiology , Muscle Proteins , Myosins/physiology , 3T3-L1 Cells , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Exocytosis , Glucose Transporter Type 4 , Insulin/pharmacology , Mice , Molecular Motor Proteins/genetics , Molecular Motor Proteins/physiology , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Morpholines/pharmacology , Myosin Type I , Myosins/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
18.
Mol Biol Cell ; 14(7): 2935-45, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12857876

ABSTRACT

Ca2+ is an essential requirement in membrane fusion, acting through binding proteins such as calmodulin (CaM). Ca2+/CaM is required for early endosome fusion in vitro, however, the molecular basis for this requirement is unknown. An additional requirement for endosome fusion is the protein Early Endosome Antigen 1 (EEA1), and its recruitment to the endosome depends on phosphatidylinositol 3-phosphate [PI(3)P] and the Rab5 GTPase. Herein, we demonstrate that inhibition of Ca2+/CaM, by using either chemical inhibitors or specific antibodies directed to CaM, results in a profound inhibition of EEA1 binding to endosomal membranes both in live cells and in vitro. The concentration of Ca2+/CaM inhibitors required for a full dissociation of EEA1 from endosomal membranes had no effect on the activity of phosphatidylinositol 3-kinases or on endogenous levels of PI(3)P. However, the interaction of EEA1 with liposomes containing PI(3)P was decreased by Ca2+/CaM inhibitors. Thus, Ca2+/CaM seems to be required for the stable interaction of EEA1 with endosomal PI(3)P, perhaps by directly or indirectly stabilizing the quaternary organization of the C-terminal FYVE domain of EEA1. This requirement is likely to underlie at least in part the essential role of Ca2+/CaM in endosome fusion.


Subject(s)
Calmodulin/metabolism , Membrane Fusion , Membrane Proteins/metabolism , Amino Acid Motifs , Animals , COS Cells , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/physiology , Chlorocebus aethiops , Endosomes/metabolism , Endosomes/physiology , Liposomes , Membrane Fusion/physiology , Membrane Proteins/physiology , Microscopy, Fluorescence , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Protein Structure, Quaternary/physiology , Recombinant Proteins/metabolism , Sulfonamides/pharmacology , Vesicular Transport Proteins , rab5 GTP-Binding Proteins/metabolism
19.
EMBO J ; 22(10): 2387-99, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12743033

ABSTRACT

Insulin stimulates glucose uptake in muscle and adipose cells by mobilizing intracellular membrane vesicles containing GLUT4 glucose transporter proteins to the plasma membrane. Here we show in live cultured adipocytes that intracellular membranes containing GLUT4-yellow fluorescent protein (YFP) move along tubulin-cyan fluorescent protein-labeled microtubules in response to insulin by a mechanism that is insensitive to the phosphatidylinositol 3 (PI3)-kinase inhibitor wortmannin. Insulin increased by several fold the observed frequencies, but not velocities, of long-range movements of GLUT4-YFP on microtubules, both away from and towards the perinuclear region. Genomics screens show conventional kinesin KIF5B is highly expressed in adipocytes and this kinesin is partially co-localized with perinuclear GLUT4. Dominant-negative mutants of conventional kinesin light chain blocked outward GLUT4 vesicle movements and translocation of exofacial Myc-tagged GLUT4-green fluorescent protein to the plasma membrane in response to insulin. These data reveal that insulin signaling targets the engagement or initiates the movement of GLUT4-containing membranes on microtubules via conventional kinesin through a PI3-kinase-independent mechanism. This insulin signaling pathway regulating KIF5B function appears to be required for GLUT4 translocation to the plasma membrane.


Subject(s)
Adipocytes/drug effects , Biological Transport/physiology , Insulin/pharmacology , Kinesins/metabolism , Microtubules/metabolism , Monosaccharide Transport Proteins/metabolism , Muscle Proteins , Adipocytes/cytology , Adipocytes/physiology , Androstadienes/pharmacology , Animals , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Glucose Transporter Type 4 , Insulin/physiology , Intracellular Membranes/metabolism , Kinesins/genetics , Luminescent Proteins/metabolism , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , Transport Vesicles/metabolism , Wortmannin
20.
J Cell Biol ; 158(7): 1239-49, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12356868

ABSTRACT

Transforming growth factor (TGF)beta is an important physiological regulator of cellular growth and differentiation. It activates a receptor threonine/serine kinase that phosphorylates the transcription factor Smad2, which then translocates into the nucleus to trigger specific transcriptional events. Here we show that activated type I and II TGF beta receptors internalize into endosomes containing the early endosomal protein EEA1. The extent of TGF beta-stimulated Smad2 phosphorylation, Smad2 nuclear translocation, and TGF beta-stimulated transcription correlated closely with the extent of internalization of the receptor. TGF beta signaling also requires SARA (Smad anchor for receptor activation), a 135-kD polypeptide that contains a FYVE Zn(++) finger motif. Here we show that SARA localizes to endosomes containing EEA1, and that disruption of this localization inhibits TGF beta-induced Smad2 nuclear translocation. These results indicate that traffic of the TGF beta receptor into the endosome enables TGF beta signaling, revealing a novel function for the endosome as a compartment specialized for the amplification of certain extracellular signals.


Subject(s)
DNA-Binding Proteins/physiology , Endosomes/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Serine Endopeptidases , Signal Transduction/physiology , Trans-Activators/physiology , Transforming Growth Factor beta/metabolism , Zinc Fingers , Animals , Binding Sites , COS Cells , Carrier Proteins/metabolism , Cricetinae , Dynamins/genetics , Dynamins/metabolism , Fluorescent Antibody Technique , Genes, Dominant/physiology , HeLa Cells , Humans , Phosphorylation , Potassium/metabolism , Protein Transport , Smad2 Protein , Subcellular Fractions , Transfection , Transferrin/metabolism , Tumor Cells, Cultured/cytology , Vesicular Transport Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...