Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Horm Metab Res ; 47(9): 681-5, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25502943

RESUMEN

Subclinical hypothyroidism, characterized by an isolated rise in TSH serum levels with normal thyroid function, is a pro-inflammatory state associated with insulin resistance. Adipocytes express TSH receptors, but it is not known if TSH can directly inhibit insulin signaling. Using primary human differentiated adipocytes, we examined the effects of TSH on insulin-stimulated Akt phosphorylation, and whether conventional PKC (cPKC) were involved. The effect of insulin on TSH-stimulated lipolysis was also investigated. TSH inhibited insulin-stimulated Akt phosphorylation in adipocytes by 54%. TSH activated cPKC, and Gö6976, a PKCα and -ß1 inhibitor, prevented the inhibitory effect of TSH on the insulin response. Insulin reduced the ability of TSH to activate cPKC and to stimulate lipolysis.Our data reveal novel interactions between TSH and insulin. TSH inhibits insulin-stimulated Akt signaling in a cPKC-dependent fashion, whereas insulin blocks TSH-stimulated cPKC activity and lipolysis. TSH and insulin act on differentiated human adipocytes to modulate their respective intracellular signals.


Asunto(s)
Adipocitos/metabolismo , Insulina/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal/fisiología , Tirotropina/metabolismo , Adulto , Diferenciación Celular , Células Cultivadas , Femenino , Humanos , Lipólisis , Masculino , Persona de Mediana Edad
2.
Atherosclerosis ; 233(1): 123-9, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24529132

RESUMEN

OBJECTIVES: Serum low density lipoprotein-cholesterol (LDL-C) correlates positively with serum PCSK9 in the general population, consistent with PCSK9 being a determinant of LDL-C levels. Patients with chronic kidney disease (CKD) on hemodialysis (HD) have lower total cholesterol (TC) and LDL-C compared to the general population. Serum PCSK9 and its relationship with serum lipids have not been reported in CKD patients on HD (CKD-HD). METHODS: We measured serum PCSK9 by ELISA and lipid levels in 66 CKD-HD patients and compared them to 178 non-CKD subjects. Since statins increase serum PCSK9 levels, CKD-HD patients were separated into those not on statin therapy (HD-NS, n = 32) and those taking statins (HD-S, n = 34). No control subjects were on statin therapy. RESULTS: Serum PCSK9, TC, LDL-C and HDL-C levels were significantly lower in the CKD-HD group (n = 66) compared to the control group. HD-NS patients showed lower PCSK9, TC and LDL-C levels than control subjects and PCSK9 levels correlated with TC and LDL-C levels (r = 0.35, p = 0.050; r = 0.423, p = 0.0158 respectively) as well as TG levels (r = 0.413, p = 0.0188). In HD-S patients, PCSK9 levels were not significantly different from the non-CKD group. There was no correlation between PCSK9 levels and TC and LDL-C levels in the HD-S group. CONCLUSION: Our data are the first quantitative analysis of serum PCSK9 levels in CKD-HD patients. We show that serum PCSK9 in HD-NS patients is decreased and it retains a positive correlation with LDL-C, suggesting that PCSK9 may remain a significant determinant of LDL-C in CKD-HD subjects. We also show that statin therapy disrupts the correlation between LDL-C and PCSK9 in CKD-HD patients. These data suggest that the regulation of LDL-C by PCSK9 remains intact in CKD-HD patients. PCSK9 may also play a role in the metabolism of triglyceride-rich lipoproteins in CKD-HD patients.


Asunto(s)
LDL-Colesterol/sangre , Proproteína Convertasas/sangre , Insuficiencia Renal Crónica/enzimología , Serina Endopeptidasas/sangre , HDL-Colesterol/sangre , Femenino , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Masculino , Persona de Mediana Edad , Proproteína Convertasa 9 , Diálisis Renal , Insuficiencia Renal Crónica/tratamiento farmacológico
3.
Horm Metab Res ; 44(11): 825-31, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22730012

RESUMEN

Adipocytes express TSH receptors, and TSH can stimulate cAMP-dependent protein kinase, perilipin phosphorylation, and lipolysis in human and mouse 3T3-L1 adipocytes. TSH activates PKC in thyrocytes. Since PKC has been implicated in lipolysis in adipocytes, we examined whether the family of conventional isoforms of PKC (cPKC) is a target of TSH in adipocytes, and whether cPKC is required for TSH-stimulated lipolysis. Differentiated 3T3-L1 and subcutaneous abdominal human adipocytes in culture were treated with TSH in the presence or absence of either PKC inhibitor Gö6976 (inhibits PKCα, ßI) or Gö6983 (inhibits PKCα, ßI, ßII, γ, δ). Activation of cPKC was assessed by phospho-(ser) PKC substrate antibody immunoblot analysis. Perilipin phosphorylation was measured by SDS-PAGE electromobility shift followed by anti-perilipin immunoblot analysis. Lipolysis was quantified by the amount of nonesterified fatty acids (NEFAs) released into the medium. TSH strongly and significantly activated cPKC in differentiated human and 3T3-L1 adipocytes from undetectable levels in control conditions. This cPKC stimulation in human adipocytes by TSH was reduced significantly by 40% or 48% in the presence of PKC inhibitor Gö6983 or Gö6976, respectively. Gö6976 inhibited TSH-stimulated human adipocyte perilipin phosphorylation and NEFA release by 80% and 50%, respectively. We conclude that cPKC is activated by TSH in human differentiated adipocytes. Based on the effects of cPKC inhibition, cPKC activation is required for TSH-stimulated perilipin phosphorylation and lipolysis in human differentiated adipocytes.


Asunto(s)
Proteínas Portadoras/metabolismo , Lipólisis , Fosfoproteínas/metabolismo , Proteína Quinasa C/metabolismo , Tirotropina/metabolismo , Adipocitos/enzimología , Adipocitos/metabolismo , Adulto , Animales , Proteínas Portadoras/genética , Diferenciación Celular , Células Cultivadas , Activación Enzimática , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Perilipina-1 , Fosfoproteínas/genética , Fosforilación , Proteína Quinasa C/genética
4.
Horm Metab Res ; 42(12): 831-6, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20803413

RESUMEN

Macrophage-secreted factors inhibit adipogenesis, but the underlying mechanism is not well understood. Our objective was to determine if anti-adipogenic signaling pathways in human preadipocytes are activated by macrophage-conditioned medium (MacCM). Human abdominal subcutaneous stromal preadipocytes were treated with adipogenic inducers in either standard medium or medium conditioned by human THP-1 macrophages. THP-1-MacCM increased inhibitor of κB kinase ß (IKKß) phosphorylation, inhibitor of NF-κB α (IκBα) degradation, and NF-κB activity in human preadipocytes in a time-dependent manner. Concomitant treatment of human abdominal subcutaneous preadipocytes with sc-514, a selective inhibitor of IKKß, prevented the inhibitory effect of THP-1-MacCM on lipid accumulation and expression of adipogenic markers. Our data indicate that activation of the preadipocyte IKKß/NF-κB pathway is required for the anti-adipogenic effect of THP-1-MacCM on human adipogenesis.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Medios de Cultivo Condicionados/metabolismo , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Macrófagos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Adulto , Línea Celular , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Femenino , Humanos , Quinasa I-kappa B/genética , Proteínas I-kappa B/genética , Persona de Mediana Edad , Inhibidor NF-kappaB alfa , FN-kappa B/genética , Fosforilación , Transducción de Señal/efectos de los fármacos
5.
Mini Rev Med Chem ; 8(1): 91-6, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18220988

RESUMEN

Thyroid stimulating hormone (TSH; also known as thyrotropin), binds cognate receptors on the surface of thyrocytes to regulate proliferation and thyroid hormone synthesis. This unidimensional view of TSH is being transformed as new evidence indicates that TSH acts on adipose tissue. Adipocyte inflammatory responses that predispose to cardiovascular disease may occur in thyroid disorders associated with elevated TSH levels.


Asunto(s)
Adipocitos/metabolismo , Receptores de Tirotropina/metabolismo , Tirotropina/fisiología , Adipocitos/efectos de los fármacos , Animales , Enfermedades Cardiovasculares/etiología , Humanos , Hipotiroidismo/complicaciones , Hipotiroidismo/metabolismo , Inflamación/metabolismo , Interleucina-6/metabolismo , Receptores de Tirotropina/efectos de los fármacos , Factores de Riesgo , Tirotropina/farmacología
6.
Horm Metab Res ; 39(8): 575-81, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17712722

RESUMEN

Differentiation of preadipocytes into functional adipocytes depends on early proliferative events (mitotic clonal expansion) and extracellular matrix interactions. We report that discoidin domain receptor (DDR) 2, a novel adhesion receptor, is expressed in 3T3-L1 preadipocytes and is downregulated during the early phase of adipogenesis. DDR2 overexpression (DDR2-L1 preadipocytes) reduced subconfluent proliferation by 56% (p<0.001) and insulin-stimulated tyrosine phosphorylation of insulin receptor substrate (IRS)-1 by 34% (p<0.05). The mitotic clonal expansion phase of differentiating confluent DDR2-L1 preadipocytes was impaired by approximately 25% (p<0.05). Although induction of peroxisome proliferator-activated receptor gamma, fatty acid synthase, and adiponectin was not altered, the resulting adipocytes were 55% larger (p<0.05), and contained 66% more triacylglycerol (p<0.01). The induction of CCAAT/enhancer binding protein alpha was reduced by 37% (p<0.05), correlating with a similar reduction in insulin-stimulated IRS-1 tyrosine phosphorylation and glucose transport in DDR2-L1 adipocytes (decreases of 22% and 27%, respectively; p<0.05 for both). Our data show that DDR2 is expressed in adipose cells and that its overexpression leads to insulin resistance.


Asunto(s)
Glucosa/metabolismo , Insulina/farmacología , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores Mitogénicos/fisiología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Animales , Diferenciación Celular , Receptores con Dominio Discoidina , Proteínas Sustrato del Receptor de Insulina , Resistencia a la Insulina/genética , Ratones , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores Mitogénicos/genética , Receptores Mitogénicos/metabolismo , Transfección
7.
J Cell Physiol ; 211(3): 598-607, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17219406

RESUMEN

In 3T3-L1 and human preadipocytes, insulin results in the isolated rise in phosphatidylinositol (PI)-3,4,5-P3, whereas PDGF produces PI(3,4)P2 in addition to PI(3,4,5)P3. SH2 domain-containing inositol 5-phosphatase 2 (SHIP2) converts PI(3,4,5)P3 into PI(3,4)P2. PDGF, but not insulin, stimulates SHIP2 tyrosine phosphorylation and its association with Shc in human and 3T3-L1 preadipocytes. We now demonstrate that SHIP2 tyrosine phosphorylation and association with Shc in PDGF-treated 3T3-L1 preadipocytes was reduced by bisindolylmaleimide I (BisI), an inhibitor of conventional/novel protein kinase C (PKC). However, the production of PI(3,4)P2 and PI(3,4,5)P3 by PDGF was unaffected by BisI. Activation of PKC by 12-O-tetradecanoylphorbol-13-acetate (TPA) was not sufficient to induce SHIP2 tyrosine phosphorylation. Furthermore, we identified threonine 958 (T958) as a novel PDGF-responsive SHIP2 phosphorylation site. Mutation of T958 to alanine reduced PDGF-stimulated SHIP2 tyrosine phosphorylation and association with Shc, but did not alter its anti-proliferative effect on preadipocytes. This study demonstrates that SHIP2 tyrosine phosphorylation and Shc association can be regulated by serine/threonine signaling pathways, either indirectly (via PKC), or directly (via T958). Interestingly, the anti-proliferative effect of SHIP2 T958A, as well as another SHIP2 mutant (Y986F, Y987F) that also displays defective tyrosine phosphorylation and Shc association, does not depend on these molecular events.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adipocitos/enzimología , Monoéster Fosfórico Hidrolasas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Células 3T3-L1 , Adipocitos/citología , Sustitución de Aminoácidos , Animales , División Celular/fisiología , Interacciones Farmacológicas , Inhibidores Enzimáticos/farmacología , Humanos , Indoles/farmacología , Inositol Polifosfato 5-Fosfatasas , Maleimidas/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Monoéster Fosfórico Hidrolasas/genética , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteínas Adaptadoras de la Señalización Shc , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src , Transfección , Tirosina/metabolismo
8.
Am J Physiol Endocrinol Metab ; 290(6): E1140-4, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16682487

RESUMEN

Adipose cells are extrathyroidal targets of thyroid-stimulating hormone (TSH). TSH stimulates interleukin-6 (IL-6) release from adipocytes. We examined TSH responsiveness as a function of stage of differentiation or adipose tissue depot in cultured adipose cells and determined the effect of TSH on extrathyroidal IL-6 production in vivo. Stromal preadipocytes, isolated from human abdominal subcutaneous or omental adipose tissue, and their differentiated counterparts were studied. IL-6 protein concentration in the medium was measured after TSH stimulation. Basal IL-6 release was greater for preadipocytes than differentiated adipocytes, whether derived from subcutaneous or omental fat depots. A depot-dependent effect (omental > subcutaneous) on basal IL-6 release was observed for preadipocytes (1.6-fold, P < 0.05); a similar trend for differentiated adipocytes was not significant (6.2-fold, P > 0.05). IL-6 responsiveness to TSH was observed upon differentiation, but only for subcutaneous adipocytes (1.9-fold over basal, P < 0.001). To determine if TSH could stimulate IL-6 release from extrathyroidal tissues in vivo, we measured serum IL-6 levels from five thyroidectomized patients who received recombinant human (rh) TSH and found that levels increased by threefold on days 3 and 4 (P < 0.05) after its administration. Our data demonstrate that stage of differentiation and fat depot origin affect basal and TSH-stimulated IL-6 release from adipose cells in culture. Furthermore, rhTSH elevates serum IL-6 response in thyroidectomized patients, indicating an extrathyroidal site of TSH action.


Asunto(s)
Abdomen/fisiología , Tejido Adiposo/metabolismo , Diferenciación Celular , Interleucina-6/metabolismo , Tirotropina/farmacología , Adipocitos/metabolismo , Medios de Cultivo Condicionados , Femenino , Humanos , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Epiplón/citología , Epiplón/metabolismo , Grasa Subcutánea/citología , Grasa Subcutánea/metabolismo , Tiroidectomía
9.
Diabetologia ; 49(6): 1402-11, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16609875

RESUMEN

AIMS/HYPOTHESIS: In obesity, a limited adipogenic capacity may promote adipocyte hypertrophy and increase the risk of insulin resistance and type 2 diabetes. Recent data indicate that macrophages reside within adipose tissue in obese rodents and humans. We hypothesised that secreted macrophage factors may inhibit adipogenesis. MATERIALS AND METHODS: Conditioned media from cultured murine J774 or human THP-1 macrophages were collected, and added to either murine 3T3-L1 preadipocytes or human abdominal stromal preadipocytes from subcutaneous or omental fat depots. RESULTS: Macrophage-conditioned medium (MacCM) strongly inhibited 3T3-L1 adipogenesis. Dose-response studies with J774-MacCM revealed that 80 and 100% of J774-MacCM completely suppressed triacylglycerol accumulation as well as the induction of fatty acid synthase, peroxisome proliferator-activated receptor gamma, CCAAT/enhancer binding protein alpha, and adiponectin. Similar inhibitory effects on 3T3-L1 preadipocytes were observed with THP-1-MacCM. Differentiation of human abdominal subcutaneous stromal preadipocytes was moderately reduced (subcutaneous>omental) by J744-MacCM. In contrast, the differentiation of both subcutaneous and omental stromal preadipocytes was completely inhibited by THP-1-MacCM, as determined on the basis of morphology and triacylglycerol accumulation, as well as fatty acid synthase and adiponectin protein expression. CONCLUSIONS/INTERPRETATION: Secreted macrophage products inhibit the differentiation of 3T3-L1 preadipocytes as well as human abdominal stromal preadipocytes.


Asunto(s)
Células 3T3/citología , Adipocitos/citología , Diferenciación Celular/fisiología , Macrófagos/fisiología , Abdomen , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular , Medios de Cultivo Condicionados , Humanos , Ratones , Monocitos/citología
10.
Int J Obes (Lond) ; 29(8): 1006-9, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15852047

RESUMEN

OBJECTIVE: Phosphoinositide 3-kinase (PI3K) is required for murine adipocyte differentiation. However, a recent report concluded that PI3K was not involved in the differentiation of human preadipocytes into adipocytes. We have re-examined the role of PI3K in human preadipocyte differentiation, enrolling more patients and using more adipogenic indices. METHODS: Human preadipocytes, isolated from nine patients, were induced to differentiate in the presence or absence of 100 nM wortmannin. After 12-15 days, triacylglycerol accumulation and the expression of adipogenic markers (fatty acid synthase and adiponectin) were measured. RESULTS: A significant inhibition in triacylglycerol accumulation and in the induction of fatty acid synthase protein expression was observed, but there was no effect on adiponectin protein expression. CONCLUSION: Inhibition of PI3K reduces the differentiation of human preadipocytes into adipocytes, suggesting a role for this enzyme in the human adipogenic process.


Asunto(s)
Adipocitos/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Adipocitos/enzimología , Análisis de Varianza , Androstadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Ácido Graso Sintasas/metabolismo , Femenino , Humanos , Immunoblotting , Masculino , Persona de Mediana Edad , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Triglicéridos/metabolismo , Wortmanina
11.
J Cell Physiol ; 204(2): 646-53, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15754337

RESUMEN

Healthy adipose tissue function depends on adipogenesis. The capacity to form new adipocytes prevents the emergence of insulin-resistant hypertrophied adipocytes, as well as the deleterious lipid deposition in muscle, liver, and pancreas. It is therefore important to understand how adipogenesis is modulated. Platelet-derived growth factor (PDGF) is anti-adipogenic, but the stage of differentiation that it targets, and the signaling pathways that it triggers, are not defined. We have studied the inhibitory effect of PDGF on murine 3T3-L1 preadipocyte and human preadipocyte differentiation. There was a significant attenuation in the protein expression of the adipogenic transcription factors, PPARgamma and C/EBPalpha, as well as in the levels of later differentiation markers, including adiponectin, aP2, and fatty acid synthase. PDGF treatment resulted in the persistence of PDGF receptor and PKCalpha expression, in contrast to the expected downregulation of both proteins that occurs during differentiation. Inactivation of conventional PKC isoforms, by bisindolylmaleimide I or PKC pseudosubstrate M20-28, partially reversed the inhibition of 3T3-L1 and human preadipocyte differentiation by PDGF, as assessed by fatty acid synthase expression and morphological appearance.


Asunto(s)
Adipocitos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Células Madre/efectos de los fármacos , Células 3T3 , Adipocitos/citología , Animales , Biomarcadores/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Femenino , Humanos , Indoles/farmacología , Masculino , Maleimidas/farmacología , Ratones , Persona de Mediana Edad , PPAR gamma/metabolismo , Proteína Quinasa C/metabolismo , Proteína Quinasa C-alfa , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/citología
13.
Int J Obes Relat Metab Disord ; 28(2): 191-8, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14970836

RESUMEN

OBJECTIVE: Insulin-responsive adipogenic signaling molecules include insulin receptor substrates (IRS)-1 and -2, phosphoinositide 3-kinase (PI3K), and protein kinase B (PKB; also known as Akt). Mammalian target of rapamycin (mTOR) is a PKB substrate, and regulates p70 S6 kinase (p70 S6K). Since p70 S6K is an insulin-responsive kinase downstream of PI3K and PKB, its potential role in adipogenic insulin signaling was investigated. DESIGN: We measured the effect of rapamycin, a specific inhibitor of mTOR, on insulin-induced 3T3-L1 adipogenesis and on insulin-stimulated p70 S6K activation. RESULTS: Rapamycin partially reduced differentiation, measured by Oil Red O staining, triacylglycerol accumulation (by up to 46%), and peroxisome proliferator-activated receptor gamma protein expression (by 50%). In contrast, rapamycin completely inhibited insulin-stimulated p70 S6K activation, assessed by phosphorylation of p70 S6K and its substrate, S6. Expression of a constitutively activated form of p70 S6K did not promote 3T3-L1 adipogenesis. The considerable residual differentiation in the presence of rapamycin, despite the complete blockade of p70 S6K activation, prompted us to measure the phosphorylation of another rapamycin-sensitive protein, eukaryotic initiation factor 4E (eIF4E) binding protein 1 (4E-BP1). Insulin-stimulated 4E-BP1 phosphorylation in 3T3-L1 preadipocytes was only partially affected by rapamycin, consistent with the differentiation data. Phosphorylation of eIF4E itself, an expected consequence of 4E-BP1 phosphorylation, was also only partially inhibited. CONCLUSION: Our data suggest that adipogenic mTOR signaling occurs via the 4E-BP1/eIF4E pathway, rather than through p70 S6K.


Asunto(s)
Adipocitos/efectos de los fármacos , Proteínas Portadoras/metabolismo , Antagonistas de Insulina/farmacología , Insulina/farmacología , Fosfoproteínas/metabolismo , Proteínas Quinasas , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteínas de Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Factores Eucarióticos de Iniciación , Ratones , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR
15.
J Cell Physiol ; 189(1): 14-22, 2001 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11573200

RESUMEN

Inhibition of insulin-induced 3T3-L1 preadipocyte differentiation by rapamycin has been attributed to a blockade of the early critical clonal expansion phase of the adipogenic program. Rapamycin binds to, and inhibits, mTOR (mammalian target of rapamycin), leading to diminution of p70 S6 kinase activity and eukaryotic initiation factor 4E binding protein 1 (eIF4E-BP1) function. Our objective was to determine if rapamycin-sensitive pathways exist subsequent to the clonal expansion phase. We determined that the mitotic clonal expansion was complete by day 4 of the differentiation protocol, based on the response to Ara-C (cytosine beta-D-arabinofuranoside), which only inhibits differentiation when administered during this phase. Treatment of differentiating 3T3-L1 cells with rapamycin, starting on day 4, exerted potent negative effects on glycerol phosphate dehydrogenase activity, and triacylglycerol accumulation, as well as on the protein expression of adipogenic transcription factors, C/EBPalpha and PPARgamma. Insulin-stimulated p70 S6 kinase activity, and its inhibition by rapamycin, were comparable in preadipocytes at day 0 vs. day 4 post-differentiation. We conclude that a component of the adipogenic program, operating after the completion of clonal expansion, is inhibited by rapamycin, suggesting an ongoing need for mTOR function in this process.


Asunto(s)
Adipocitos/fisiología , Proteínas Quinasas , Sirolimus/farmacología , Células 3T3 , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular , División Celular , Citarabina/farmacología , Glicerolfosfato Deshidrogenasa/antagonistas & inhibidores , Insulina/farmacología , Cinética , Ratones , Inhibidores de Proteínas Quinasas , Receptores Citoplasmáticos y Nucleares/metabolismo , Proteínas Quinasas S6 Ribosómicas/antagonistas & inhibidores , Células Madre/citología , Células Madre/metabolismo , Serina-Treonina Quinasas TOR , Factores de Transcripción/metabolismo , Triglicéridos/biosíntesis
16.
Endocrinology ; 142(1): 205-12, 2001 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11145583

RESUMEN

Adipocyte number, a determinant of adipose tissue mass, reflects the balance between the rates of proliferation/differentiation vs. apoptosis of preadipocytes. The percentage of 3T3-L1 preadipocytes undergoing cell death following serum deprivation was reduced by 10 nM insulin-like growth factor (IGF)-1 (from 50.0 +/- 0.7% for control starved cells to 27.5 +/- 3.1%). TUNEL staining confirmed the apoptotic nature of the cell death. The protective effect of IGF-1 was blocked by phosphoinositide 3-kinase (PI3K) inhibitors, wortmannin, and LY294002, but was unaffected by rapamycin, PD98059, or SB203580, which inhibit mammalian target of rapamycin (mTOR), ERK kinase (MEK1), and p38 MAPK respectively. Exogenous PI(3,4,5)P3 (10 microM), the principal product of IGF-1-stimulated PI3K in 3T3-L1 preadipocytes, had a modest survival effect on its own, reducing cell death from 47.9 +/- 3.4% to 35.6 +/- 3.5%. When added to the combination of IGF-1 and LY294002, PI(3,4,5)P3 reversed most of the inhibitory effect of LY294002 on IGF-1-dependent cell survival, protein kinase B/Akt phosphorylation, and caspase-3 activity. Taken together, these results implicate PI(3,4,5)P3 as a necessary signal for the anti-apoptotic action of IGF-1 on 3T3-L1 preadipocytes.


Asunto(s)
Adipocitos/citología , Factor I del Crecimiento Similar a la Insulina/farmacología , Fosfatos de Fosfatidilinositol/metabolismo , Células 3T3 , Adipocitos/efectos de los fármacos , Androstadienos/farmacología , Animales , Apoptosis/efectos de los fármacos , Cromonas/farmacología , Medio de Cultivo Libre de Suero , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Imidazoles/farmacología , Etiquetado Corte-Fin in Situ , MAP Quinasa Quinasa 1 , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Morfolinas/farmacología , Fosfatos de Fosfatidilinositol/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Piridinas/farmacología , Sirolimus/farmacología , Wortmanina , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Int J Obes Relat Metab Disord ; 24 Suppl 4: S3-7, 2000 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11126237

RESUMEN

Apoptosis is critical for mammalian tissue homeostasis, and its disruption has been linked to a wide variety of disorders, including cancer, neurodegenerative disease, autoimmune disease and diabetes. This review will focus on recent investigations that have begun to address the potential role of apoptosis in adipose tissue growth. Evidence for apoptosis occurring in mature adipocytes has been obtained through the use of in vitro cell culture models as well as in vivo studies in rodents and humans. Preadipocytes, fibroblast-like adipocyte precursor cells, can also undergo apoptotic cell death. As they differentiate, preadipocytes acquire a relative resistance to apoptosis. The levels of the cell survival proteins Bcl-2 and neuronal apoptosis inhibitory protein (NAIP) have been observed to increase during adipogenesis. Further research on the effect of apoptosis on adipose tissue cellularity should clarify its influence on adipose tissue mass and distribution.


Asunto(s)
Adipocitos/citología , Tejido Adiposo/crecimiento & desarrollo , Apoptosis , Diferenciación Celular , Metabolismo Energético , Animales , Humanos
18.
Am J Physiol Cell Physiol ; 279(2): C335-40, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10912999

RESUMEN

Controversy continues about whether, and to what levels of abundance, thyroid-stimulating hormone receptors (TSHR) are found in human tissues other than the thyroid gland. Restricted expression to the thyroid and orbit would suggest that TSHR represents the target autoantigen in thyroid-associated ophthalmopathy. A more generalized pattern of tissue expression would be inconsistent with TSHR acting as the autoantigen that is solely responsible for selectively targeting the immune system to the orbit. We have detected TSHR mRNA in human abdominal adipose tissue by Northern blot analysis. TSHR protein was also detected, by immunoblotting with two different antibodies, in preadipocytes isolated from human abdominal subcutaneous and omental adipose tissue and in derivative adipocytes differentiated in primary culture. Preadipocytes treated with thyroid-stimulating hormone (TSH) exhibited a sevenfold increase in the activity of p70 S6 kinase, a serine/threonine kinase recently recognized as a downstream target of TSHR in thyroid cells. Activation of p70 S6 kinase by TSH was also observed in orbital fibroblasts. Thus TSHR protein expression is found in fibroblasts from several anatomic locations, suggesting that factors other than site-limited TSHR expression must be involved in restricting the distribution of Graves' disease manifestations. Furthermore, the presence of functional TSHR in preadipocytes raises the possibility of a novel role for TSHR signaling in adipose tissue development.


Asunto(s)
Adipocitos/metabolismo , Fibroblastos/metabolismo , Receptores de Tirotropina/metabolismo , Proteínas Quinasas S6 Ribosómicas/metabolismo , Abdomen , Músculos Abdominales , Adipocitos/efectos de los fármacos , Animales , Células CHO , Células Cultivadas , Cricetinae , Fibroblastos/efectos de los fármacos , Humanos , Células Jurkat , Ratones , Epiplón , Ratas , Receptores de Tirotropina/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas/efectos de los fármacos , Glándula Tiroides/efectos de los fármacos , Glándula Tiroides/metabolismo , Tirotropina/farmacología
19.
Obes Res ; 8(3): 249-54, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10832768

RESUMEN

OBJECTIVE: The immunosuppressant drug rapamycin, has been reported to inhibit 3T3-L1 adipocyte differentiation by interfering with critical postconfluent mitoses that are required early on for successful differentiation of this cell line (clonal expansion phase). In contrast to the murine 3T3-L1 preadipocyte cell line, human preadipocytes in primary culture do not undergo clonal expansion during differentiation. We investigated whether rapamycin could inhibit human adipocyte differentiation. RESEARCH METHODS AND PROCEDURES: The effect of rapamycin on the induction of differentiation of human preadipocytes in primary culture into adipocytes was measured using Oil Red O staining and glycerol phosphate dehydrogenase activity. RESULTS: We have observed that rapamycin severely curtails human adipocyte differentiation of both omental and abdominal subcutaneous preadipocytes (to 14% and 19% of standard differentiation, respectively). The rapamycin-mediated inhibition of human adipocyte differentiation could be reversed in the presence of excess amounts of FK-506, which displaces rapamycin from its intracellular receptor, FKPB12. Measurement of cytosolic protein and [3H]thymidine incorporation into DNA confirmed the absence of proliferation during differentiation of human preadipocytes in primary culture. DISCUSSION: Our data indicate that rapamycin exerts important negative regulatory effects on adipogenesis in human preadipocytes, through a mechanism that does not depend on interruption of clonal expansion.


Asunto(s)
Adipocitos/efectos de los fármacos , Tejido Adiposo/citología , Diferenciación Celular/efectos de los fármacos , Inmunosupresores/farmacología , Sirolimus/farmacología , Adipocitos/citología , Compuestos Azo/química , Técnicas de Cultivo de Célula , Diferenciación Celular/fisiología , Células Cultivadas , Colorantes/química , Femenino , Glicerolfosfato Deshidrogenasa/análisis , Humanos , Masculino , Persona de Mediana Edad , Conteo por Cintilación , Timidina/química
20.
AIDS ; 14(16): 2467-73, 2000 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-11101056

RESUMEN

OBJECTIVE: A novel lipodystrophy syndrome characterized by truncal adiposity, peripheral fat atrophy, type 2 diabetes mellitus, and dyslipidemia occurs in HIV-infected individuals, and may be aggravated by HIV-1 protease inhibitors. The increase in truncal fat could be due to enhanced preadipocyte differentiation. Using the 3T3-L1 preadipocyte model, we reported that ritonavir enhances adipocyte differentiation in culture. The goal of this study was to characterize the molecular mechanism of ritonavir on preadipocyte differentiation. DESIGNS AND METHODS: Time course studies of 3T3-L1 preadipocytes placed in standard differentiation medium (insulin, dexamethasone, and isobutylmethylxanthine) were performed. Glycerol phosphate dehydrogenase (GPDH) was assayed enzymatically, and triacylglycerol (TG) mass was quantified. The adipogenic transcription factors adipocyte determination and differentiation-dependent factor 1 (ADD-1)/sterol regulatory element binding protein 1 (SREBP-1), CCAAT/enhancer-binding protein-alpha (CEBPalpha), and peroxisome proliferator activated receptor-gamma (PPARgamma), were measured by Western analysis. RESULTS: Ritonavir (10 microg/ml) enhanced 3T3-L1 preadipocyte differentiation (30% increase in TG mass; 50% increase in GPDH activity), and transiently raised levels of the 68 kDa active mature form of ADD-1/SREBP-1 during adipogenesis by threefold, compared with standard differentiation. In contrast, ritonavir attenuated the differentiation-induced increase in CEBPalpha and PPARgamma. CONCLUSIONS: Our data suggest that ritonavir enhances 3T3-L1 adipogenesis by increasing the level of active mature ADD-1/SREBP-1. This effect may be due to reduced proteolysis of ADD-1/SREBP-1, as ritonavir inhibits an N-acetyl-leucyl-leucyl-norleucinal (ALLN)-sensitive proteosomal degradation pathway in lymphocytes, and ALLN itself inhibits the breakdown of mature ADD-1/SREBP-1. As mature ADD-1/SREBP-1 regulates several lipogenic enzymes, higher levels may explain the effect of ritonavir on TG accumulation and GPDH activity. Studying ADD-1/SREBP-1 may lead to better understanding and prevention of the lipodystrophy syndrome.


Asunto(s)
Adipocitos/citología , Adipocitos/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proteínas de Unión al ADN/metabolismo , Inhibidores de la Proteasa del VIH/farmacología , Ritonavir/farmacología , Factores de Transcripción , Animales , Proteínas Potenciadoras de Unión a CCAAT/efectos de los fármacos , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Glicerolfosfato Deshidrogenasa/metabolismo , Ratones , Proteína 1 de Unión a los Elementos Reguladores de Esteroles , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...