Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell ; 183(1): 94-109.e23, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32937105

RESUMEN

Cardiomyocytes are subjected to the intense mechanical stress and metabolic demands of the beating heart. It is unclear whether these cells, which are long-lived and rarely renew, manage to preserve homeostasis on their own. While analyzing macrophages lodged within the healthy myocardium, we discovered that they actively took up material, including mitochondria, derived from cardiomyocytes. Cardiomyocytes ejected dysfunctional mitochondria and other cargo in dedicated membranous particles reminiscent of neural exophers, through a process driven by the cardiomyocyte's autophagy machinery that was enhanced during cardiac stress. Depletion of cardiac macrophages or deficiency in the phagocytic receptor Mertk resulted in defective elimination of mitochondria from the myocardial tissue, activation of the inflammasome, impaired autophagy, accumulation of anomalous mitochondria in cardiomyocytes, metabolic alterations, and ventricular dysfunction. Thus, we identify an immune-parenchymal pair in the murine heart that enables transfer of unfit material to preserve metabolic stability and organ function. VIDEO ABSTRACT.


Asunto(s)
Macrófagos/metabolismo , Mitocondrias/metabolismo , Miocitos Cardíacos/metabolismo , Anciano , Animales , Apoptosis , Autofagia , Femenino , Corazón/fisiología , Homeostasis , Humanos , Macrófagos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/fisiología , Infarto del Miocardio/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/fisiología , Fagocitosis/fisiología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa c-Mer/metabolismo
2.
Nat Metab ; 2(9): 974-988, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32943786

RESUMEN

Proinflammatory macrophages are key in the development of obesity. In addition, reactive oxygen species (ROS), which activate the Fgr tyrosine kinase, also contribute to obesity. Here we show that ablation of Fgr impairs proinflammatory macrophage polarization while preventing high-fat diet (HFD)-induced obesity in mice. Systemic ablation of Fgr increases lipolysis and liver fatty acid oxidation, thereby avoiding steatosis. Knockout of Fgr in bone marrow (BM)-derived cells is sufficient to protect against insulin resistance and liver steatosis following HFD feeding, while the transfer of Fgr-expressing BM-derived cells reverts protection from HFD feeding in Fgr-deficient hosts. Scavenging of mitochondrial peroxides is sufficient to prevent Fgr activation in BM-derived cells and HFD-induced obesity. Moreover, Fgr expression is higher in proinflammatory macrophages and correlates with obesity traits in both mice and humans. Thus, our findings reveal the mitochondrial ROS-Fgr kinase as a key regulatory axis in proinflammatory adipose tissue macrophage activation, diet-induced obesity, insulin resistance and liver steatosis.


Asunto(s)
Dieta Alta en Grasa , Inflamación/fisiopatología , Activación de Macrófagos , Obesidad/enzimología , Obesidad/fisiopatología , Proteínas Proto-Oncogénicas/metabolismo , Familia-src Quinasas/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Hígado Graso/genética , Hígado Graso/fisiopatología , Resistencia a la Insulina , Interleucina-1beta/biosíntesis , Imagen por Resonancia Magnética , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias Hepáticas/metabolismo , Obesidad/genética , Proteínas Proto-Oncogénicas/genética , Especies Reactivas de Oxígeno/metabolismo , Familia-src Quinasas/genética
3.
Sci Adv ; 6(31): eaba5345, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32832682

RESUMEN

Heteroplasmy, multiple variants of mitochondrial DNA (mtDNA) in the same cytoplasm, may be naturally generated by mutations but is counteracted by a genetic mtDNA bottleneck during oocyte development. Engineered heteroplasmic mice with nonpathological mtDNA variants reveal a nonrandom tissue-specific mtDNA segregation pattern, with few tissues that do not show segregation. The driving force for this dynamic complex pattern has remained unexplained for decades, challenging our understanding of this fundamental biological problem and hindering clinical planning for inherited diseases. Here, we demonstrate that the nonrandom mtDNA segregation is an intracellular process based on organelle selection. This cell type-specific decision arises jointly from the impact of mtDNA haplotypes on the oxidative phosphorylation (OXPHOS) system and the cell metabolic requirements and is strongly sensitive to the nuclear context and to environmental cues.

4.
Thyroid ; 26(5): 618-26, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26701289

RESUMEN

BACKGROUND: Monocarboxylate transporter 8 (MCT8) is a thyroid hormone-specific cell membrane transporter. Mutations in the MCT8 gene lead to profound psychomotor retardation and abnormal thyroid hormone serum levels with low thyroxine (T4) and high triiodothyronine (T3). Currently, therapeutic options for patients are limited. Triiodothyroacetic acid (TRIAC) has potential therapeutic value. The aim of this study was to evaluate the effects and efficacy of therapeutic doses of TRIAC on Mct8-deficient mice (Mct8KO). METHODS: Wild-type (Wt) and Mct8KO mice were treated with 30 ng TRIAC/g of body weight/day, given in drinking water, from postnatal day 21 to 30. TRIAC, T4 and T3 levels in plasma, as well as T3 and TRIAC content in the cerebral cortex and striatum were measured by specific radioimmunoassays. The activities of deiodinases 1 and 2 were measured in liver and cortex. The effect of TRIAC treatment in the expression of T3-dependent genes was measured in the heart, cerebral cortex, and striatum. RESULTS: Plasma TRIAC concentration were the same in Wt and Mct8KO animals after treatment. TRIAC treatment greatly decreased plasma T4 in Wt and Mct8KO mice, and reduced T3 to normal levels in the Mct8KO mice. Deiodinase 1 activity and gene expression in the liver increased, while it did not have any effect on the expression of Serca2a in the heart. TRIAC treatment did not induce the expression of T3-dependent genes in the cerebral cortex or striatum, but further decreased expression of Flywch2 in the cortex and Aldh1a1 and Flywch2 in the striatum. Direct measurements of TRIAC and T3 content in the cortex and striatum revealed a decrease in T3 after treatment with no significant increase in the level of endogenous TRIAC. CONCLUSIONS: Therapeutic doses of TRIAC in Mct8KO mice restored plasma T3 levels but severely decreased T4 levels. TRIAC has a direct effect on deiodinase 1 in the liver and does not have an effect on gene expression in the heart. The increase in the plasma TRIAC levels after treatment is not sufficient to increase TRIAC levels in the brain and to promote the expression of T3-dependent genes in brain cells. Instead, it leads to a state of brain hypothyroidism with reduced T3 content.


Asunto(s)
Proteínas de Transporte de Membrana/genética , Tiroxina/sangre , Triyodotironina/análogos & derivados , Triyodotironina/sangre , Animales , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Yoduro Peroxidasa/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Ratones , Ratones Noqueados , Transportadores de Ácidos Monocarboxílicos , Simportadores , Triyodotironina/farmacología
5.
PLoS One ; 9(5): e96915, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24819605

RESUMEN

Thyroid hormone entry into cells is facilitated by transmembrane transporters. Mutations of the specific thyroid hormone transporter, MCT8 (Monocarboxylate Transporter 8, SLC16A2) cause an X-linked syndrome of profound neurological impairment and altered thyroid function known as the Allan-Herndon-Dudley syndrome. MCT8 deficiency presumably results in failure of thyroid hormone to reach the neural target cells in adequate amounts to sustain normal brain development. However during the perinatal period the absence of Mct8 in mice induces a state of cerebral cortex hyperthyroidism, indicating increased brain access and/or retention of thyroid hormone. The contribution of other transporters to thyroid hormone metabolism and action, especially in the context of MCT8 deficiency is not clear. We have analyzed the role of the heterodimeric aminoacid transporter Lat2 (Slc7a8), in the presence or absence of Mct8, on thyroid hormone concentrations and on expression of thyroid hormone-dependent cerebral cortex genes. To this end we generated Lat2-/-, and Mct8-/yLat2-/- mice, to compare with wild type and Mct8-/y mice during postnatal development. As described previously the single Mct8 KO neonates had a transient increase of 3,5,3'-triiodothyronine concentration and expression of thyroid hormone target genes in the cerebral cortex. Strikingly the absence of Lat2 in the double Mct8Lat2 KO prevented the effect of Mct8 inactivation in newborns. The Lat2 effect was not observed from postnatal day 5 onwards. On postnatal day 21 the Mct8 KO displayed the typical pattern of thyroid hormone concentrations in plasma, decreased cortex 3,5,3'-triiodothyronine concentration and Hr expression, and concomitant Lat2 inactivation produced little to no modifications. As Lat2 is expressed in neurons and in the choroid plexus, the results support a role for Lat2 in the supply of thyroid hormone to the cerebral cortex during early postnatal development.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Corteza Cerebral/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Hipertiroidismo/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Animales , Animales Recién Nacidos , Femenino , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Hipertiroidismo/genética , Masculino , Proteínas de Transporte de Membrana/genética , Ratones , Transportadores de Ácidos Monocarboxílicos , Simportadores , Triyodotironina/metabolismo
6.
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
7.
Endocrinology ; 151(10): 5074-83, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20719854

RESUMEN

Brown adipose tissue (BAT) thermogenesis increases when uncoupling protein-1 (UCP1) is activated adrenergically and requires T3. In humans, UCP1 activation in BAT seems involved in body weight maintenance. BAT type 2 deiodinase (D2) increases in response to adrenergic agents, producing the T3 required for UCP1 expression. T3 actions are mediated by thyroid hormone nuclear T3 receptors (TR), TRα and TRß. Studies in mice suggest that TRß is required for UCP1 induction, whereas TRα regulates body temperature and adrenergic sensitivity. In the present study, we compare the effects of T3 vs. specific TRß1 and TRα1 agonists [GC-1 and CO23] on the adrenergic induction of UCP1 and D2 in cultured rat brown adipocytes. T3 and GC-1 produced similar increases on UCP1, whereas CO23 increased UCP1 only at high doses (50 nm). GC-1 at low doses (0.2-10 nm) was less potent than T3, increasing the adrenergic stimulation of D2 activity and mRNA. At higher doses, GC-1 further stimulated whereas T3 inhibited D2 activity but not D2 mRNA, suggesting posttranscriptional effects. CO23 had no effect on D2 activity but increased D2 mRNA. T3, GC-1, or CO23 by themselves did not increase UCP1 or D2 mRNA. High T3 doses shortened D2 half-life and increased D2 turnover via proteasome, whereas GC-1 did not change D2 stability. The α1- and α2-adrenergic D2 responses increased using high T3 doses. In summary, T3 increases the adrenergic stimulation of UCP1 and D2 expression mostly via the TRß1 isoform, and in brown adipocytes, D2 is protected from degradation by the action of T3 on TRß1.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Yoduro Peroxidasa/genética , Canales Iónicos/genética , Proteínas Mitocondriales/genética , Receptores beta de Hormona Tiroidea/fisiología , Acetatos/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Adrenérgicos/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Hidantoínas/farmacología , Yoduro Peroxidasa/metabolismo , Canales Iónicos/metabolismo , Proteínas Mitocondriales/metabolismo , Norepinefrina/farmacología , Fenoles/farmacología , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores beta de Hormona Tiroidea/agonistas , Receptores beta de Hormona Tiroidea/metabolismo , Triyodotironina/farmacología , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
8.
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
9.
J Clin Endocrinol Metab ; 89(7): 3117-28, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15240580

RESUMEN

Thyroid hormones are required for human brain development, but data on local regulation are limited. We describe the ontogenic changes in T(4), T(3), and rT(3) and in the activities of the types I, II, and III iodothyronine deiodinases (D1, D2, and D3) in different brain regions in normal fetuses (13-20 wk postmenstrual age) and premature infants (24-42 wk postmenstrual age). D1 activity was undetectable. The developmental changes in the concentrations of the iodothyronines and D2 and D3 activities showed spatial and temporal specificity but with divergence in the cerebral cortex and cerebellum. T(3) increased in the cortex between 13 and 20 wk to levels higher than adults, unexpected given the low circulating T(3). Considerable D2 activity was found in the cortex, which correlated positively with T(4) (r = 0.65). Cortex D3 activity was very low, as was D3 activity in germinal eminence and choroid plexus. In contrast, cerebellar T(3) was very low and increased only after midgestation. Cerebellum D3 activities were the highest (64 fmol/min.mg) of the regions studied, decreasing after midgestation. Other regions with high D3 activities (midbrain, basal ganglia, brain stem, spinal cord, hippocampus) also had low T(3) until D3 started decreasing after midgestation. D3 was correlated with T(3) (r = -0.682) and rT(3)/T(3) (r = 0.812) and rT(3)/T(4) (r = 0.889). Our data support the hypothesis that T(3) is required by the human cerebral cortex before midgestation, when mother is the only source of T(4). D2 and D3 play important roles in the local bioavailability of T(3). T(3) is produced from T(4) by D2, and D3 protects brain regions from excessive T(3) until differentiation is required.


Asunto(s)
Encéfalo/embriología , Recien Nacido Prematuro/metabolismo , Yoduro Peroxidasa/metabolismo , Tiroxina/metabolismo , Triyodotironina Inversa/metabolismo , Triyodotironina/metabolismo , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario y Fetal , Humanos , Recién Nacido , Concentración Osmolar , Distribución Tisular , Yodotironina Deyodinasa Tipo II
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA