Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 151(2): 248-50, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-23063118

RESUMO

Developmental genes are essential in the formation and function of adipose tissue and muscle. In this issue of Cell, Teperino et al. demonstrate that noncanonical hedgehog signaling increases glucose uptake into brown fat and muscle. Modulation of developmental pathways may serve as a potential target for new treatments of diabetes and other metabolic disorders.

2.
EMBO J ; 38(3)2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30530479

RESUMO

Recent studies suggest that, even within a single adipose depot, there may be distinct subpopulations of adipocytes. To investigate this cellular heterogeneity, we have developed multiple conditionally immortalized clonal preadipocyte lines from white adipose tissue of mice. Analysis of these clones reveals at least three white adipocyte subpopulations. These subpopulations have differences in metabolism and differentially respond to inflammatory cytokines, insulin, and growth hormones. These also have distinct gene expression profiles and can be tracked by differential expression of three marker genes: Wilms' tumor 1, transgelin, and myxovirus 1. Lineage tracing analysis with dual-fluorescent reporter mice indicates that these adipocyte subpopulations have differences in gene expression and metabolism that mirror those observed in the clonal cell lines. Furthermore, preadipocytes and adipocytes from these subpopulations differ in their abundance in different fat depots. Thus, white adipose tissue, even in a single depot, is comprised of distinct subpopulations of white adipocytes with different physiological phenotypes. These differences in adipocyte composition may contribute to the differences in metabolic behavior and physiology of different fat depots.


Assuntos
Adipócitos Brancos/classificação , Adipócitos Brancos/citologia , Adipogenia , Tecido Adiposo/citologia , Biomarcadores/análise , Adipócitos Brancos/fisiologia , Tecido Adiposo/fisiologia , Animais , Citocinas/metabolismo , Metabolismo Energético , Hormônio do Crescimento Humano/metabolismo , Mediadores da Inflamação/metabolismo , Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/metabolismo , Proteínas Repressoras/metabolismo , Transcriptoma , Proteínas WT1
3.
Am J Physiol Endocrinol Metab ; 316(1): E34-E42, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30325658

RESUMO

The lipolytic effects of growth hormone (GH) have been known for half a century and play an important physiological role for substrate metabolism during fasting. In addition, sustained GH-induced lipolysis is causally linked to insulin resistance. However, the underlying molecular mechanisms remain elusive. In the present study, we obtained experimental data in human subjects and used human adipose-derived stromal vascular cells (hADSCs) as a model system to elucidate GH-triggered molecular signaling that stimulates adipose tissue lipolysis and insulin resistance in human adipocytes. We discovered that GH downregulates the expression of fat-specific protein (FSP27), a negative regulator of lipolysis, by impairing the transcriptional ability of the master transcriptional regulator, peroxisome proliferator-activated receptor-γ (PPARγ) via MEK/ERK activation. Ultimately, GH treatment promotes phosphorylation of PPARγ at Ser273 and causes its translocation from nucleus to the cytosol. Surprisingly, FSP27 overexpression inhibited PPARγ Ser273 phosphorylation and promoted its nuclear retention. GH antagonist treatment had similar effects. Our study identifies a novel signaling mechanism by which GH transcriptionally induces lipolysis via the MEK/ERK pathway that acts along PPARγ-FSP27 in human adipose tissue.


Assuntos
Adipócitos Brancos/metabolismo , Hormônio do Crescimento Humano/metabolismo , Lipólise/genética , Sistema de Sinalização das MAP Quinases , PPAR gama/metabolismo , Proteínas/genética , Proteínas Reguladoras de Apoptose , Regulação da Expressão Gênica , Humanos , Técnicas In Vitro , Masculino , Fosforilação , Proteínas/metabolismo , Adulto Jovem
4.
Proc Natl Acad Sci U S A ; 110(28): 11409-14, 2013 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-23798383

RESUMO

Visceral and s.c. fat exhibit different intrinsic properties, including rates of lipolysis, and are associated with differential risk for the development of type 2 diabetes. These effects are in part related to cell autonomous differences in gene expression. In the present study, we show that expression of Shox2 (Short stature homeobox 2) is higher in s.c. than visceral fat in both rodents and humans and that levels are further increased in humans with visceral obesity. Fat-specific disruption of Shox2 in male mice results in protection from high fat diet-induced obesity, with a preferential loss of s.c. fat. The reduced adipocyte size is secondary to a twofold increase in the expression of ß3 adrenergic receptor (Adrb3) at both the mRNA and protein level and a parallel increase in lipolytic rate. These effects are mimicked by knockdown of Shox2 in C3H10T1/2 cells. Conversely, overexpression of Shox2 leads to a repression of Adrb3 expression and decrease lipolytic rate. Shox2 does not affect differentiation but directly interacts with CCAAT/enhancer binding protein alpha and attenuates its transcriptional activity of the Adrb3 promoter. Thus, Shox2 can regulate the expression of Adrb3 and control the rate of lipolysis and, in this way, exerts control of the phenotypic differences between visceral and s.c. adipocytes.


Assuntos
Adipócitos/citologia , Proteínas de Homeodomínio/fisiologia , Animais , Dieta , Proteínas de Homeodomínio/genética , Resistência à Insulina , Lipólise , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/genética
5.
Proc Natl Acad Sci U S A ; 108(7): 2771-6, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21282637

RESUMO

Increased intraabdominal (visceral) fat is associated with a high risk of diabetes and metabolic syndrome. We have previously shown that the mesodermal developmental transcription factor Tbx15 is highly differentially expressed between visceral and subcutaneous (s.c.) fat in both humans and rodents, and in humans visceral fat Tbx15 expression is decreased in obesity. Here we show that, in mice, Tbx15 is 260-fold more highly expressed in s.c. preadipocytes than in epididymal preadipocytes. Overexpression of Tbx15 in 3T3-L1 preadipocytes impairs adipocyte differentiation and decreases triglyceride content. This defect in differentiation can be corrected by stimulating cells with the PPARγ agonist rosiglitazone (Rosi). However, triglyceride accumulation remains decreased by ∼50%, due to a decrease in basal lipogenic rate and increase in basal lipolytic rate. 3T3-L1 preadipocytes overexpressing Tbx15 also have a 15% reduction in mitochondrial mass and a 28% reduction in basal mitochondrial respiration (P = 0.004) and ATP turnover (P = 0.02), and a 45% (P = 0.003) reduction in mitochondrial respiratory capacity. Thus, differential expression of Tbx15 between fat depots plays an important role in the interdepot differences in adipocyte differentiation, triglyceride accumulation, and mitochondrial function that may contribute to the risk of diabetes and metabolic disease.


Assuntos
Adipócitos/fisiologia , Diferenciação Celular/genética , Respiração Celular/genética , Mitocôndrias/fisiologia , Gordura Subcutânea/metabolismo , Proteínas com Domínio T/metabolismo , Células 3T3-L1 , Trifosfato de Adenosina/metabolismo , Animais , Compostos Azo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Respiração Celular/fisiologia , Clonagem Molecular , Primers do DNA/genética , Metabolismo Energético/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Consumo de Oxigênio/fisiologia , PPAR gama/agonistas , Reação em Cadeia da Polimerase , Rosiglitazona , Proteínas com Domínio T/genética , Tiazolidinedionas/farmacologia
6.
Proc Natl Acad Sci U S A ; 108(35): 14608-13, 2011 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-21873205

RESUMO

Sirt3 is a member of the sirtuin family of protein deacetylases that is localized in mitochondria and regulates mitochondrial function. Sirt3 expression in skeletal muscle is decreased in models of type 1 and type 2 diabetes and regulated by feeding, fasting, and caloric restriction. Sirt3 knockout mice exhibit decreased oxygen consumption and develop oxidative stress in skeletal muscle, leading to JNK activation and impaired insulin signaling. This effect is mimicked by knockdown of Sirt3 in cultured myoblasts, which exhibit reduced mitochondrial oxidation, increased reactive oxygen species, activation of JNK, increased serine and decreased tyrosine phosphorylation of IRS-1, and decreased insulin signaling. Thus, Sirt3 plays an important role in diabetes through regulation of mitochondrial oxidation, reactive oxygen species production, and insulin resistance in skeletal muscle.


Assuntos
Resistência à Insulina , Mitocôndrias/metabolismo , Músculo Esquelético/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Sirtuína 3/fisiologia , Envelhecimento/metabolismo , Animais , Células Cultivadas , Diabetes Mellitus Experimental/metabolismo , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mioblastos/metabolismo , Oxirredução , Fosforilação
7.
J Endocrinol ; 260(3)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38174979

RESUMO

Studies in humans and mice have determined that distinct subpopulations of adipocytes reside even within individual adipose tissue depots. Previously, our lab defined three white adipocyte subpopulations with stable and unique gene expression profiles, which were termed type 1, 2, and 3 adipocytes, respectively. Our previous studies demonstrated that type 2 adipocytes were highly responsive to the inflammatory cytokine, tumor necrosis factor alpha (TNFα). This study extends these findings to investigate the role of type 2 adipocytes in obesity. We found that treatment with TNFα increased lipolysis specifically in type 2 adipocytes, at least in part, through the reduction of fat-specific protein 27 (FSP27) expression. To assess the physiological role of lipolysis from this adipocyte subpopulation, a type2Ad-hFSP27tg mouse model was generated by overexpressing human FSP27 specifically in type 2 adipocytes. Glucose and insulin tolerance test analysis showed that male type2Ad-hFSP27tg mice on 60% high-fat diet exhibited improved glucose tolerance and insulin sensitivity, with no change in body weight compared to controls. These metabolic changes may, at least in part, be explained by the reduced lipolysis rate in the visceral fat of type2Ad-hFSP27tg mice. Although FSP27 overexpression in primary type 2 adipocytes was sufficient to acutely reduce TNFα-induced apoptosis in vitro, it failed to reduce macrophage infiltration in obesity in vivo. Taken together, these results strongly suggest that type 2 adipocytes contribute to the regulation of lipolysis and could serve as a potential therapeutic target for obesity-associated insulin resistance.


Assuntos
Resistência à Insulina , Lipólise , Masculino , Camundongos , Humanos , Animais , Lipólise/genética , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Adipócitos/metabolismo , Obesidade/genética , Obesidade/metabolismo , Dieta Hiperlipídica/efeitos adversos , Glucose/metabolismo , Camundongos Endogâmicos C57BL
9.
ACS Catal ; 13(12): 7812-7821, 2023 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-37342831

RESUMO

Electrochemical conversion of CO2 requires selective catalysts and high solubility of CO2 in the electrolyte to reduce the energy requirement and increase the current efficiency. In this study, the CO2 reduction reaction (CO2RR) over Ag electrodes in acetonitrile-based electrolytes containing 0.1 M [EMIM][2-CNpyr] (1-ethyl-3-methylimidazolium 2-cyanopyrolide), a reactive ionic liquid (IL), is shown to selectively (>94%) convert CO2 to CO with a stable current density (6 mA·cm-2) for at least 12 h. The linear sweep voltammetry experiments show the onset potential of CO2 reduction in acetonitrile shifts positively by 240 mV when [EMIM][2-CNpyr] is added. This is attributed to the pre-activation of CO2 through the carboxylate formation via the carbene intermediate of the [EMIM]+ cation and the carbamate formation via binding to the nucleophilic [2-CNpyr]- anion. The analysis of the electrode-electrolyte interface by surface-enhanced Raman spectroscopy (SERS) confirms the catalytic role of the functionalized IL where the accumulation of the IL-CO2 adduct between -1.7 and -2.3 V vs Ag/Ag+ and the simultaneous CO formation are captured. This study reveals the electrode surface species and the role of the functionalized ions in lowering the energy requirement of CO2RR for the design of multifunctional electrolytes for the integrated capture and conversion.

10.
Endocrinology ; 164(11)2023 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-37897489

RESUMO

In adipose tissue, growth hormone (GH) stimulates lipolysis, leading to an increase in plasma free fatty acid levels and a reduction in insulin sensitivity. In our previous studies, we have found that GH increases lipolysis by reducing peroxisome proliferator-activated receptor γ (PPARγ) transcription activity, leading to a reduction of tat-specific protein 27 (FSP27, also known as CIDEC) expression. In previous studies, our laboratory uncovered 3 developmentally distinct subpopulations of white adipocytes. In this manuscript, we show that one of the subpopulations, termed type 2 adipocytes, has increased GH-induced signaling and lipolysis compared to other adipocyte subtypes. To assess the physiological role of GH-mediated lipolysis mediated by this adipocyte subpopulation, we specifically expressed human FSP27 (hFSP27) transgene in type 2 adipocytes (type2Ad-hFSP27tg mice). Systemically, male type2Ad-hFSP27tg mice displayed reduced serum glycerol release and nonesterified fatty acids levels after acute GH treatment, and improvement in acute, but not chronic, GH-induced glucose intolerance. Furthermore, we demonstrate that type2Ad-hFSP27tg mice displayed improved hepatic insulin signaling. Taken together, these results indicate that this adipocyte subpopulation is a critical regulator of the GH-mediated lipolytic and metabolic response. Thus, further investigation of adipocyte subpopulations may provide novel treatment strategies to regulate GH-induced glucose intolerance in patients with growth and metabolic disorders.


Assuntos
Intolerância à Glucose , Hormônio do Crescimento Humano , Humanos , Masculino , Camundongos , Animais , Hormônio do Crescimento/metabolismo , Lipólise/genética , Intolerância à Glucose/genética , Hormônio do Crescimento Humano/farmacologia , Hormônio do Crescimento Humano/metabolismo , Adipócitos Brancos/metabolismo , Glucose
11.
FASEB J ; 25(4): 1176-87, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21163860

RESUMO

WNT4, a member of the Wnt family of ligands, is critical for the development of the female reproductive tract. Analysis of Wnt4 expression in the adult uterus during pregnancy indicates that it may play a role in the regulation of endometrial stromal cell proliferation, survival, and differentiation, which is required to support the developing embryo. To investigate the role of Wnt4 in adult uterine physiology, conditional ablation of Wnt4 using the PR(cre) mouse model was accomplished. Ablation of Wnt4 rendered female mice subfertile due to a defect in embryo implantation and subsequent defects in endometrial stromal cell survival, differentiation, and responsiveness to progesterone signaling. In addition to altered stromal cell function, the uteri of PR(cre/+)Wnt4(f/f) (Wnt4(d/d)) mice displayed altered epithelial differentiation characterized by a reduction in the number of uterine glands and the emergence of a p63-positive basal cell layer beneath the columnar luminal epithelial cells. The altered epithelial cell phenotype was further escalated by chronic estrogen treatment, which caused squamous cell metaplasia of the uterine epithelium in the Wnt4(d/d) mice. Thus, WNT4 is a critical regulator not only of proper postnatal uterine development, but also embryo implantation and decidualization.


Assuntos
Decídua/fisiologia , Útero/fisiologia , Proteínas Wnt/fisiologia , Animais , Apoptose/efeitos dos fármacos , Implantação do Embrião/fisiologia , Feminino , Camundongos , Gravidez , Progesterona/fisiologia , Transdução de Sinais/fisiologia , Útero/crescimento & desenvolvimento , Proteína Wnt4
12.
Proc Natl Acad Sci U S A ; 106(21): 8677-82, 2009 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-19439667

RESUMO

Normal endometrial function requires a balance of progesterone (P4) and estrogen (E2) effects. An imbalance caused by increased E2 action and/or decreased P4 action can result in abnormal endometrial proliferation and, ultimately, endometrial adenocarcinoma, the fourth most common cancer in women. We have identified mitogen-inducible gene 6 (Mig-6) as a downstream target of progesterone receptor (PR) and steroid receptor coactivator (SRC-1) action in the uterus. Here, we demonstrate that absence of Mig-6 in mice results in the inability of P4 to inhibit E2-induced uterine weight gain and E2-responsive target genes expression. At 5 months of age, the absence of Mig-6 results in endometrial hyperplasia. Ovariectomized Mig-6(d/d) mice exhibit this hyperplastic phenotype in the presence of E2 and P4 but not without ovarian hormone. Ovariectomized Mig-6(d/d) mice treated with E2 developed invasive endometrioid-type endometrial adenocarcinoma. Importantly, the observation that endometrial carcinomas from women have a significant reduction in MIG-6 expression provides compelling support for an important growth regulatory role for Mig-6 in the uterus of both humans and mice. This demonstrates the Mig-6 is a critical regulator of the response of the endometrium to E2 in regulating tissue homeostasis. Since Mig-6 is regulated by both PR and SRC-1, this identifies a PR, SRC-1, Mig-6 regulatory pathway that is critical in the suppression of endometrial cancer.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Hiperplasia Endometrial/metabolismo , Neoplasias do Endométrio/metabolismo , Estrogênios/metabolismo , Progesterona/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Adolescente , Adulto , Animais , Regulação para Baixo , Hiperplasia Endometrial/genética , Hiperplasia Endometrial/patologia , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/patologia , Feminino , Histona Acetiltransferases/deficiência , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Knockout , Coativador 1 de Receptor Nuclear , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor
13.
Genes (Basel) ; 13(5)2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35627243

RESUMO

Mutations in Dentin Sialophosphoprotein (DSPP) are known to cause, in order of increasing severity, dentin dysplasia type-II (DD-II), dentinogenesis imperfecta type-II (DGI-II), and dentinogenesis imperfecta type-III (DGI-III). DSPP mutations fall into two groups: a 5'-group that affects protein targeting and a 3'-group that shifts translation into the −1 reading frame. Using whole-exome sequence (WES) analyses and Single Molecule Real-Time (SMRT) sequencing, we identified disease-causing DSPP mutations in 12 families. Three of the mutations are novel: c.53T>C/p.(Val18Ala); c.3461delG/p.(Ser1154Metfs*160); and c.3700delA/p.(Ser1234Alafs*80). We propose genetic analysis start with WES analysis of proband DNA to identify mutations in COL1A1 and COL1A2 causing dominant forms of osteogenesis imperfecta, 5'-DSPP mutations, and 3'-DSPP frameshifts near the margins of the DSPP repeat region, and SMRT sequencing when the disease-causing mutation is not identified. After reviewing the literature and incorporating new information showing distinct differences in the cell pathology observed between knockin mice with 5'-Dspp or 3'-Dspp mutations, we propose a modified Shields Classification based upon the causative mutation rather than phenotypic severity such that patients identified with 5'-DSPP defects be diagnosed as DGI-III, while those with 3'-DSPP defects be diagnosed as DGI-II.


Assuntos
Dentinogênese Imperfeita , Animais , Dentinogênese Imperfeita/genética , Proteínas da Matriz Extracelular/genética , Humanos , Camundongos , Mutação , Linhagem , Fosfoproteínas/genética , Sialoglicoproteínas/genética
14.
Methods Protoc ; 4(2)2021 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-34199437

RESUMO

Organic solvent dibenzyl ether (DBE)-based protocols have been widely used in adipose tissue clearing. However, benzyl alcohol/benzyl benzoate (BABB)-based clearing has been shown to offer better transparency in other tissues. The addition of diphenyl ether (DPE) to BABB (BABB-D4) is often included to preserve fluorescent signals, but its effects on adipose tissue transparency and shrinkage have not been explored. Distinct adipocyte subpopulations contribute to its cellular composition and biological activity. Here, we compared clearing solvents to create an optimized clearing methodology for the study of adipocyte subpopulations. Adipose tissues were cleared with BABB, BABB-D4, and DBE, and post-clearing transparency and tissue shrinkage were measured. An optimized protocol, including BABB-D4 clearing, delipidation, and extensive immunofluorescence blocking steps, was created to examine the spatial distribution of Wt-1 positive progenitor-derived (Type-1) adipocytes in intact mesenteric fat. Both BABB and BABB-D4 lead to significantly increased tissue transparency with reduced tissue shrinkage compared to DBE-cleared adipose tissue. Type-1 adipocytes are found in a clustered distribution with predominant residence in fat associated with the ileum and colon. This paper details an optimized clearing methodology for adipose tissue with increased tissue transparency and reduced shrinkage, and therefore will be a useful tool for investigating adipose tissue biology.

15.
Biol Reprod ; 83(3): 396-403, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20484741

RESUMO

During embryonic development, Foxa2 is required for the formation of the node and notochord, and ablation of this gene results in defects in gastrulation, neural tube patterning, and gut morphogenesis. Foxa2 has been shown to be expressed specifically in the glandular epithelium of the murine uterus. To study the uterine function of Foxa2, this gene was conditionally ablated in the mouse uterus by crossing mice with floxed Foxa2 alleles, Foxa2(loxP/loxP), with the Pgr(cre) mouse model. Pgr(cre/+) Foxa2(loxP/loxP) mice showed significantly reduced fertility. Analysis of the uterus on Day 5.5 of pregnancy showed disrupted blastocyst implantation. Pgr(cre/+) Foxa2(loxP/loxP) mice also showed a severe impairment of the uterus to respond to the artificial induction of the decidual response. Morphological examination of the uteri of these mice showed a severe reduction in the number of endometrial glands. The loss of endometrial glands resulted in the reduction of leukemia inhibitory factor (Lif) expression. The lack of a decidual response could be partially rescued by an intrauterine injection of LIF before the initiation of the decidual response. This analysis demonstrates that Foxa2 regulates endometrial gland development and that mice with a loss of endometrial glands cannot support implantation in part due to the loss of LIF, which is a requisite for fertility in the mouse.


Assuntos
Implantação do Embrião/fisiologia , Endométrio/crescimento & desenvolvimento , Fator 3-beta Nuclear de Hepatócito/metabolismo , Placentação , Placentação/fisiologia , Análise de Variância , Animais , Implantação do Embrião/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Endométrio/metabolismo , Estradiol/farmacologia , Feminino , Fator 3-beta Nuclear de Hepatócito/genética , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Placenta/efeitos dos fármacos , Placenta/metabolismo , Placentação/efeitos dos fármacos , Gravidez , Progesterona/farmacologia , Pseudogravidez/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Biol Reprod ; 82(5): 991-9, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20130264

RESUMO

Previous work has identified Indian hedgehog (Ihh) as a major mediator of progesterone signaling during embryo implantation. Ihh acts through its downstream effector smoothened (Smo) to activate the GLI family of transcription factors. In order to gain a better understanding of Ihh action during embryo implantation, we expressed a Cre-recombinase-dependent constitutively activated SMO in the murine uterus using the Pgr(tm2(cre)Lyd) (PR(cre)) mouse model [Pgr(tm2(cre)Lyd+)Gt(ROSA)26Sor(tm1(Smo/EYFP)Amc)(+) (PR(cre/+)SmoM2(+))]. Female PR(cre/+)SmoM2(+) mice were infertile. They exhibited normal serum progesterone levels and normal ovulation, but their ova failed to be fertilized in vivo and their uterus failed to undergo the artificially induced decidual response. Examination of the PR(cre/+)SmoM2(+) uteri revealed numerous features such as uterine hypertrophy, the presence of a stratified luminal epithelial cell layer, a reduced number of uterine glands, and an endometrial stroma that had lost its normal morphologic characteristics. Microarray analysis of 3-mo-old PR(cre/+)SmoM2(+) uteri demonstrated a chondrocytic signature and confirmed that constitutive activation of PR(cre/+)SmoM2(+) increased extracellular matrix production. Thus, constitutive activation of Smo in the mouse uterus alters postnatal uterine differentiation which interferes with early pregnancy. These results provide new insight into the role of Hedgehog signaling during embryo implantation.


Assuntos
Implantação do Embrião/fisiologia , Infertilidade Feminina/metabolismo , Progesterona/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Útero/crescimento & desenvolvimento , Animais , Diferenciação Celular/fisiologia , Decídua/crescimento & desenvolvimento , Feminino , Regulação da Expressão Gênica/fisiologia , Proteínas Hedgehog/metabolismo , Metaboloma , Camundongos , Camundongos Mutantes , Gravidez , Análise Serial de Proteínas , Receptores de Progesterona/metabolismo , Transdução de Sinais/fisiologia , Receptor Smoothened , Útero/citologia , Útero/metabolismo
17.
Biol Reprod ; 82(4): 783-90, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20056671

RESUMO

Conditional ablation of Indian hedgehog (Ihh) in the murine uterus results in mice that are sterile because of defects in embryo implantation. We performed microarray analysis on these mice at the time point at which the Ihh target genes are induced by the administration of exogenous hormone to mimic Day 3.5 of pregnancy. This analysis identified 863 genes altered by the conditional ablation of Ihh. Of these, genes that regulated the cell cycle were overrepresented. In addition, genes involved in epidermal growth factor (EGF) and estrogen (E2) signaling were found to be deregulated upon Ihh ablation. Furthermore, upon conditional ablation of Ihh, 15-mo-old mice exhibited hallmarks of estrogenized uteri, such as cystically dilated glands and hyalinized stroma. Thus, Ihh regulates embryo implantation by having an impact on the cell cycle, EGF signaling, and E2 signaling.


Assuntos
Ciclo Celular/genética , Fator de Crescimento Epidérmico/metabolismo , Estradiol/metabolismo , Deleção de Genes , Proteínas Hedgehog/genética , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Implantação do Embrião/genética , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/fisiologia , Estradiol/farmacologia , Estradiol/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Genes cdc/efeitos dos fármacos , Genes cdc/fisiologia , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/fisiologia , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Gravidez , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Útero/efeitos dos fármacos , Útero/metabolismo , Útero/fisiologia
18.
Mol Cell Biol ; 27(15): 5468-78, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17515606

RESUMO

The process of implantation, necessary for all viviparous birth, consists of tightly regulated events, including apposition of the blastocyst, attachment to the uterine lumen, and differentiation of the uterine stroma. In rodents and primates the uterine stroma undergoes a process called decidualization. Decidualization, the process by which the uterine endometrial stroma proliferates and differentiates into large epithelioid decidual cells, is critical to the establishment of fetal-maternal communication and the progression of implantation. The role of bone morphogenetic protein 2 (Bmp2) in regulating the transformation of the uterine stroma during embryo implantation in the mouse was investigated by the conditional ablation of Bmp2 in the uterus using the (PR-cre) mouse. Bmp2 gene ablation was confirmed by real-time PCR analysis in the PR-cre; Bmp2fl/fl (termed Bmp2d/d) uterus. While littermate controls average 0.9 litter of 6.2+/-0.7 pups per month, Bmp2d/d females are completely infertile. Analysis of the infertility indicates that whereas embryo attachment is normal in the Bmp2d/d as in control mice, the uterine stroma is incapable of undergoing the decidual reaction to support further embryonic development. Recombinant human BMP2 can partially rescue the decidual response, suggesting that the observed phenotypes are not due to a developmental consequence of Bmp2 ablation. Microarray analysis demonstrates that ablation of Bmp2 leads to specific gene changes, including disruption of the Wnt signaling pathway, Progesterone receptor (PR) signaling, and the induction of prostaglandin synthase 2 (Ptgs2). Taken together, these data demonstrate that Bmp2 is a critical regulator of gene expression and function in the murine uterus.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Decídua/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/deficiência , Diferenciação Celular , Proliferação de Células , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Decídua/citologia , Decídua/patologia , Implantação do Embrião , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Humanos , Infertilidade Feminina/patologia , Camundongos , Análise em Microsséries , Modelos Genéticos , Neovascularização Fisiológica , Ovário/patologia , Transdução de Sinais , Proteínas de Ligação a Tacrolimo/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Fator de Crescimento Transformador beta/deficiência , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
19.
Nat Rev Endocrinol ; 16(3): 135-146, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31780780

RESUMO

The ability of growth hormone (GH) to induce adipose tissue lipolysis has been known for over five decades; however, the molecular mechanisms that mediate this effect and the ability of GH to inhibit insulin-stimulated glucose uptake have scarcely been documented. In this same time frame, our understanding of adipose tissue has evolved to reveal a complex structure with distinct types of adipocyte, depot-specific differences, a biologically significant extracellular matrix and important endocrine properties mediated by adipokines. All these aforementioned features, in turn, can influence lipolysis. In this Review, we provide a historical and current overview of the lipolytic effect of GH in humans, mice and cultured cells. More globally, we explain lipolysis in terms of GH-induced intracellular signalling and its effect on obesity, insulin resistance and lipotoxicity. In this regard, findings that define molecular mechanisms by which GH induces lipolysis are described. Finally, data are presented for the differential effect of GH on specific adipose tissue depots and on distinct classes of metabolically active adipocytes. Together, these cellular, animal and human studies reveal novel cellular phenotypes and molecular pathways regulating the metabolic effects of GH on adipose tissue.


Assuntos
Tecido Adiposo/metabolismo , Hormônio do Crescimento/metabolismo , Animais , Humanos , Mutação , Hormônios Tireóideos/metabolismo
20.
Physiol Rep ; 8(3): e14373, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-32073221

RESUMO

AIM: Since GH stimulates lipolysis in vivo after a 2-hr lag phase, we studied whether this involves GH signaling and gene expression in adipose tissue (AT). METHODS: Human subjects (n = 9) each underwent intravenous exposure to GH versus saline with measurement of serum FFA, and GH signaling, gene array, and protein in AT biopsies after 30-120 min. Human data were corroborated in adipose-specific GH receptor knockout (FaGHRKO) mice versus wild-type mice. Expression of candidate genes identified in the array were investigated in 3T3-L1 adipocytes. RESULTS: GH increased serum FFA and AT phosphorylation of STAT5b in human subjects. This was replicated in wild-type mice, but not in FaGHRKO mice. The array identified 53 GH-regulated genes, and Ingenuity Pathway analysis showed downregulation of PDE3b, an insulin-dependent antilipolytic signal, upregulation of PTEN that inhibits insulin-dependent antilipolysis, and downregulation of G0S2 and RASD1, both encoding antilipolytic proteins. This was confirmed in 3T3-L1 adipocytes, except for PDE3B, including reciprocal effects of GH and insulin on mRNA expression of PTEN, RASD1, and G0S2. CONCLUSION: (a) GH directly stimulates AT lipolysis in a GHR-dependent manner, (b) this involves suppression of antilipolytic signals at the level of gene expression, (c) the underlying GH signaling pathways remain to be defined.


Assuntos
Tecido Adiposo/metabolismo , Hormônio do Crescimento Humano/metabolismo , Lipólise , Células 3T3 , Tecido Adiposo/efeitos dos fármacos , Adulto , Animais , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Ácidos Graxos não Esterificados/sangue , Feminino , Hormônio do Crescimento Humano/farmacologia , Humanos , Insulina/sangue , Masculino , Camundongos , Pessoa de Meia-Idade , PTEN Fosfo-Hidrolase/metabolismo , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Fator de Transcrição STAT5/metabolismo , Proteínas ras/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA