Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Diabetologia ; 62(1): 123-135, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30327824

RESUMEN

AIMS/HYPOTHESIS: Pleiotrophin, a developmentally regulated and highly conserved cytokine, exerts different functions including regulation of cell growth and survival. Here, we hypothesise that this cytokine can play a regulatory role in glucose and lipid homeostasis. METHODS: To test this hypothesis, we performed a longitudinal study characterising the metabolic profile (circulating variables and tissue mRNA expression) of gene-targeted Ptn-deficient female mice and their corresponding wild-type counterparts at different ages from young adulthood (3 months) to older age (15 months). Metabolic cages were used to investigate the respiratory exchange ratio and energy expenditure, at both 24°C and 30°C. Undifferentiated immortalised mouse brown adipocytes (mBAs) were treated with 0.1 µg/ml pleiotrophin until day 6 of differentiation, and markers of mBA differentiation were analysed by quantitative real-time PCR (qPCR). RESULTS: Ptn deletion was associated with a reduction in total body fat (20.2% in Ptn+/+ vs 13.9% in Ptn-/- mice) and an enhanced lipolytic response to isoprenaline in isolated adipocytes from 15-month-old mice (189% in Ptn+/+ vs 273% in Ptn-/- mice). We found that Ptn-/- mice exhibited a significantly lower QUICKI value and an altered lipid profile; plasma triacylglycerols and NEFA did not increase with age, as happens in Ptn+/+ mice. Furthermore, the contribution of cold-induced thermogenesis to energy expenditure was greater in Ptn-/- than Ptn+/+ mice (42.6% and 33.6%, respectively). Body temperature and the activity and expression of deiodinase, T3 and mitochondrial uncoupling protein-1 in the brown adipose tissue of Ptn-/- mice were higher than in wild-type controls. Finally, supplementing brown pre-adipocytes with pleiotrophin decreased the expression of the brown adipocyte markers Cidea (20% reduction), Prdm16 (21% reduction), and Pgc1-α (also known as Ppargc1a, 11% reduction). CONCLUSIONS/INTERPRETATION: Our results reveal for the first time that pleiotrophin is a key player in preserving insulin sensitivity, driving the dynamics of adipose tissue lipid turnover and plasticity, and regulating energy metabolism and thermogenesis. These findings open therapeutic avenues for the treatment of metabolic disorders by targeting pleiotrophin in the crosstalk between white and brown adipose tissue.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Metabolismo Energético/fisiología , Termogénesis/fisiología , Animales , Proteínas Portadoras/genética , Citocinas/genética , Metabolismo Energético/genética , Femenino , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Estudios Longitudinales , Ratones , Ratones Noqueados , Termogénesis/genética
2.
Cereb Cortex ; 28(5): 1783-1793, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28407057

RESUMEN

Thyroid hormones (THs, T4 and the transcriptionally active hormone T3) play an essential role in neurodevelopment; however, the mechanisms underlying T3 brain delivery during mice fetal development are not well known. This work has explored the sources of brain T3 during mice fetal development using biochemical, anatomical, and molecular approaches. The findings revealed that during late gestation, a large amount of fetal brain T4 is of maternal origin. Also, in the developing mouse brain, fetal T3 content is regulated through the conversion of T4 into T3 by type-2 deiodinase (D2) activity, which is present from earlier prenatal stages. Additionally, D2 activity was found to be essential to mediate expression of T3-dependent genes in the cerebral cortex, and also necessary to generate the transient cerebral cortex hyperthyroidism present in mice lacking the TH transporter Monocarboxylate transporter 8. Notably, the gene encoding for D2 (Dio2) was mainly expressed at the blood-cerebrospinal fluid barrier (BCSFB). Overall, these data signify that T4 deiodinated by D2 may be the only source of T3 during neocortical development. We therefore propose that D2 activity at the BCSFB converts the T4 transported across the choroid plexus into T3, thus supplying the brain with active hormone to maintain TH homeostasis.


Asunto(s)
Corteza Cerebral , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hormonas Tiroideas/metabolismo , Factores de Edad , Animales , Animales Recién Nacidos , Peso Corporal/fisiología , Corteza Cerebral/embriología , Corteza Cerebral/crecimiento & desarrollo , Corteza Cerebral/metabolismo , Femenino , Edad Gestacional , Yoduro Peroxidasa/deficiencia , Yoduro Peroxidasa/genética , Isótopos de Yodo/metabolismo , Hígado/embriología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Proteínas de Transporte de Membrana/deficiencia , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Ácidos Monocarboxílicos , Embarazo , ARN Mensajero/metabolismo , Simportadores , Hormonas Tiroideas/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Yodotironina Deyodinasa Tipo II
3.
Biochim Biophys Acta ; 1861(12 Pt A): 1929-1941, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27686967

RESUMEN

New onset diabetes after transplantation (NODAT) is a metabolic disorder that affects 40% of patients on immunosuppressive agent (IA) treatment, such as rapamycin (also known as sirolimus). IAs negatively modulate insulin action in peripheral tissues including skeletal muscle, liver and white fat. However, the effects of IAs on insulin sensitivity and thermogenesis in brown adipose tissue (BAT) have not been investigated. We have analyzed the impact of rapamycin on insulin signaling, thermogenic gene-expression and mitochondrial respiration in BAT. Treatment of brown adipocytes with rapamycin for 16h significantly decreased insulin receptor substrate 1 (IRS1) protein expression and insulin-mediated protein kinase B (Akt) phosphorylation. Consequently, both insulin-induced glucose transporter 4 (GLUT4) translocation to the plasma membrane and glucose uptake were decreased. Early activation of the N-terminal Janus activated kinase (JNK) was also observed, thereby increasing IRS1 Ser 307 phosphorylation. These effects of rapamycin on insulin signaling in brown adipocytes were partly prevented by a JNK inhibitor. In vivo treatment of rats with rapamycin for three weeks abolished insulin-mediated Akt phosphorylation in BAT. Rapamycin also inhibited norepinephrine (NE)-induced lipolysis, the expression of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) and uncoupling protein (UCP)-1 in brown adipocytes. Importantly, basal mitochondrial respiration, proton leak and maximal respiratory capacity were significantly decreased in brown adipocytes treated with rapamycin. In conclusion, we demonstrate, for the first time the important role of brown adipocytes as target cells of rapamycin, suggesting that insulin resistance in BAT might play a major role in NODAT development.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Glucosa/metabolismo , Insulina/metabolismo , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteína Desacopladora 1/metabolismo , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Respiración de la Célula/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 4/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Termogénesis/efectos de los fármacos
4.
Environ Res ; 136: 413-23, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25460663

RESUMEN

Previous studies have shown relationships between organohalogen contaminants (OHCs) and circulating levels of thyroid hormones (THs) in arctic wildlife. However, there is a lack of knowledge concerning the possible functional effects of OHCs on TH status in target tissues for TH-dependent activity. The relationships between circulating (plasma) levels of OHCs and various TH variables in plasma as well as in liver, muscle and kidney tissues from East Greenland sub-adult polar bears (Ursus maritimus) sampled in 2011 (n=7) were therefore investigated. The TH variables included 3.3',5.5'-tetraiodothyronine or thyroxine (T4), 3.3',5-triiodothyronine (T3) and type 1 (D1) and type 2 (D2) deiodinase activities. Principal component analysis (PCA) combined with correlation analyses demonstrated negative relationships between individual polychlorinated biphenyls (PCBs) and their hydroxylated (OH-) metabolites and T4 in both plasma and muscle. There were both positive and negative relationships between individual OHCs and D1 and D2 activities in muscle, liver and kidney tissues. In general, PCBs, OH-PCBs and polybrominated dipehenyl ethers (PBDEs) were positively correlated to D1 and D2 activities, whereas organochlorine pesticides and byproducts (OCPs) were negatively associated with D1 and D2 activities. These results support the hypothesis that OHCs can affect TH status and action in the target tissues of polar bears. TH levels and deiodinase activities in target tissues can be sensitive endpoints for exposure of TH-disrupting compounds in arctic wildlife, and thus, tissue-specific responses in target organs should be further considered when assessing TH disruption in wildlife studies.


Asunto(s)
Halógenos/análisis , Yoduro Peroxidasa/metabolismo , Compuestos Orgánicos/análisis , Hormonas Tiroideas/metabolismo , Ursidae , Contaminantes Químicos del Agua/análisis , Animales , Groenlandia , Halógenos/toxicidad , Yoduro Peroxidasa/sangre , Compuestos Orgánicos/toxicidad , Hormonas Tiroideas/sangre , Contaminantes Químicos del Agua/toxicidad
5.
Biochim Biophys Acta ; 1821(10): 1309-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22766489

RESUMEN

Arachidonic acid (AA) is a polyunsaturated fatty acid that stimulates the proliferation of many cellular types. We studied the mitogenic potential of AA in rat brown preadipocytes in culture and the signaling pathways involved. AA is a potent mitogen which induces 4-fold DNA synthesis in brown preadipocytes. The AA mitogenic effect increases by NE addition. AA also increases the mitogenic action of different growth factor combinations. Other unsaturated and saturated fatty acids do not stimulate DNA synthesis to the same extent as AA. We analyzed the role of PKC and MEK/MAPK signaling pathways. PKC inhibition by bisindolilmaleimide I (BIS) abolishes AA and phorbol ester stimulation of DNA synthesis and reduces the mitogenic activity of different growth factors in brown preadipocytes. Brown preadipocytes in culture express PKC α, δ, ε and ζ isoforms. Pretreatment with high doses of the phorbol ester PDBu, induces downregulation of PKCs ε and δ and reproduces the effect of BIS indicating that AA-dependent induction of DNA synthesis requires PKC activity. AA also activates MEK/MAPK pathway and the inhibition of MEK activity inhibits AA stimulation of DNA synthesis and brown adipocyte proliferation. Inhibition of PKC δ by rottlerin abolishes AA-dependent stimulation of DNA synthesis and MAPK activation, whereas PKC ε inhibition does not produce any effect. In conclusion, our results identify AA as a potent mitogen for brown adipocytes and demonstrate the involvement of the PDBu-sensitive PKC δ isoform and MEK/MAPK pathway in AA-induced proliferation of brown adipocytes. Increased proliferative activity might increase the thermogenic capacity of brown fat.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Ácido Araquidónico/farmacología , ADN/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Proteína Quinasa C/fisiología , Adipocitos Marrones/metabolismo , Secuencia de Aminoácidos , Animales , Activación Enzimática/efectos de los fármacos , Masoprocol/farmacología , Quinasas de Proteína Quinasa Activadas por Mitógenos/fisiología , Datos de Secuencia Molecular , Ratas
6.
Biochim Biophys Acta ; 1811(12): 1194-200, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21840420

RESUMEN

ChREBP is an essential transcription factor for lipogenesis. Its physiological role in adipose tissue has been studied only to a small extent and the control of its expression remains unknown in human adipocytes. We have studied ChREBP mRNA and protein expression levels in the liver and the omental (OM) and subcutaneous (SC) adipose tissues from obese and lean subjects, as well as in human differentiated preadipocytes. Liver and OM and SC adipose tissue biopsies were obtained from lean and obese patients. Human preadipocytes were isolated from the adipose tissues from obese patients and differentiated under adipogenic conditions. ChREBP expression levels were quantified by RT-PCR and Western blot analysis. We found opposing results in terms of ChREBP regulation in the liver and adipose samples. ChREBP increased in the liver from obese compared to lean subjects, whereas the expression decreased in both adipose tissues. The mRNAs of other adipogenic markers were checked in these tissues. The pattern of FASN was similar to the one for ChREBP, ADCY3 decreased in both adipose tissues from obese patients, AP2 decreased only in OM adipose tissue of obese patients and ATGL did not change. The levels of ChREBP mRNA and protein showed dramatic increases during the differentiation of human OM and SC preadipocytes. In conclusion, ChREBP expression has an opposite regulation in the liver and adipose tissue from obese subjects which is compatible with the increased hepatic lipogenesis and decreased adipocytic lipogenesis found in these patients. The dramatic increase of ChREBP mRNA and protein levels during preadipocyte differentiation suggests a role in adipogenesis.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Hígado/metabolismo , Obesidad/genética , Obesidad/metabolismo , Epiplón/metabolismo , Grasa Subcutánea/metabolismo , Adenilil Ciclasas/genética , Adenilil Ciclasas/metabolismo , Adipocitos/citología , Adulto , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Western Blotting , Diferenciación Celular , Acido Graso Sintasa Tipo I/genética , Acido Graso Sintasa Tipo I/metabolismo , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Femenino , Expresión Génica , Humanos , Lipasa/genética , Lipasa/metabolismo , Lipogénesis , Masculino , Persona de Mediana Edad , Obesidad/patología , Epiplón/citología , Cultivo Primario de Células , ARN Mensajero/análisis , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Grasa Subcutánea/citología
7.
BMC Physiol ; 12: 4, 2012 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-22471305

RESUMEN

BACKGROUND: The expansion of adipose tissue is linked to the development of its vasculature, which appears to have the potential to regulate the onset of obesity. However, at present, there are no studies highlighting the relationship between human adipose tissue angiogenesis and obesity-associated insulin resistance (IR). RESULTS: Our aim was to analyze and compare angiogenic factor expression levels in both subcutaneous (SC) and omentum (OM) adipose tissues from morbidly obese patients (n = 26) with low (OB/L-IR) (healthy obese) and high (OB/H-IR) degrees of IR, and lean controls (n = 17). Another objective was to examine angiogenic factor correlations with obesity and IR.Here we found that VEGF-A was the isoform with higher expression in both OM and SC adipose tissues, and was up-regulated 3-fold, together with MMP9 in OB/L-IR as compared to leans. This up-regulation decreased by 23% in OB/-H-IR compared to OB/L-IR. On the contrary, VEGF-B, VEGF-C and VEGF-D, together with MMP15 was down-regulated in both OB/H-IR and OB/L-IR compared to lean patients. Moreover, MMP9 correlated positively and VEGF-C, VEGF-D and MMP15 correlated negatively with HOMA-IR, in both SC and OM. CONCLUSION: We hereby propose that the alteration in MMP15, VEGF-B, VEGF-C and VEGF-D gene expression may be caused by one of the relevant adipose tissue processes related to the development of IR, and the up-regulation of VEGF-A in adipose tissue could have a relationship with the prevention of this pathology.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Resistencia a la Insulina/fisiología , Metaloproteasas/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Inductores de la Angiogénesis , Biomarcadores/metabolismo , Expresión Génica , Humanos , Metaloproteasas/genética , Neovascularización Fisiológica , Obesidad/fisiopatología , Obesidad Mórbida/metabolismo , Epiplón
8.
J Clin Invest ; 118(3): 975-83, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18259611

RESUMEN

Thyroid hormone is a critical determinant of cellular metabolism and differentiation. Precise tissue-specific regulation of the active ligand 3,5,3'-triiodothyronine (T3) is achieved by the sequential removal of iodine groups from the thyroid hormone molecule, with type 3 deiodinase (D3) comprising the major inactivating pathway that terminates the action of T3 and prevents activation of the prohormone thyroxine. Using cells endogenously expressing D3, we found that hypoxia induced expression of the D3 gene DIO3 by a hypoxia-inducible factor-dependent (HIF-dependent) pathway. D3 activity and mRNA were increased both by hypoxia and by hypoxia mimetics that increase HIF-1. Using ChIP, we found that HIF-1alpha interacted specifically with the DIO3 promoter, indicating that DIO3 may be a direct transcriptional target of HIF-1. Endogenous D3 activity decreased T3-dependent oxygen consumption in both neuronal and hepatocyte cell lines, suggesting that hypoxia-induced D3 may reduce metabolic rate in hypoxic tissues. Using a rat model of cardiac failure due to RV hypertrophy, we found that HIF-1alpha and D3 proteins were induced specifically in the hypertrophic myocardium of the RV, creating an anatomically specific reduction in local T3 content and action. These results suggest a mechanism of metabolic regulation during hypoxic-ischemic injury in which HIF-1 reduces local thyroid hormone signaling through induction of D3.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Hipoxia/metabolismo , Yoduro Peroxidasa/fisiología , Isquemia/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Inducción Enzimática , Hipertrofia Ventricular Derecha/metabolismo , Masculino , Ratas , Ratas Wistar , Transducción de Señal , Triyodotironina/fisiología
9.
Cell Physiol Biochem ; 28(5): 969-80, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178948

RESUMEN

UNLABELLED: Uncoupling protein 1 (UCP-1), the specific marker of brown adipose tissue, is transcriptionally activated in response to adrenergic stimuli and thyroid hormones are necessary for its full expression. We describe differences in the regulation of UCP-1 mRNA expression between rat and mouse brown adipocytes in culture, using norepinephrine (NE), triiodothyronine (T3), insulin and retinoic acid (RA). RESULTS: NE and cAMP-elevating agents strongly increase UCP-1 mRNA levels in cultures of mouse adipocytes, but increases are low in those from rat. In rat adipocytes NE poorly increases UCP-1 mRNA expression and T3 markedly increases the adrenergic response of UCP-1, an effect not observed in mouse adipocytes. In the absence of insulin, T3 itself increases UCP-1 mRNA in rat adipocytes and enhances the response to NE, while in mouse adipocytes no effect of T3 is observed. RA by itself stimulates UCP-1 mRNA in mouse adipocytes, but not in those from rat. In rat cultures, RA requires the presence of NE and/or T3. CONCLUSIONS: We find important differences in the hormonal regulation of UCP-1 mRNA expression in cultured preadipocytes depending on the species used as donor; those differences are observed using identical culture conditions and should be considered when doing cultures from these species.


Asunto(s)
Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/metabolismo , Insulina/farmacología , Canales Iónicos/genética , Proteínas Mitocondriales/genética , Norepinefrina/farmacología , Tretinoina/farmacología , Triyodotironina/farmacología , Adipocitos Marrones/citología , Animales , Células Cultivadas , Hormonas/farmacología , Canales Iónicos/metabolismo , Ratones , Proteínas Mitocondriales/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Desacopladora 1
10.
Medicina (B Aires) ; 81(3): 346-358, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34137693

RESUMEN

Hepatocellular carcinoma (HCC) is the most common primary liver tumor. Hexachlorobenzene (HCB) is an endocrine disruptor and a liver tumor promoter. Deregulation of thyroid hormone (TH) homeostasis may play a significant role in early neoplastic transformation. The aim of this study was to evaluate the relation between TH metabolism and the regulation of cell growth in an in vivo and in vitro model. We examined the role of transforming growth factor-ß1 (TGF-ß1) on TH deiodinase expression and hepatocyte proliferation. An initiation (DEN)/promotion (HCB) tumor model from rat liver and HepG2 cells were used. We evaluated PCNA, p21, p27, SMAD2/3, TGF-ß1, deiodinase 1 (D1), D3, protein expression levels; D1 and D3 mRNA expression; TH and TGF-ß1, D1, D3, and GST-P protein levels in focal/non-focal areas. In vivo, HCB decreased triiodothyronine (T3) and D1 mRNA levels and increased thyroxine (T4) and D3 mRNA levels in liver from DEN+HCB vs. DEN group. HCB increased protein levels from D3, TGF-ß1, and PCNA and decreased D1 in focal-areas. In vitro, HCB increased PCNA, pSMAD 2/3, and TGF-ß1 protein levels and mRNA expression and decreased p21 and p27 protein levels. Exogenous T3 treatment prevent HCB induced molecular alterations related to hepatocyte proliferation whereas T4 did not have any effect. These effects were prevented by using a TGF-ß1 receptor II inhibitor. Results suggest that alteration of TH homeostasis, through D1 function, play a key role in hepatocyte proliferation and that TGF-ß1-SMAD pathway is involved in this process confirming their role in early neoplastic transformation in HCC.


El hepatocarcinoma (HCC) es un tumor hepático primario. El hexaclorobenceno (HCB) es un disruptor endocrino y un promotor de tumores hepáticos. La desregulación de la homeostasis de las hormonas tiroideas (HT) puede ser un proceso importante para la transformación neoplásica temprana. Nuestro objetivo fue evaluar la relación entre el metabolismo de las HT y la regulación de la proliferación celular. Se utilizó un modelo tumoral de iniciación (DEN)/promoción (HCB) de hígado de rata (in vivo) (DEN/HCB) y células HepG2 (in vitro). Evaluamos los niveles de PCNA, p21, p27, SMAD2/3, TGF-ß1, D1, D3, ARNm de D1 y D3, HT y los niveles de TGF-ß1, D1, D3 y GST-P en áreas focales/no focales. In vivo, HCB disminuyó los niveles de T3 y ARNm de la D1 y aumentó los niveles de T4 y ARNm de D3 del grupo DEN + HCB frente al grupo DEN. El HCB aumentó los niveles de D3, TGF-ß1 y PCNA y disminuyó el D1 en las áreas focales. In vitro, HCB aumentó los niveles de PCNA, pSMAD 2/3 y TGF-ß1 y la expresión de ARNm mientras que disminuyó los niveles de p21 y p27. El tratamiento con T3 exógeno previno las alteraciones moleculares relacionadas con la proliferación hepatocitaria. Estos efectos se evitaron utilizando un inhibidor del receptor II de TGF-ß1. Los resultados sugieren que la alteración de la homeostasis de HT, a través de la D1 y la vía TGF-ß1-SMAD, juega un papel clave en la proliferación celular y en las transformaciones neoplásicas tempranas en el HCC.


Asunto(s)
Carcinoma Hepatocelular , Yoduro Peroxidasa , Neoplasias Hepáticas , Factor de Crecimiento Transformador beta1 , Animales , Proliferación Celular , Yoduro Peroxidasa/genética , Ratas
11.
Redox Biol ; 48: 102171, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34736121

RESUMEN

Therapeutic potential of metformin in obese/diabetic patients has been associated to its ability to combat insulin resistance. However, it remains largely unknown the signaling pathways involved and whether some cell types are particularly relevant for its beneficial effects. M1-activation of macrophages by bacterial lipopolysaccharide (LPS) promotes a paracrine activation of hypoxia-inducible factor-1α (HIF1α) in brown adipocytes which reduces insulin signaling and glucose uptake, as well as ß-adrenergic sensitivity. Addition of metformin to M1-polarized macrophages blunted these signs of brown adipocyte dysfunction. At the molecular level, metformin inhibits an inflammatory program executed by HIF1α in macrophages by inducing its degradation through the inhibition of mitochondrial complex I activity, thereby reducing oxygen consumption in a reactive oxygen species (ROS)-independent manner. In obese mice, metformin reduced inflammatory features in brown adipose tissue (BAT) such as macrophage infiltration, proinflammatory signaling and gene expression, and restored the response to cold exposure. In conclusion, the impact of metformin on macrophages by suppressing a HIF1α-dependent proinflammatory program is likely responsible for a secondary beneficial effect on insulin-mediated glucose uptake and ß-adrenergic responses in brown adipocytes.

12.
Sci Rep ; 10(1): 12317, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32704052

RESUMEN

The contribution of the nucleotide-binding oligomerization domain protein NOD1 to obesity has been investigated in mice fed a high fat diet (HFD). Absence of NOD1 accelerates obesity as early as 2 weeks after feeding a HFD. The obesity was due to increases in abdominal and inguinal adipose tissues. Analysis of the resting energy expenditure showed an impaired function in NOD1-deficient animals, compatible with an alteration in thyroid hormone homeostasis. Interestingly, free thyroidal T4 increased in NOD1-deficient mice fed a HFD and the expression levels of UCP1 in brown adipose tissue were significantly lower in NOD1-deficient mice than in the wild type animals eating a HFD, thus contributing to the observed adiposity in NOD1-deficient mice. Feeding a HFD resulted in an alteration of the proinflammatory profile of these animals, with an increase in the infiltration of inflammatory cells in the liver and in the white adipose tissue, and an elevation of the circulating levels of TNF-α. In addition, alterations in the gut microbiota in NOD1-deficient mice correlate with increased vulnerability of their ecosystem to the HFD challenge and affect the immune-metabolic phenotype of obese mice. Together, the data are compatible with a protective function of NOD1 against low-grade inflammation and obesity under nutritional conditions enriched in saturated lipids. Moreover, one of the key players of this early obesity onset is a dysregulation in the metabolism and release of thyroid hormones leading to reduced energy expenditure, which represents a new role for these hormones in the metabolic actions controlled by NOD1.


Asunto(s)
Dieta Alta en Grasa , Conducta Alimentaria , Microbioma Gastrointestinal , Homeostasis , Proteína Adaptadora de Señalización NOD1/deficiencia , Hormonas Tiroideas/metabolismo , Adipocitos/metabolismo , Tejido Adiposo Blanco/patología , Animales , Biodiversidad , Peso Corporal , Hígado Graso/patología , Prueba de Tolerancia a la Glucosa , Inflamación/patología , Intestinos/patología , Lípidos/química , Hígado/patología , Metabolómica , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Adaptadora de Señalización NOD1/metabolismo , Obesidad/sangre , Obesidad/microbiología , Obesidad/patología , Glándula Tiroides/patología , Glándula Tiroides/fisiopatología , Hormonas Tiroideas/sangre
13.
Mol Metab ; 42: 101097, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33049408

RESUMEN

OBJECTIVE: Metainflammation is a chronic low-grade inflammatory state induced by obesity and associated comorbidities, including peripheral insulin resistance. Brown adipose tissue (BAT), a therapeutic target against obesity, is an insulin target tissue sensitive to inflammation. Therefore, it is necessary to find strategies to protect BAT against the effects of inflammation in energy balance. In this study, we explored the impact of moderate sirtuin 1 (SIRT1) overexpression on insulin sensitivity and ß-adrenergic responses in BAT and brown adipocytes (BA) under pro-inflammatory conditions. METHODS: The effect of inflammation on BAT functionality was studied in obese db/db mice and lean wild-type (WT) mice or mice with moderate overexpression of SIRT1 (SIRT1Tg+) injected with a low dose of bacterial lipopolysaccharide (LPS) to mimic endotoxemia. We also conducted studies on differentiated BA (BA-WT and BA-SIRT1Tg+) exposed to a macrophage-derived pro-inflammatory conditioned medium (CM) to evaluate the protection of SIRT1 overexpression in insulin signaling and glucose uptake, mitochondrial respiration, fatty acid oxidation (FAO), and norepinephrine (NE)-mediated-modulation of uncoupling protein-1 (UCP-1) expression. RESULTS: BAT from the db/db mice was susceptible to metabolic inflammation manifested by the activation of pro-inflammatory signaling cascades, increased pro-inflammatory gene expression, tissue-specific insulin resistance, and reduced UCP-1 expression. Impairment of insulin and noradrenergic responses were also found in the lean WT mice upon LPS injection. In contrast, BAT from the mice with moderate overexpression of SIRT1 (SIRT1Tg+) was protected against LPS-induced activation of pro-inflammatory signaling, insulin resistance, and defective thermogenic-related responses upon cold exposure. Importantly, the decline in triiodothyronine (T3) levels in the circulation and intra-BAT after exposure of the WT mice to LPS and cold was markedly attenuated in the SIRT1Tg+ mice. In vitro BA experiments in the two genotypes revealed that upon differentiation with a T3-enriched medium and subsequent exposure to a macrophage-derived pro-inflammatory CM, only BA-SIRT1Tg+ fully recovered insulin and noradrenergic responses. CONCLUSIONS: This study has ascertained the benefit of the moderate overexpression of SIRT1 to confer protection against defective insulin and ß-adrenergic responses caused by BAT inflammation. Our results have potential therapeutic value in combinatorial therapies for BAT-specific thyromimetics and SIRT1 activators to combat metainflammation in this tissue.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Sirtuina 1/metabolismo , Adipocitos/metabolismo , Adipocitos/fisiología , Adipocitos Marrones/metabolismo , Adipocitos Marrones/fisiología , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/fisiología , Animales , Metabolismo Energético , Femenino , Expresión Génica/genética , Regulación de la Expresión Génica/genética , Inflamación/prevención & control , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Obesidad/metabolismo , Receptores Adrenérgicos beta/metabolismo , Sirtuina 1/genética , Sirtuina 1/fisiología , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1/metabolismo
15.
J Exp Zool B Mol Dev Evol ; 312B(3): 231-46, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19306324

RESUMEN

Thyroid hormone-induced metamorphosis seems to represent an ancestral feature of chrordates (urochordates, cephalochordates and vertebrates), but also of nonchordate animals. Although thyroid hormones and thyroid hormone receptor profiles during metamorphosis have been analyzed in different vertebrate taxa, including fish, developmental expression and activity of type 2 (dio2, D2) and type 3 (dio3, D3) iodothyronine deiodinases, two key enzymes in anuran metamorphosis, remain unknown in any fish species. The aim of this work was to investigate the development of thyroid hormone system during the metamorphosis of a flatfish species, the Senegalese sole, focusing on the deiodinases developmental profile. We have cloned sole D2 and D3 and analyzed several parameters of thyroid hormones system in pre-, early-, middle-, and late-metamorphic larvae. Both deiodinases contain in their catalytic centers an UGA triplet encoding for a selenocystein (Sec) residue as expected. Left eye migration and rotation in body position were associated with a significant increase in both thyroid hormones and thyroid hormone receptors at the middle-late metamorphic stages. Although dio2 expression slightly increased during metamorphosis, D2 activity augmentation was much more significant. Sole dio3 expression declined only slightly, whereas the D3 activity clearly decreased at mid-late metamorphic period. This developmental profile of deiodinases sustained the rise of thyroid hormones levels observed during sole metamorphosis. No clear cut daily rhythms were observed in the parameters analyzed although it seemed that thyroid hormone system was more active during daytime, in particular at late metamorphic stages. These developmental changes point out the importance not only of thyroid hormones and their receptors but also of dio2 and dio3 in mediating flatfish metamorphosis, as it has been described in amphibians.


Asunto(s)
Peces Planos/fisiología , Yoduro Peroxidasa/metabolismo , Metamorfosis Biológica/fisiología , Receptores de Hormona Tiroidea/metabolismo , Tiroxina/metabolismo , Triyodotironina/metabolismo , Animales , Química Encefálica , Clonación Molecular , Peces Planos/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Yoduro Peroxidasa/genética , Filogenia , ARN Mensajero/metabolismo , Receptores de Hormona Tiroidea/genética
16.
Mol Cell Biol ; 26(6): 2317-26, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16508007

RESUMEN

dickkopf (dkk) genes encode a small family of secreted Wnt antagonists, except for dkk3, which is divergent and whose function is poorly understood. Here, we describe the generation and characterization of dkk3 mutant mice. dkk3-deficient mice are viable and fertile. Phenotypic analysis shows no major alterations in organ morphology, physiology, and most clinical chemistry parameters. Since Dkk3 was proposed to function as thyroid hormone binding protein, we have analyzed deiodinase activities, as well as thyroid hormone levels. Mutant mice are euthyroid, and the data do not support a relationship of dkk3 with thyroid hormone metabolism. Altered phenotypes in dkk3 mutant mice were observed in the frequency of NK cells, immunoglobulin M, hemoglobin, and hematocrit levels, as well as lung ventilation. Furthermore, dkk3-deficient mice display hyperactivity.


Asunto(s)
Conducta Animal/fisiología , Sistema Inmunológico/fisiología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Ventilación Pulmonar/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Antígenos Helmínticos/genética , Antígenos Helmínticos/inmunología , Eritrocitos/patología , Femenino , Inmunoglobulina M/sangre , Péptidos y Proteínas de Señalización Intercelular/inmunología , Yoduro Peroxidasa/metabolismo , Pulmón/fisiopatología , Masculino , Ratones , Ratones Mutantes , Tiroxina/metabolismo , Triyodotironina/metabolismo , Yodotironina Deyodinasa Tipo II
17.
Aging Cell ; 18(3): e12948, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30920127

RESUMEN

Age-related increased adiposity is an important contributory factor in the development of insulin resistance (IR) and is associated with metabolic defects. Caloric restriction (CR) is known to induce weight loss and to decrease adiposity while preventing metabolic risk factors. Here, we show that moderate 20% CR delays early deleterious effects of aging on white and brown adipose tissue (WAT and BAT, respectively) function and improves peripheral IR. To elucidate the role of CR in delaying early signs of aging, young (3 months), middle-aged (12 months), and old (20 months) mice fed al libitum and middle-aged and old mice subjected to early-onset CR were used. We show that impaired plasticity of subcutaneous WAT (scWAT) contributes to IR, which is already evident in middle-aged mice. Moreover, alteration of thyroid axis status with age is an important factor contributing to BAT dysfunction in middle-aged animals. Both defects in WAT and BAT/beige cells are ameliorated by CR. Accordingly, CR attenuated the age-related decline in scWAT function and decreased the extent of fibro-inflammation. Furthermore, CR promoted scWAT browning. In brief, our study identifies the contribution of scWAT impairment to age-associated metabolic dysfunction and identifies browning in response to food restriction, as a potential therapeutic strategy to prevent the adverse metabolic effects in middle-aged animals.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Envejecimiento/metabolismo , Restricción Calórica , Animales , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones de la Cepa 129 , Tomografía Computarizada por Tomografía de Emisión de Positrones
18.
J Clin Endocrinol Metab ; 93(1): 267-77, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17940114

RESUMEN

CONTEXT: We studied two families from Galicia (northwest Spain) with Pendred syndrome (PS) and unusual thyroid phenotypes. In family A, the proposita had a large goiter and hypothyroxinemia but normal TSH and free T3 (FT3). In family B, some affected members showed deafness but not goiter. OBJECTIVE: Our objective was to identify the mutations causing PS and molecular mechanisms underlying the thyroid phenotypes. INTERVENTIONS: Interventions included extraction of DNA and of thyroid tissue. PATIENTS: Propositi and 10 members of the two families participated in the study. MAIN OUTCOME MEASURES: Main outcome measures included SLC26A4 gene analysis, deiodinase activities in thyroid tissue, and c.416-1G-->A effects on SLC26A4 splicing. In addition, a primary PS thyrocyte culture, T-PS2, was obtained from propositus B and compared with another culture of normal human thyrocytes, NT, by Western blotting, confocal microscopy, and iodine uptake kinetics. RESULTS: Proposita A was heterozygous for c.578C-->T and c.279delT, presented with goiter, and had normal TSH and FT3 but low FT4 attributable to high type 1 and type 2 iodothyronine deiodinase activities in the goiter. Propositus B bore c.279delT and a novel mutation c.416-1G-->A; some deaf relatives were homozygous for c.416-1G-->A but did not present goiter. The c.279delT mutation was associated with identical haplotype in the two families. T-PS2 showed truncated pendrin retained intracellularly and high iodine uptake with low efflux leading to iodine retention. CONCLUSIONS: c.279delT is a founder mutation in Galicia. Proposita A adapted to poor organification by increasing deiodinase activities in the goiter, avoiding hypothyroidism. Lack of goiter in subjects homozygous for c.416-1G-->A was due to incomplete penetrance allowing synthesis of some wild-type pendrin. Intracellular iodine retention, as seen in T-PS2, could play a role in thyroid alterations in PS.


Asunto(s)
Bocio Nodular/genética , Pérdida Auditiva Sensorineural/genética , Adulto , Secuencia de Aminoácidos , Femenino , Bocio Nodular/enzimología , Bocio Nodular/patología , Haplotipos , Pérdida Auditiva Sensorineural/enzimología , Pérdida Auditiva Sensorineural/patología , Humanos , Inmunohistoquímica , Yoduro Peroxidasa/biosíntesis , Yoduro Peroxidasa/genética , Yodo/farmacocinética , Masculino , Proteínas de Transporte de Membrana/biosíntesis , Proteínas de Transporte de Membrana/genética , Persona de Mediana Edad , Datos de Secuencia Molecular , Transportadores de Ácidos Monocarboxílicos/biosíntesis , Transportadores de Ácidos Monocarboxílicos/genética , Linaje , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , España , Transportadores de Sulfato , Simportadores , Síndrome , Pruebas de Función de la Tiroides , Yodotironina Deyodinasa Tipo II
19.
Thyroid ; 18(2): 185-95, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18279019

RESUMEN

Thyroid hormones act as pleiotropic factors in many tissues during development, by regulating genes involved in differentiation. The adipose tissue, a target of thyroid hormones, is the main place for energy storage and acts as a regulator of energy balance, sending signals to keep metabolic control. Adipogenesis is a complex process that involves proliferation of preadipocytes and its differentiation into mature adipocytes. This process is regulated by several transcription factors (CCAAT/enhancer-binding proteins [C/EBPs], peroxisome proliferator-activated receptors [PPARs]) that act coordinately, activating adipocyte-specific genes that will provide the adipocytic phenotype. Thyroid hormones regulate many of those genes, markers of differentiation of adipocytes, those involved in lipogenesis, lipolysis, and thermogenesis in the brown adipose tissue (BAT). Triiodothyronine (T3) actions are achieved either directly through specific thyroid response elements (TREs), by regulating other key genes as PPARs, or through specific isoforms of the nuclear T3 receptors. The availability of T3 is regulated through the deiodinases D3, D2, and D1. D3 is activated by serum and mitogens during proliferation of preadipocytes, while D2 is linked to the differentiation program of adipocytes, through the C/EBPs that govern its functionality, providing the T3 required for thermogenesis and lipogenesis. The relationship between white adipose tissue (WAT) and BAT and the possible reactivation of WAT by activation of uncoupling protein-1 (UCP1) is discussed.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis , Transducción de Señal , Triyodotironina/metabolismo , Adipocitos Marrones/enzimología , Adipocitos Blancos/enzimología , Adipogénesis/genética , Animales , Proliferación Celular , Metabolismo Energético , Humanos , Yoduro Peroxidasa/metabolismo , Canales Iónicos/metabolismo , Lipogénesis , Proteínas Mitocondriales/metabolismo , Receptores de Hormona Tiroidea/metabolismo , Transducción de Señal/genética , Termogénesis , Factores de Transcripción/metabolismo , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
20.
Trends Endocrinol Metab ; 18(9): 338-43, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17962037

RESUMEN

Iodine deficiency remains the most frequent cause worldwide, after starvation, of preventable mental retardation in children. It causes maternal hypothyroxinemia, which affects pregnant women even in apparently iodine-sufficient areas, and often goes unnoticed because L-thyroxine (T4) levels remain within the normal range, and thyroid-stimulating hormone (TSH) is not increased. Even a mild hypothyroxinemia during pregnancy increases the risk of neurodevelopmental abnormalities, and experimental data clearly demonstrate that it damages the cortical cytoarchitecture of the fetal brain. The American Thyroid Association (ATA) recommends a supplement of 150 microg iodine/day during pregnancy and lactation, in addition to the use of iodized salt. We discuss the importance of iodine supplementation to ensure adequate T4 levels in all women who are considering conception and throughout pregnancy and lactation.


Asunto(s)
Yodo/administración & dosificación , Yodo/deficiencia , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/prevención & control , Oligoelementos/administración & dosificación , Animales , Femenino , Humanos , Lactante , Recién Nacido , Yodo/efectos adversos , Lactancia/metabolismo , Política Nutricional , Embarazo , Atención Prenatal , Salud Pública , Roedores , Cloruro de Sodio Dietético/administración & dosificación , Oligoelementos/efectos adversos , Oligoelementos/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA