Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
EMBO Rep ; 21(11): e50085, 2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33043581

RESUMEN

The cultured brown adipocytes can oxidize glucose in vitro, but it is still not fully clear whether brown adipose tissue (BAT) could completely oxidize glucose in vivo. Although positron emission tomography (PET) with 18 F-fluorodeoxyglucose (18 F-FDG) showed a high level of glucose uptake in the activated BAT, the non-metabolizable 18 F-FDG cannot fully demonstrate intracellular glucose metabolism. Through in vivo [U-13 C]glucose tracing, here we show that chronic cold exposure dramatically activates glucose oxidation in BAT and the browning/beiging subcutaneous white adipose tissue (sWAT). Specifically, chronic cold exposure enhances glucose flux into the mitochondrial TCA cycle. Metabolic flux analysis models that ß3-adrenergic receptor (ß3-AR) agonist significantly enhances the flux of mitochondrial pyruvate uptake through mitochondrial pyruvate carrier (MPC) in the differentiated primary brown adipocytes. Furthermore, in vivo MPC inhibition blocks cold-induced glucose oxidation and impairs body temperature maintenance in mice. Together, mitochondrial pyruvate uptake and oxidation serve an important energy source in the chronic cold exposure activated BAT and beige adipose tissue, which supports a role for glucose oxidation in brown fat thermogenesis.


Asunto(s)
Tejido Adiposo Pardo , Glucosa , Tejido Adiposo Blanco , Animales , Frío , Fluorodesoxiglucosa F18 , Ratones , Termogénesis
2.
Am J Pathol ; 188(1): 173-183, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29128567

RESUMEN

The neuropeptides arginine vasopressin (Avp) and vasoactive intestinal polypeptide (Vip) are critical for the communication and coupling of suprachiasmatic nucleus neurons, which organize daily rhythms of physiology and behavior in mammals. However, how these peptides are regulated remains uncharacterized. We found that heterogeneous nuclear ribonucleoprotein U (hnRNP U) is essential for the expression of Avp and Vip. Loss of one copy of the Hnrnpu gene resulted in fragmented locomotor activities and disrupted metabolic rhythms. Hnrnpu+/- mice were more active than wild-type mice in the daytime but more inactive at night. These phenotypes were partially rescued by microinfusion of Avp and Vip into free-moving animals. In addition, hnRNP U modulated Avp and Vip via directly binding to their promoters together with brain and muscle Arnt-like protein-1/circadian locomotor output cycles kaput heterodimers. Our work identifies hnRNP U as a novel regulator of the circadian pacemaker and provides new insights into the mechanism of rhythm output.


Asunto(s)
Ritmo Circadiano/genética , Ribonucleoproteína Heterogénea-Nuclear Grupo U/genética , Actividad Motora/genética , Animales , Arginina Vasopresina/genética , Arginina Vasopresina/metabolismo , Arginina Vasopresina/farmacología , Ritmo Circadiano/efectos de los fármacos , Femenino , Regulación de la Expresión Génica , Haploinsuficiencia , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Péptido Intestinal Vasoactivo/genética , Péptido Intestinal Vasoactivo/metabolismo , Péptido Intestinal Vasoactivo/farmacología
3.
Am J Pathol ; 186(8): 2117-2128, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27418549

RESUMEN

Duodenum-jejunum gastric bypass (DJB) has been used to treat morbid diabetic patients. However, neither the suitability among patients nor the mechanisms of this surgical treatment is clear. Previously, we reported a new mouse strain named Timo as type 2 diabetes model caused by brain-derived neurotrophic factor (Bdnf) deficiency. In this study, we found that DJB on Timo mice reversed their metabolic abnormalities without altering the expression of Bdnf. Glucose tolerance and insulin sensitivity were improved greatly, along with reduction of fat accumulation in liver and white adipose tissue. The gut flora population was altered by DJB with increased proportion of Firmicutes and decreased Actinobacteria and Proteobacteria in the ileum after surgery. Systemic inflammation in Timo mice was greatly suppressed with less macrophage infiltration and lower tumor necrosis factor-α levels in liver and white adipose tissue after surgery. Interestingly, the alteration of gut microflora abundance and improved metabolism preceded the inflammation alleviation after DJB surgery. These results suggested that DJB can reverse Bdnf deficiency-associated metabolic abnormality. In addition, the reduced inflammation may not be the initial cause for the DJB-associated metabolic and microbiota alterations. The increased BDNF protein levels in hypothalamus and hippocampus may result from microbiota change after DJB surgery.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/deficiencia , Diabetes Mellitus Experimental/cirugía , Diabetes Mellitus Tipo 2/cirugía , Derivación Gástrica , Microbioma Gastrointestinal , Animales , Western Blotting , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa
4.
Biochem J ; 468(2): 325-36, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25825937

RESUMEN

Gasdermin A3 (Gsdma3) was originally identified in association with hair-loss phenotype in mouse mutants. Our previous study found that AE mutant mice, with a Y344H substitution at the C-terminal domain of Gsdma3, display inflammation-dependent alopecia and excoriation [Zhou et al. (2012) Am. J. Pathol. 180, 763-774]. Interestingly, we found that the newly-generated null mutant of Gsdma3 mice did not display the skin dysmorphology, indicating that Gsdma3 is not essential for differentiation of epidermal cells and maintenance of the hair cycle in normal physiological conditions. Consistently, human embryonic kidney (HEK)293 and HaCaT cells transfected with wild-type (WT) Gsdma3 did not show abnormal morphology. However, Gsdma3 Y344H mutation induced autophagy. Gsdma3 N-terminal domain, but not the C-terminal domain, also displayed the similar pro-autophagic activity. The Gsdma3 Y344H mutant protein and N-terminal domain-induced autophagy was associated with mitochondria and ROS generation. Co-expression of C-terminal domain reversed the cell autophagy induced by N-terminal domain. Moreover, C-terminal domain could be co-precipitated with N-terminal domain. These data indicated that the potential pro-autophagic activity of WT Gsdma3 protein is suppressed through an intramolecular inhibition mechanism. Studies on other members of the GSDM family suggested this mechanism is conserved in several sub-families.


Asunto(s)
Autofagia , Muerte Celular , Mutación/genética , Proteínas/fisiología , Animales , Western Blotting , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias , Fenotipo , Especies Reactivas de Oxígeno
5.
Nat Metab ; 6(1): 78-93, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38191667

RESUMEN

The coexistence of brown adipocytes with low and high thermogenic activity is a fundamental feature of brown adipose tissue heterogeneity and plasticity. However, the mechanisms that govern thermogenic adipocyte heterogeneity and its significance in obesity and metabolic disease remain poorly understood. Here we show that in male mice, a population of transcription factor jun-B (JunB)-enriched (JunB+) adipocytes within the brown adipose tissue exhibits lower thermogenic capacity compared to high-thermogenic adipocytes. The JunB+ adipocyte population expands in obesity. Depletion of JunB in adipocytes increases the fraction of adipocytes exhibiting high thermogenic capacity, leading to enhanced basal and cold-induced energy expenditure and protection against diet-induced obesity and insulin resistance. Mechanistically, JunB antagonizes the stimulatory effects of PPARγ coactivator-1α on high-thermogenic adipocyte formation by directly binding to the promoter of oestrogen-related receptor alpha, a PPARγ coactivator-1α downstream effector. Taken together, our study uncovers that JunB shapes thermogenic adipocyte heterogeneity, serving a critical role in maintaining systemic metabolic health.


Asunto(s)
Resistencia a la Insulina , Ratones , Masculino , Animales , PPAR gamma/metabolismo , Adipocitos Marrones/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Dieta Alta en Grasa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
6.
Elife ; 122023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37417552

RESUMEN

The production of beige adipocytes following cold exposure is blocked as mice get older and leads to changes in the expression of metabolic genes.


Asunto(s)
Adipocitos Beige , Ratones , Animales , Proteína Desacopladora 1/metabolismo , Termogénesis , Frío
7.
Compr Physiol ; 12(4): 4119-4132, 2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36214190

RESUMEN

As a dynamic endocrine organ, white adipose tissue (WAT) stores lipids and plays a critical role in maintaining whole-body energy homeostasis and insulin sensitivity. A large group of the population over 65 years old suffer from increased WAT mass, especially in the visceral location. Visceral adiposity accelerates aging through promoting age-associated chronic conditions, significantly shortening life expectancy. Unlike WAT, brown adipose tissue (BAT) functions as an effective energy sink that burns and disposes of excess lipids and glucose upon activation of thermogenesis. Unfortunately, the thermogenic activity of BAT declines during aging. New appreciation of cellular and functional remodeling of WAT and BAT during aging has emerged in recent years. Efforts are underway to explore the potential underlying mechanisms behind these age-associated alterations in WAT and BAT and the impact of these alterations on whole-body metabolism. Lastly, it is intriguing to translate our knowledge obtained from animal models to the clinic to prevent and treat age-associated metabolic disorders. © 2022 American Physiological Society. Compr Physiol 12: 4119-4132, 2022.


Asunto(s)
Metabolismo Energético , Termogénesis , Tejido Adiposo Pardo/metabolismo , Envejecimiento , Animales , Glucosa/metabolismo , Lípidos
8.
Biology (Basel) ; 11(7)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-36101404

RESUMEN

Breastfeeding offers a broad spectrum of health benefits for infants. However, overnutrition and a steady increase in maternal obesity in the U.S. have made it harder for many mothers to produce and express breastmilk, and the quality of milk from obese mothers is also frequently compromised. Adipocytes, the primary cell type in the non-lactating breast, display a drastic morphological and functional change during lactation in mice. Lipid-filled adipocytes undergo lipolysis, and lipid droplets disappear to provide fatty acids and energy for breastmilk production. Once the animal stops lactation, these lipid-depleted adipocytes return as lipid-laden cells. This dynamic remodeling of the tissue is likely the result of active intercellular communications. Connexin43 (Cx43) is the most abundant connexin in the mammary adipose tissue that makes up the gap junctions for direct intercellular communications. Its expression is increased during lactation and reduced in obese mammary adipose tissue, which is resistant to lactation-induced remodeling. However, whether Cx43 is required for adipocyte remodeling and breastmilk production to support neonates' growth has not been established. In this study, we used doxycycline-inducible adipocyte-specific Cx43-deleted mice and demonstrated that adipocyte Cx43 played a vital role in determining the carbohydrate levels in breastmilk, which may subsequently affect neonates' growth.

9.
Front Endocrinol (Lausanne) ; 12: 651763, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33953697

RESUMEN

Brown adipocyte in brown adipose tissue (BAT) specializes in expending energy through non-shivering thermogenesis, a process that produces heat either by uncoupling protein 1 (UCP1) dependent uncoupling of mitochondrial respiration or by UCP1 independent mechanisms. Apart from this, there is ample evidence suggesting that BAT has an endocrine function. Studies in rodents point toward its vital roles in glucose and lipid homeostasis, making it an important therapeutic target for treating metabolic disorders related to morbidities such as obesity and type 2 diabetes. The rediscovery of thermogenically active BAT depots in humans by several independent research groups in the last decade has revitalized interest in BAT as an even more promising therapeutic intervention. Over the last few years, there has been overwhelming interest in understanding brown adipocyte's developmental lineages and how brown adipocyte uniquely utilizes energy beyond UCP1 mediated uncoupling respiration. These new discoveries would be leveraged for designing novel therapeutic interventions for metabolic disorders.


Asunto(s)
Tejido Adiposo Pardo/patología , Metabolismo Energético , Obesidad/metabolismo , Proteína Desacopladora 1/biosíntesis , Adipocitos/citología , Adipocitos Marrones/metabolismo , Animales , Sistema Endocrino , Ácidos Grasos/metabolismo , Homeostasis , Humanos , Enfermedades Metabólicas/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Termogénesis
10.
J Clin Invest ; 130(1): 247-257, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31573981

RESUMEN

Brown adipose tissue (BAT), as the main site of adaptive thermogenesis, exerts beneficial metabolic effects on obesity and insulin resistance. BAT has been previously assumed to contain a homogeneous population of brown adipocytes. Utilizing multiple mouse models capable of genetically labeling different cellular populations, as well as single-cell RNA sequencing and 3D tissue profiling, we discovered a brown adipocyte subpopulation with low thermogenic activity coexisting with the classical high-thermogenic brown adipocytes within the BAT. Compared with the high-thermogenic brown adipocytes, these low-thermogenic brown adipocytes had substantially lower Ucp1 and Adipoq expression, larger lipid droplets, and lower mitochondrial content. Functional analyses showed that, unlike the high-thermogenic brown adipocytes, the low-thermogenic brown adipocytes have markedly lower basal mitochondrial respiration, and they are specialized in fatty acid uptake. Upon changes in environmental temperature, the 2 brown adipocyte subpopulations underwent dynamic interconversions. Cold exposure converted low-thermogenic brown adipocytes into high-thermogenic cells. A thermoneutral environment had the opposite effect. The recruitment of high-thermogenic brown adipocytes by cold stimulation is not affected by high-fat diet feeding, but it does substantially decline with age. Our results revealed a high degree of functional heterogeneity of brown adipocytes.


Asunto(s)
Adipocitos Marrones/metabolismo , Adiponectina/biosíntesis , Tejido Adiposo Pardo/metabolismo , Regulación de la Expresión Génica/fisiología , Termogénesis/fisiología , Proteína Desacopladora 1/biosíntesis , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Animales , Ratones
11.
Diabetes ; 68(10): 1874-1885, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31540940

RESUMEN

Activated beige adipocytes have therapeutic potential due to their ability to improve glucose and lipid homeostasis. To date, the origin of beige adipocytes remains enigmatic. Whether beige cells arise through de novo differentiation from resident precursors or through reprogramming of mature white adipocytes has been a topic of intense discussion. Here, we offer our perspective on the natural origin of beige adipocytes in mice. In particular, we revisit recent lineage-tracing studies that shed light on this issue and offer new insight into how environmental housing temperatures early in life influence the mode of beige adipocyte biogenesis upon cold exposure later in life. We suggest a unified model in which beige adipocytes (UCP1+ multilocular cells) in rodents initially arise predominantly from progenitors (i.e., de novo beige adipogenesis) upon the first exposure to cold temperatures and then interconvert between "dormant beige" and "active beige" phenotypes (i.e., beige cell activation) upon subsequent changes in environmental temperature. Importantly, we highlight experimental considerations needed to visualize de novo adipogenesis versus beige cell activation in mice. A precise understanding of the cellular origins of beige adipocytes emanating in response to physiological and pharmacological stimuli may better inform therapeutic strategies to recruit beige adipocytes in vivo.


Asunto(s)
Adipocitos Beige/citología , Adipogénesis/fisiología , Tejido Adiposo Blanco/citología , Animales , Humanos , Termogénesis/fisiología
12.
Stem Cell Res Ther ; 9(1): 47, 2018 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-29482657

RESUMEN

BACKGROUND: Changes in metabolic pathway preferences are key events in the reprogramming process of somatic cells to induced pluripotent stem cells (iPSCs). The optimization of metabolic conditions can enhance reprogramming; however, the detailed underlying mechanisms are largely unclear. By comparing the gene expression profiles of somatic cells, intermediate-phase cells, and iPSCs, we found that carnitine palmitoyltransferase (Cpt)1b, a rate-limiting enzyme in fatty acid oxidation, was significantly upregulated in the early stage of the reprogramming process. METHODS: Mouse embryonic fibroblasts isolated from transgenic mice carrying doxycycline (Dox)-inducible Yamanaka factor constructs were used for reprogramming. Various fatty acid oxidation-related metabolites were added during the reprogramming process. Colony counting and fluorescence-activated cell sorting (FACS) were used to calculate reprogramming efficiency. Fatty acid oxidation-related metabolites were measured by liquid chromatography-mass spectrometry. Seahorse was used to measure the level of oxidative phosphorylation. RESULTS: We found that overexpression of cpt1b enhanced reprogramming efficiency. Furthermore, palmitoylcarnitine or acetyl-CoA, the primary and final products of Cpt1-mediated fatty acid oxidation, also promoted reprogramming. In the early reprogramming process, fatty acid oxidation upregulated oxidative phosphorylation and downregulated protein kinase C activity. Inhibition of protein kinase C also promoted reprogramming. CONCLUSION: We demonstrated that fatty acid oxidation promotes reprogramming by enhancing oxidative phosphorylation and inhibiting protein kinase C activity in the early stage of the reprogramming process. This study reveals that fatty acid oxidation is crucial for the reprogramming efficiency.


Asunto(s)
Reprogramación Celular , Embrión de Mamíferos/metabolismo , Ácidos Grasos/metabolismo , Fibroblastos/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Fosforilación Oxidativa , Proteína Quinasa C/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Embrión de Mamíferos/citología , Fibroblastos/citología , Células Madre Pluripotentes Inducidas/citología , Ratones , Oxidación-Reducción , Proteína Quinasa C/antagonistas & inhibidores
13.
J Endocrinol ; 239(2): 167-180, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30121577

RESUMEN

Peroxisome proliferator-activated receptor-γ (PPARγ) is a master regulator of adipogenesis and a target of the thiazolidinedione (TZD) class of antidiabetic drugs; therefore, identifying novel regulators of PPARγ action in adipocytes is essential for the future development of therapeutics for diabetes. MAGE family member D1 (MAGED1), by acting as an adaptor for ubiquitin-dependent degradation pathways and a co-factor for transcription, plays an important role in neural development, cell differentiation and circadian rhythm. Here, we showed that MAGED1 expression was downregulated during adipogenesis and loss of MAGED1 promoted preadipocyte proliferation and differentiation in vitro. MAGED1 bound to PPARγ and suppressed the stability and transcriptional activity of PPARγ. Compared to WT littermates, MAGED1-deficient mice showed increased levels of PPARγ protein and its target genes, more CD29+CD34+Sca-1+ adipocyte precursors and hyperplasia of white adipose tissues (WATs). Moreover, MAGED1-deficient mice developed late-onset obesity as a result of decreased energy expenditure and physical activity. However, these mice were metabolically healthy as shown by improved glucose clearance and insulin sensitivity, normal levels of serum lipids and enhanced secretion of adipokines such as leptin and adiponectin. Taken together, our data identify MAGED1 as a novel negative regulator of PPARγ activity, adipogenesis and insulin sensitivity in mice. MAGED1 might therefore serve as a novel pharmaceutical target to treat obesity-associated insulin resistance.


Asunto(s)
Adipogénesis , Resistencia a la Insulina , Proteínas de Neoplasias/metabolismo , PPAR gamma/metabolismo , Adiposidad , Animales , Glucosa/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/etiología , Obesidad/metabolismo
14.
Stem Cell Res Ther ; 9(1): 111, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29673401

RESUMEN

The original article [1] mistakenly omitted a source of funding, and the authors would like to rectify this by acknowledging the additional support of the Natural Science Foundation in Jiangsu Province (BK20150687).

15.
Cell Mol Immunol ; 15(6): 630-639, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28626237

RESUMEN

Body weight regain often causes failure of obesity therapies while the underlying mechanism remains largely unknown. In this study, we report that immune cells, especially CD4+ T cells, mediate the 'memory' of previous obese status. In a weight gain-loss-regain model, we found that C57BL/6J mice with an obesity history showed a much faster rate of body weight regain. This obesity memory could last for at least 2 months after previously obese mice were kept at the same body weight as non-obese mice. Surprisingly, such obesity memory was abrogated by dexamethasone treatment, whereas immunodeficient Rag1-/- and H2A-/- mice failed to establish such memory. Rag1-/- mice repossessed the obesity memory when immune cells or CD4+ T cells isolated from previously obese mice were transferred. Furthermore, depletion of CD4+ T cells led to obesity memory ablation. Taken together, we conclude that CD4+ T cells mediate obesity memory and promote weight regain.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Obesidad/inmunología , Aumento de Peso/inmunología , Animales , Metabolismo Basal , Hiperfagia/complicaciones , Ratones Endogámicos C57BL , Termogénesis , Pérdida de Peso/inmunología
16.
Cell Metab ; 28(2): 282-288.e3, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-29909970

RESUMEN

Adipose tissue in the mammary gland undergoes dramatic remodeling during reproduction. Adipocytes are replaced by mammary alveolar structures during pregnancy and lactation, then reappear upon weaning. The fate of the original adipocytes during lactation and the developmental origin of the re-appearing adipocyte post involution are unclear. Here, we reveal that adipocytes in the mammary gland de-differentiate into Pdgfrα+ preadipocyte- and fibroblast-like cells during pregnancy and remain de-differentiated during lactation. Upon weaning, de-differentiated fibroblasts proliferate and re-differentiate into adipocytes. This cycle occurs over multiple pregnancies. These observations reveal the potential of terminally differentiated adipocytes to undergo repeated cycles of de-differentiation and re-differentiation in a physiological setting.


Asunto(s)
Adipocitos Blancos/metabolismo , Adipogénesis , Tejido Adiposo , Lactancia/metabolismo , Glándulas Mamarias Animales , Adipocitos Blancos/citología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Femenino , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Destete
17.
Endocrinology ; 158(6): 1798-1811, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28323958

RESUMEN

The hypothalamic-pituitary-gonadal axis controls development, reproduction, and metabolism. Although most studies have focused on the hierarchy from the brain to the gonad, many questions remain unresolved concerning the feedback from the gonad to the central nervous system, especially regarding the potential epigenetic modifications in hypothalamic neurons. In the present report, we generated genetically modified mice lacking histone H3 lysine 27 (H3K27) demethylase Jumonji domain-containing 3 (JMJD3) in hypothalamic rat-insulin-promoter-expressing neurons (RIP-Cre neurons). The female mutant mice displayed late-onset obesity owing to reduced locomotor activity and decreased energy expenditure. JMJD3 deficiency in RIP-Cre neurons also results in delayed pubertal onset, an irregular estrous cycle, impaired fertility, and accelerated ovarian failure in female mice owing to the dysregulation of the hypothalamic-ovarian axis. We found that JMJD3 directly regulates Kiss1 gene expression by binding to the Kiss1 promoter and triggering H3K27me3 demethylation in the anteroventral periventricular (AVPV) nucleus. Further study confirmed that the aberrations arose from impaired kisspeptin signaling in the hypothalamic AVPV nucleus and subsequent estrogen deficiency. Estrogen replacement therapy can reverse obesity in mutant mice. Moreover, we demonstrated that Jmjd3 is an estrogen target gene in the hypothalamus. These results provide direct genetic and molecular evidence that JMJD3 is a key mediator for the kisspeptin-estrogen feedback loop.


Asunto(s)
Estrógenos/metabolismo , Retroalimentación Fisiológica/fisiología , Hipotálamo Anterior/metabolismo , Histona Demetilasas con Dominio de Jumonji/fisiología , Kisspeptinas/metabolismo , Neuronas/metabolismo , Reproducción/fisiología , Animales , Femenino , Insulina/genética , Integrasas/genética , Integrasas/metabolismo , Histona Demetilasas con Dominio de Jumonji/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Ratas , Reproducción/genética
18.
Onco Targets Ther ; 10: 4753-4763, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29033588

RESUMEN

Non-small cell lung cancer (NSCLC) is one of the most common malignancies worldwide, and it occurs at a higher frequency in males. HOXD-AS1, an important cancer-associated long noncoding RNA (lncRNA), contributes to the development and progression of several cancers. However, the exact roles of HOXD-AS1 in NSCLC progression are still unknown. Here, we investigated the underlying mechanisms of HOXD-AS1 in human NSCLC tissues. We found that lncRNA HOXD-AS1 was specifically upregulated (P<0.001) in NSCLC tissues and promoted cancer cell growth by targeting miR-147a. Moreover, HOXD-AS1 expression positively correlated with NSCLC clinical pathologic characteristics (tumor size, P=0.006; tumor stage, P=0.044; recurrence, P=0.031) and survival rate (P=0.003). HOXD-AS1 knockdown reduced proliferation and promoted apoptosis of NSCLC cells. The dual-luciferase reporter assay showed that HOXD-AS1 could negatively regulate the expression of miR-147a. miR-147a inhibition abrogated the effect of HOXD-AS1 knockdown on the proliferation and apoptosis of NSCLC cells. Furthermore, HOXD-AS1 positively regulated the expression of pRB (a tumor suppressor protein) in NSCLC cells. Taken together, our data indicated that HOXD-AS1 might be an oncogenic lncRNA that promotes proliferation of NSCLC and could be a therapeutic target in NSCLC.

19.
Cell Cycle ; 15(11): 1450-61, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27096707

RESUMEN

Studies using in vitro cultured oocytes have indicated that the protein phosphatase 2A (PP2A), a major serine/threonine protein phosphatase, participates in multiple steps of meiosis. Details of oocyte maturation regulation by PP2A remain unclear and an in vivo model can provide more convincing information. Here, we inactivated PP2A by mutating genes encoding for its catalytic subunits (PP2Acs) in mouse oocytes. We found that eliminating both PP2Acs caused female infertility. Oocytes lacking PP2Acs failed to complete 1(st) meiotic division due to chromosome misalignment and abnormal spindle assembly. In mitosis, PP2A counteracts Aurora kinase B/C (AurkB/C) to facilitate correct kinetochore-microtubule (KT-MT) attachment. In meiosis I in oocyte, we found that PP2Ac deficiency destabilized KT-MT attachments. Chemical inhibition of AurkB/C in PP2Ac-null oocytes partly restored the formation of lateral/merotelic KT-MT attachments but not correct KT-MT attachments. Taken together, our findings demonstrate that PP2Acs are essential for chromosome alignments and regulate the formation of correct KT-MT attachments in meiosis I in oocytes.


Asunto(s)
Cinetocoros/metabolismo , Meiosis , Microtúbulos/metabolismo , Oocitos/metabolismo , Proteína Fosfatasa 2/genética , Subunidades de Proteína/genética , Secuencia de Aminoácidos , Animales , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Aurora Quinasa C/genética , Aurora Quinasa C/metabolismo , Cromosomas de los Mamíferos/metabolismo , Cromosomas de los Mamíferos/ultraestructura , Femenino , Regulación del Desarrollo de la Expresión Génica , Infertilidad Femenina/genética , Infertilidad Femenina/metabolismo , Cinetocoros/ultraestructura , Ratones , Ratones Transgénicos , Microtúbulos/ultraestructura , Mitosis , Oocitos/ultraestructura , Cultivo Primario de Células , Proteína Fosfatasa 2/metabolismo , Subunidades de Proteína/metabolismo , Transducción de Señal , Huso Acromático/metabolismo , Huso Acromático/ultraestructura
20.
Aging (Albany NY) ; 7(4): 223-32, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25888638

RESUMEN

Protein phosphatase 2A (PP2A) is a key negative regulator of phosphatidylinositol 3-kinase/Akt pathway. Previous study showed that, in the liver, the catalytic subunit of PP2A (PP2Ac) is closely associated with insulin resistance syndrome, which is characterized by glucose intolerance and dyslipidemia. Here we studied the role of liver PP2Ac in glucose metabolism and evaluated whether PP2Ac is a suitable therapeutic target for treating insulin resistance syndrome. Liver-specific Ppp2cα knockout mice (Ppp2cα(loxp/loxp): Alb) exhibited improved glucose homeostasis compared with littermate controls in both normal and high-fat diet conditions, despite no significant changes in body weight and liver weight under chow diet. Ppp2cα(loxp/loxp): Alb mice showed enhanced glycogen deposition, serum triglyceride, cholesterol, low density lipoprotein and high density lipoprotein, activated insulin signaling, decreased expressions of gluconeogenic genes G6P and PEPCK, and lower liver triglyceride. Liver-specific Ppp2cα knockout mice showed enhanced glucose homeostasis and increased insulin sensitivity by activation of insulin signaling through Akt. These findings suggest that inhibition of hepatic Ppp2cα may be a useful strategy for the treatment of insulin resistance syndrome.


Asunto(s)
Intolerancia a la Glucosa/metabolismo , Glucosa/metabolismo , Resistencia a la Insulina/fisiología , Insulina/metabolismo , Hígado/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Dieta Alta en Grasa , Homeostasis/fisiología , Lípidos/sangre , Ratones , Ratones Noqueados , Fosfoproteínas Fosfatasas/genética , Proteína Fosfatasa 2C , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA