Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 11966, 2019 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-31427604

RESUMO

Progesterone receptor (PGR) co-ordinately regulates ovulation, fertilisation and embryo implantation through tissue-specific actions, but the mechanisms for divergent PGR action are poorly understood. Here we characterised PGR activity in mouse granulosa cells using combined ChIP-seq for PGR and H3K27ac and gene expression microarray. Comparison of granulosa, uterus and oviduct PGR-dependent genes showed almost complete tissue specificity in PGR target gene profiles. In granulosa cells 82% of identified PGR-regulated genes bound PGR within 3 kb of the gene and PGR binding sites were highly enriched in proximal promoter regions in close proximity to H3K27ac-modified active chromatin. Motif analysis showed highly enriched PGR binding to the PGR response element (GnACAnnnTGTnC), but PGR also interacted significantly with other transcription factor binding motifs. In uterus PGR showed far more tendency to bind intergenic chromatin regions and low evidence of interaction with other transcription factors. This is the first genome-wide description of PGR action in granulosa cells and systematic comparison of diverse PGR action in different reproductive tissues. It clarifies finely-tuned contextual PGR-chromatin interactions with implications for more targeted reproductive medicine.


Assuntos
Cromatina/genética , Cromatina/metabolismo , Regulação da Expressão Gênica , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Sequência de Bases , Sítios de Ligação , Feminino , Células da Granulosa/metabolismo , Histonas/metabolismo , Humanos , Motivos de Nucleotídeos , Especificidade de Órgãos , Ovário/metabolismo , Ovulação/genética , Matrizes de Pontuação de Posição Específica , Ligação Proteica , Elementos de Resposta
2.
Oncogene ; 34(43): 5418-26, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25684138

RESUMO

Rates of the most common gynecologic cancer, endometrioid adenocarcinoma (EAC), continue to rise, mirroring the global epidemic of obesity, a well-known EAC risk factor. Thus, identifying novel molecular targets to prevent and/or mitigate EAC is imperative. The prevalent Type 1 EAC commonly harbors loss of the tumor suppressor, Pten, leading to AKT activation. The major endoplasmic reticulum (ER) chaperone, GRP78, is a potent pro-survival protein to maintain ER homeostasis, and as a cell surface protein, is known to regulate the phosphatidylinositol 3-kinase (PI3K)/AKT pathway. To determine whether targeting GRP78 could suppress EAC development, we created a conditional knockout mouse model using progesterone receptor-Cre-recombinase to achieve Pten and Grp78 (cPten(f/f)Grp78(f/f)) deletion in the endometrial epithelium. Mice with a single Pten (cPten(f/f)) deletion developed well-differentiated EAC by 4 weeks. In contrast, no cPten(f/f)Grp78(f/f) mice developed EAC, even after more than 8 months of observation. Histologic examination of uteri from cPten(f/f)Grp78(f/f) mice also revealed no complex atypical hyperplasia, a well-established EAC precursor. These histologic observations among the cPten(f/f)Grp78(f/f) murine uteri also corresponded to abrogation of AKT activation within the endometrium. We further observed that GRP78 co-localized with activated AKT on the surface of EAC, thus providing an opportunity for therapeutic targeting. Consistent with previous findings that cell surface GRP78 is an upstream regulator of PI3K/AKT signaling, we show here that in vivo short-term systemic treatment with a highly specific monoclonal antibody against GRP78 suppressed AKT activation and increased apoptosis in the cPten(f/f) tumors. Collectively, these findings present GRP78-targeting therapy as an efficacious therapeutic option for EAC.


Assuntos
Anticorpos Monoclonais/farmacologia , Carcinogênese/efeitos dos fármacos , Carcinoma Endometrioide/tratamento farmacológico , Carcinoma Endometrioide/metabolismo , Proteínas de Choque Térmico/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinogênese/metabolismo , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Chaperona BiP do Retículo Endoplasmático , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Chaperonas Moleculares/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Transdução de Sinais/efeitos dos fármacos
3.
Oncogene ; 34(19): 2471-82, 2015 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-24998851

RESUMO

Type II endometrial carcinomas (ECs) are estrogen independent, poorly differentiated tumors that behave in an aggressive manner. As TP53 mutation and CDH1 inactivation occur in 80% of human endometrial type II carcinomas, we hypothesized that mouse uteri lacking both Trp53 and Cdh1 would exhibit a phenotype indicative of neoplastic transformation. Mice with conditional ablation of Cdh1 and Trp53 (Cdh1(d/d)Trp53(d/d)) clearly demonstrate architectural features characteristic of type II ECs, including focal areas of papillary differentiation, protruding cytoplasm into the lumen (hobnailing) and severe nuclear atypia at 6 months of age. Further, Cdh1(d/d)Trp53(d/d) tumors in 12-month-old mice were highly aggressive, and metastasized to nearby and distant organs within the peritoneal cavity, such as abdominal lymph nodes, mesentery and peri-intestinal adipose tissues, demonstrating that tumorigenesis in this model proceeds through the universally recognized morphological intermediates associated with type II endometrial neoplasia. We also observed abundant cell proliferation and complex angiogenesis in the uteri of Cdh1(d/d)Trp53(d/d) mice. Our microarray analysis found that most of the genes differentially regulated in the uteri of Cdh1(d/d)Trp53(d/d) mice were involved in inflammatory responses. CD163 and Arg1, markers for tumor-associated macrophages, were also detected and increased in the uteri of Cdh1(d/d)Trp53(d/d) mice, suggesting that an inflammatory tumor microenvironment with immune cell recruitment is augmenting tumor development in Cdh1(d/d)Trp53(d/d) uteri. Further, inflammatory mediators secreted from CDH1-negative, TP53 mutant endometrial cancer cells induced normal macrophages to express inflammatory-related genes through activation of nuclear factor-κB signaling. These results indicate that absence of CDH1 and TP53 in endometrial cells initiates chronic inflammation, promotes tumor microenvironment development following the recruitment of macrophages and promotes aggressive ECs.


Assuntos
Proteínas Cdh1/genética , Neoplasias do Endométrio/genética , Inflamação/genética , Macrófagos/imunologia , Proteína Supressora de Tumor p53/genética , Animais , Antígenos CD/genética , Antígenos de Diferenciação Mielomonocítica/genética , Arginase/genética , Linhagem Celular , Proliferação de Células/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Inflamação/imunologia , Inflamação/patologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Neovascularização Patológica/genética , Receptores de Superfície Celular/genética , Microambiente Tumoral/imunologia , Útero/citologia , Útero/patologia
4.
Semin Cell Dev Biol ; 24(10-12): 724-35, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23994285

RESUMO

The regulation of blastocyst implantation in the uterus is orchestrated by the ovarian hormones estrogen and progesterone. These hormones act via their nuclear receptors to direct the transcriptional activity of the endometrial compartments and create a defined period in which the uterus is permissive to embryo implantation termed the "window of receptivity". Additional members of the nuclear receptor family have also been described to have a potential role in endometrial function. Much of what we know about the function of these nuclear receptors during implantation we have learned from the use of mouse models. Transgenic murine models with targeted gene ablation have allowed us to identify a complex network of paracrine signaling between the endometrial epithelium and stroma. While some of the critical molecules have been identified, the mechanism underlying the intricate communication between endometrial compartments during the implantation window has not been fully elucidated. Defining this mechanism will help identify markers of a receptive uterine environment, ultimately providing a useful tool to help improve the fertility outlook for reproductively challenged couples. The aim of this review is to outline our current understanding of how nuclear receptors and their effector molecules regulate blastocyst implantation in the endometrium.


Assuntos
Implantação do Embrião , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Ritmo Circadiano , Humanos , Transdução de Sinais
5.
Oncogene ; 28(1): 31-40, 2009 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-18806829

RESUMO

Endometrioid adenocarcinoma is the most frequent form of endometrial cancer, usually developing in pre- and peri-menopausal women. beta-catenin abnormalities are common in endometrioid type endometrial carcinomas with squamous differentiation. To investigate the role of beta-catenin (Ctnnb1) in uterine development and tumorigenesis, mice were generated which expressed a dominant stabilized beta-catenin or had beta-catenin conditionally ablated in the uterus by crossing the PR(Cre) mouse with the Ctnnb1(f(ex3)/+) mouse or Ctnnb1(f/f) mouse, respectively. Both of the beta-catenin mutant mice have fertility defects and the ability of the uterus to undergo a hormonally induced decidual reaction was lost. Expression of the dominant stabilized beta-catenin, PR(cre/+)Ctnnb1(f(ex3)/+), resulted in endometrial glandular hyperplasia, whereas ablation of beta-catenin, PR(cre/+)Ctnnb1(f/f), induced squamous cell metaplasia in the murine uterus. Therefore, we have demonstrated that correct regulation of beta-catenin is important for uterine function as well as in the regulation of endometrial epithelial differentiation.


Assuntos
Transformação Celular Neoplásica/genética , Hiperplasia Endometrial/genética , Endométrio/crescimento & desenvolvimento , beta Catenina/fisiologia , Animais , Diferenciação Celular/genética , Proliferação de Células , Transformação Celular Neoplásica/patologia , Modelos Animais de Doenças , Hiperplasia Endometrial/patologia , Endométrio/patologia , Feminino , Infertilidade Feminina/genética , Camundongos , Camundongos Mutantes , beta Catenina/genética
6.
Artigo em Inglês | MEDLINE | ID: mdl-18540568

RESUMO

The importance of the progesterone receptor (PR) in transducing the progestin signal is firmly established in female reproductive and mammary gland biology; however, the coregulators preferentially recruited by PR in these systems have yet to be comprehensively investigated. Using an innovative genetic approach, which ablates gene function specifically in murine cell-lineages that express PR, steroid receptor coactivator 2 (SRC-2, also known as TIF-2 or GRIP-1) was shown to exert potent coregulator properties in progestin-dependent responses in the uterus and mammary gland. Uterine cells positive for PR (but devoid of SRC-2) led to an early block in embryo implantation, a phenotype not shared by knockouts for SRC-1 or SRC-3. In the case of the mammary gland, progestin-dependent branching morphogenesis and alveologenesis failed to occur in the absence of SRC-2, thereby establishing a critical coactivator role for SRC-2 in cellular proliferative programs initiated by progestins in this tissue. Importantly, the recent detection of SRC-2 in both human endometrium and breast suggests that this coregulator may provide a new clinical target for the future management of female reproductive health and/or breast cancer.


Assuntos
Glândulas Mamárias Animais/crescimento & desenvolvimento , Coativador 2 de Receptor Nuclear/fisiologia , Progestinas/fisiologia , Útero/crescimento & desenvolvimento , Animais , Feminino , Humanos , Morfogênese
7.
Artigo em Inglês | MEDLINE | ID: mdl-18543433

RESUMO

The progesterone receptor (PR) in cooperation with coregulator complexes coordinates crucial processes in female reproduction. To investigate the dynamic regulation of PR activity in vivo, a new transgenic mouse model utilizing a PR activity indicator (PRAI) system was generated. Studies utilizing the PRAI mouse have revealed that progesterone temporally regulates PR activity in female reproductive tissues. Specifically, progesterone rapidly enhances PR activity immediately after administration. However, chronic progesterone stimulation represses PR activity in female reproductive organs. Like progesterone, RU486 also temporally regulates PR activity in female reproductive organs. However, the temporal regulation of PR activity by RU486 is the inverse of progesterone's activity. RU486 acutely represses PR activity after injection but increases PR activity after chronic treatment in female reproductive tissues. Treatment with a mixed antagonist/agonist of PR, when compared to natural hormone, results in dramatically different tissue-specific patterns of intracellular PR activity, coregulator levels, and kinase activity. Transcriptional regulation of gene expression by PR is facilitated by coordinate interactions with the steroid receptor coactivators (SRCs). Bigenic PRAI-SRC knockout mouse models enabled us to draw a tissue-specific coactivator atlas for PR activity in vivo. Based on this atlas, we conclude that the endogenous physiological function of PR in distinct tissues is modulated by different SRCs. SRC-3 is the primary coactivator for PR in the breast and SRC-1 is the primary coactivator for PR in the uterus.


Assuntos
Genitália Feminina/fisiologia , Receptores de Progesterona/fisiologia , Reprodução , Animais , Feminino , Humanos , Camundongos , Mifepristona/farmacologia , Receptores de Progesterona/efeitos dos fármacos
8.
J Steroid Biochem Mol Biol ; 102(1-5): 41-50, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17067792

RESUMO

The ovarian steroid hormone progesterone is a major regulator of uterine function. The actions of this hormone is mediated through its cognate receptor, the progesterone receptor, Pgr. Ablation of the Pgr has shown that this receptor is critical for all female reproductive functions including the ability of the uterus to support and maintain the development of the implanting mouse embryo. High density DNA microarray analysis has identified direct and indirect targets of Pgr action. One of the targets of Pgr action is a member of the Hedgehog morphogen Indian Hedgehog, Ihh. Ihh and members of the Hh signaling cascade show a coordinate expression pattern in the mouse uterus during the preimplantation period of pregnancy. The expression of Ihh and its receptor Patched-1, Ptc1, as well as, down stream targets of Ihh-Ptch1 signaling, such as the orphan nuclear receptor COUP-TF II show that this morphogen pathway mediates communication between the uterine epithelial and stromal compartments. The members of the Ihh signaling axis may function to coordinate the proliferation, vascularization and differentiation of the uterine stroma during pregnancy. This analysis demonstrates that progesterone regulates uterine function in the mouse by coordinating the signals from the uterine epithelium to stroma in the preimplantation mouse uterus.


Assuntos
Receptores de Progesterona/metabolismo , Útero/fisiologia , Animais , Feminino , Proteínas Hedgehog/metabolismo , Humanos , Transdução de Sinais , Útero/citologia
9.
J Endocrinol ; 189(3): 473-84, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731779

RESUMO

Progesterone (P4) and its cognate receptor, the progesterone receptor (PGR), have important roles in the establishment and maintenance of pregnancy in the murine uterus. In previous studies, using high-density DNA microarray analysis, we identified a subset of genes whose expression is repressed by chronic P4-PGR activation in the uterus. The Clca3 gene is one of the genes whose expression is the most significantly downregulated by P4 and PGR. In the present study, we performed real-time RT-PCR and in situ hybridization to investigate the regulation of Clca3 by P4 and determine the pattern of expression of Clca3 in the uterus during early pregnancy. This analysis shows that Clca3 mRNA transcripts were detected in the luminal and glandular epithelium of the pseudopregnant uterus at day 0.5 and that the expression of Clca3 was not detected after day 3.5. P4 represses Clca3 mRNA synthesis in the luminal epithelial and glandular epithelial cells of the uterus in ovariectomized wild-type mice, but not in Pgr knockout (PRKO) mice. Conversely, estrogen (E2) induces Clca3 expression in the luminal epithelium and glandular epithelium, and this induction was repressed by P4 in the murine uterus. Analysis of the promoter region of Clca3 by in silico and transient transfection analysis in HEC-1A cells identified the regulation of Clca3 by estrogen receptor-alpha (ESR1) within the first 528 bp of 5'-flanking region of the Clca3 gene. Our studies identified Clca3 as a novel downregulated gene of PGR that is a direct target of E2 regulation.


Assuntos
Canais de Cloreto/genética , Regulação da Expressão Gênica , Progesterona/metabolismo , Regiões Promotoras Genéticas , Receptores de Progesterona/metabolismo , Útero/metabolismo , Animais , Linhagem Celular , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Hibridização In Situ/métodos , Camundongos , Camundongos Knockout , Ovariectomia , Gravidez , Progesterona/genética , Pseudogravidez , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção/métodos
10.
Mol Cell Endocrinol ; 247(1-2): 82-90, 2006 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-16406265

RESUMO

SSTR1 is found on the majority of human pancreatic beta cells, however, its role in insulin secretion has yet to be elucidated. In this study, we used the SSTR1 knockout mouse model to examine the role of SSTR1 in insulin secretion and glucose homeostasis in mice. Despite the reported effect of SSTR1 in inhibiting growth hormone secretion, SSTR1-/- mice had significantly reduced body weight with growth retardation. Perfusion of isolated mouse pancreata at 3 months of age demonstrated a significant increase in insulin secretion in SSTR1-/- mice compared with that of WT controls. We also found that at 3 months of age, SSTR1-/- mice had significantly decreased levels of systemic insulin secretion and were glucose intolerant. However, SSTR1 gene-ablated mice had a much higher rate of insulin clearance compared to WT mice at the same age. When challenged at 12 months of age, we found SSTR1-/- mice had increased glucose tolerance with exaggerated increase of insulin levels at the end of the experiment. Immunochemical analysis showed that the pancreatic islets of SSTR1-/- mice had significantly decreased levels of somatostatin staining and a significant decrease of SSTR5 expression. These results demonstrate that SSTR1 plays an important role in the regulation of insulin secretion in the endocrine pancreas in mice.


Assuntos
Glucose/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Somatostatina/metabolismo , Fatores Etários , Animais , Crescimento , Homeostase , Insulina/metabolismo , Resistência à Insulina , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Knockout , Receptores de Somatostatina/genética
11.
J Surg Res ; 129(1): 64-72, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16026801

RESUMO

INTRODUCTION: The purpose of this study was to examine the effect of global gene ablation of SSTR5 on the endocrine pancreas, insulin secretion, and glucose tolerance in aging mice, as SSTR5 is a primary regulator of insulin secretion in the mouse pancreas. METHODS: Global SSTR5-/- mice were generated and genotypes were verified using Southern blot and RT-PCR. Glucose tolerance and in vivo insulin secretion in SSTR5-/- and WT mice were examined using intraperitoneal glucose tolerance test (IPGTT;1.2-2.0 mg/kg) at 3 and 12 months of age (n = 8 per group). Basal and glucose-stimulated insulin secretion in vitro was studied using the isolated perfused mouse pancreas model at 3 and 12 months. Pancreata were removed and levels of insulin, glucagon, somatostatin, and SSTR1 were studied using immunohistochemical analysis along with H&E staining of the pancreata. RESULTS: Genotyping verified the absence of SSTR5 in SSTR5-/- mice. IPGTT demonstrated that 3-month-old SSTR5-/- mice were glucose intolerant despite similar insulin secretion both in vivo and in vitro and enlarged islets. At 12 months of age, SSTR5-/- mice had basal hypoglycemia and improved glucose intolerance associated with hyperinsulinemia in vivo and in vitro and enlarged islets. SSTR5-/- mice had increased insulin clearance at 3 and 12 months of age. SSTR1 expression was significantly increased in islets at 3 months of age, but was nearly absent in islets at 12 months of age, as was somatostatin staining in SSTR5-/- mice. CONCLUSIONS: These results suggest that both SSTR5 and SSTR1 play a pivotal role in insulin secretion and glucose regulation in mice and that their regulatory effects are age-related.


Assuntos
Envelhecimento , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Receptores de Somatostatina/deficiência , Receptores de Somatostatina/genética , Animais , Glicemia/metabolismo , Peso Corporal , Feminino , Glucagon/análise , Intolerância à Glucose/genética , Teste de Tolerância a Glucose , Transtornos do Crescimento/genética , Imuno-Histoquímica , Insulina/análise , Insulina/sangue , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Masculino , Taxa de Depuração Metabólica , Camundongos , Camundongos Knockout , Receptores de Somatostatina/fisiologia
12.
FEBS Lett ; 579(14): 3107-14, 2005 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-15919085

RESUMO

Somatostatin (SST) peptide is a potent inhibitor of insulin secretion and its effect is mediated via somatostatin receptor 5 (SSTR5) in the endocrine pancreas. To investigate the consequences of gene ablation of SSTR5 in the mouse pancreas, we have generated a mouse model in which the SSTR5 gene was specifically knocked down in the pancreatic beta cells (betaSSTR5Kd) using the Cre-lox system. Immunohistochemistry analysis showed that SSTR5 gene expression was absent in beta cells at three months of age. At the time of gene ablation, betaSSTR5Kd mice demonstrated glucose intolerance with lack of insulin response and significantly reduced serum insulin levels. Insulin tolerance test demonstrated a significant increase of insulin clearance in vivo at the same age. In vitro studies demonstrated an absence of response to SST-28 stimulation in the betaSSTR5Kd mouse islet, which was associated with a significantly reduced SST expression level in betaSSTR5Kd mice pancreata. In addition, betaSSTR5Kd mice had significantly reduced serum glucose levels and increased serum insulin levels at 12 months of age. Glucose tolerance test at an older age also indicated a persistently higher insulin level in betaSSTR5Kd mice. Further studies of betaSSTR5Kd mice had revealed elevated serum C-peptide levels at both 3 and 12 months of age, suggesting that these mice are capable of producing and releasing insulin to the periphery. These results support the hypothesis that SSTR5 plays a pivotal role in the regulation of insulin secretion in the mouse pancreas.


Assuntos
Glucose/metabolismo , Homeostase , Ilhotas Pancreáticas/metabolismo , Receptores de Somatostatina/deficiência , Receptores de Somatostatina/metabolismo , Animais , Peptídeo C/metabolismo , Quimera , Regulação da Expressão Gênica , Glucose/farmacologia , Intolerância à Glucose , Insulina/metabolismo , Secreção de Insulina , Camundongos , Receptores de Somatostatina/genética , Somatostatina/metabolismo
13.
Surgery ; 136(3): 585-92, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15349106

RESUMO

BACKGROUND: Previous studies conducted in our laboratory showed that single-gene ablation of somatostatin receptor (SSTR)1 or 5 results in diabetes in mice. The objective of this study was to determine the effect of double-gene ablation of SSTR1 and SSTR5 on insulin secretion and glucose homeostasis in mice. METHODS: SSTR1/5 -/- mice and wild-type (WT) control mice were generated and their genotype verified via polymerase chain reaction. Insulin secretion and glucose levels in these mice were examined with the use of an intraperitoneal glucose tolerance test (1.2-2.0 g/kg body weight). In vitro glucose-stimulated insulin secretion was studied with the use of the isolated perfused mouse pancreas model and islet culture techniques. Pancreata morphologic alterations were determined, and an immunohistochemistry analysis was performed. RESULTS: In vitro incubation of isolated islets from WT mice with somatostatin peptides resulted in significant reduction in insulin secretion, whereas SSTR1/5 -/- mouse islets had no response to somatostatin peptides confirming SSTR1/5 gene ablation. SSTR1/5 -/- mice also had significant increase of both basal and glucose-stimulated insulin levels in vitro. During the intraperitoneal glucose tolerance test, SSTR1/5 -/- mice had significantly improved glucose tolerance and sustained an increase in late-phase insulin secretion in vivo. Histological analysis demonstrated significant islet hyperplasia in the SSTR 1/5 -/- mouse pancreas. Immunostaining revealed an overall increase of glucagon and pancreatic polypeptide-producing cells in the islets of SSTR1/5 -/- mice. CONCLUSIONS: Double-gene ablation of SSTR1 and SSTR5 in mice resulted in a distinct phenotype with islet cell hyperplasia, hyperinsulinemia, and improved glucose tolerance. This form of diabetes differs from that seen in mice in which only the SSTR1 or SSTR5 gene was ablated. These results demonstrate that SSTR1 and SSTR5 are important regulators of insulin secretion and glucose regulation, and suggest that SSTR1 and SSTR5 are coordinately regulated.


Assuntos
Intolerância à Glucose/genética , Hiperinsulinismo/genética , Receptores de Somatostatina/genética , Animais , Glucose/metabolismo , Homeostase/genética , Homeostase/fisiologia , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/patologia , Ilhotas Pancreáticas/fisiopatologia , Camundongos , Camundongos Knockout , Modelos Animais , Receptores de Somatostatina/fisiologia
14.
J Endocrinol ; 180(2): 287-95, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14765981

RESUMO

Progesterone, via its nuclear receptor, is mandatory not only for the induction and specification of mammary gland ductal side-branching and lobuloalveologenesis but also for carcinogen-induced mammary tumorigenesis. Notwithstanding these recent advances, a more comprehensive molecular explanation of progesterone-induced mammary morphogenesis is contingent upon the identification and characterization of mammary molecular targets that are responsive to the progesterone signal. Toward this goal, we report that calcitonin, a 32 amino acid peptide hormone involved in calcium homeostasis, is exclusively expressed in, and secreted from, luminal epithelial cells within the mammary gland of the pregnant mouse, and, importantly, its expression is progesterone-dependent. Conversely, the calcitonin receptor is present during all stages of post-natal mammary development examined, is localized to the myoepithelial cell lineage, and is not regulated by progesterone. Because calcitonin induction spatiotemporally correlates with increases in progesterone-induced mammary gland proliferation and structural remodeling, we posit that calcitonin - through its receptor - may be involved in one or both of these progesterone-dependent processes.


Assuntos
Calcitonina/metabolismo , Células Epiteliais/metabolismo , Glândulas Mamárias Animais/metabolismo , Prenhez/metabolismo , Progesterona/metabolismo , Animais , Calcitonina/análise , Calcitonina/genética , Divisão Celular , Estrogênios/farmacologia , Feminino , Imuno-Histoquímica/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Gravidez , Progesterona/genética , Progesterona/farmacologia , RNA/análise , Receptores da Calcitonina/análise , Receptores da Calcitonina/metabolismo
15.
Annu Rev Physiol ; 66: 647-63, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-14977417

RESUMO

The lung is a complex organ consisting of numerous cell types that function to ensure sufficient gas exchange to oxygenate the blood. In order to accomplish this function, the lung must be exposed to the external environment and at the same time maintain a homeostatic balance between its function in gas exchange and the maintenance of inflammatory balance. During the past two decades, as molecular methodologies have evolved with the sequencing of entire genomes, the use of in vivo models to elucidate the molecular mechanisms involved in pulmonary physiology and disease have increased. The mouse has emerged as a potent model to investigate pulmonary physiology due to the explosion in molecular methods that now allow for the developmental and tissue-specific regulation of gene transcription. Initial efforts to manipulate gene expression in the mouse genome resulted in the generation of transgenic mice characterized by the constitutive expression of a specific gene and knockout mice characterized by the ablation of a specific gene. The utility of these original mouse models was limited, in many cases, by phenotypes resulting in embryonic or neonatal lethality that prevented analysis of the impact of the genetic manipulation on pulmonary biology. Second-generation transgenic mouse models employ multiple strategies that can either activate or silence gene expression thereby providing extensive temporal and spatial control of the experimental parameters of gene expression. These highly regulated mouse models are intended to serve as a foundation for further investigation of the molecular basis of human disease such as tumorigenesis. This review describes the principles, progress, and application of systems that are currently employed in the conditional regulation of gene expression in the investigation of lung cancer.


Assuntos
Modelos Animais de Doenças , Engenharia Genética , Neoplasias Pulmonares/genética , Animais , Genes de Troca , Camundongos , Camundongos Knockout/genética , Camundongos Transgênicos/genética
16.
FASEB J ; 17(14): 2142-4, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14500549

RESUMO

The Clara cell secretory protein (CCSP) imparts a protective effect to the lung during oxidant injury. However, exposure to supplemental oxygen, a common therapeutic modality for lung disease, represses the expression of CCSP in the adult mouse lung. We investigated the mechanisms of hyperoxia-induced repression of the mouse CCSP promoter. Deletion experiments in vivo and in vitro indicated that the hyperoxia-responsive elements are localized to the proximal -166 bp of the CCSP promoter. Electrophoretic mobility shift and supershift analyses demonstrated increased binding of c-Jun at the activator protein-1 site, increased binding of CCAAT/enhancer binding protein (C/EBP) beta at the C/EBP sites, and decreased binding at the Nkx2.1 sites. Western analyses revealed that hyperoxia exposure induced an increase in the expression of the C/EBPbeta isoform liver-inhibiting protein (LIP) and an increase in cytoplasmic Nkx2.1. Cotransfection of LIP or c-Jun expression plasmids decreased the transcriptional activity of the proximal -166-bp CCSP promoter. These observations suggest that hyperoxia-induced repression of the CCSP gene is mediated, at least in part, at the level of transcription and that multiple mechanisms mediate this repression. Moreover, these novel observations may provide insights for generation of therapeutic interventions for the amelioration of oxidant-induced lung injury.


Assuntos
Inativação Gênica , Proteínas/genética , Uteroglobina , Região 5'-Flanqueadora , Animais , Sítios de Ligação , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular Transformada , Citoplasma/química , Proteínas de Homeodomínio/análise , Camundongos , Modelos Genéticos , Oxigênio/toxicidade , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Sequências Reguladoras de Ácido Nucleico , Fatores de Transcrição/metabolismo , Transcrição Gênica
17.
Pancreas ; 26(3): e67-73, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12657967

RESUMO

INTRODUCTION: The function of pancreatic somatostatin in insulin secretion is controversial, and the receptor(s) mediating such event has not been exclusively investigated. AIM AND METHODOLOGY: To differentiate the specific role of SSTR5 in the mouse pancreas, we generated a mouse SSTR5 gene ablation model. Mice homozygous for the deletion (SSTR5-/-) and wild type (WT) littermate controls underwent whole pancreas perfusion to determine the effect of SSTR5 gene ablation on glucose-stimulated insulin secretion. The perfusion was done with and without octreotide added to the infusion buffer. Furthermore, pancreatic somatostatin was immunoneutralized by using a potent somatostatin monoclonal antibody to determine whether pancreatic somatostatin regulates insulin secretion in these mice. RESULTS: Results showed that at 3 months of age, there were no alterations in insulin secretion compared with WT controls. However, glucose-stimulated insulin secretion was significantly enhanced in 12-month-old SSTR5-/- mice compared with WT controls. The addition of octreotide to the perfusion significantly suppressed insulin secretion in WT controls, while it had no effect on SSTR5-/- mice. Immunoneutralization of pancreatic somatostatin resulted in enhanced glucose-stimulated insulin secretion in WT controls, but decreased levels of insulin secretion in SSTR5-/- mice. CONCLUSION: These results suggest that, in the mouse, pancreatic somatostatin regulates insulin secretion through SSTR5, and that the effect is age-specific.


Assuntos
Insulina/metabolismo , Pâncreas/metabolismo , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Glucose/farmacologia , Imuno-Histoquímica , Secreção de Insulina , Camundongos , Camundongos Knockout , Octreotida/farmacologia , Pâncreas/química , Pâncreas/efeitos dos fármacos , Perfusão , Receptores de Somatostatina/análise , Receptores de Somatostatina/genética , Somatostatina/antagonistas & inibidores , Somatostatina/imunologia
18.
Am J Physiol Lung Cell Mol Physiol ; 284(1): L108-18, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12388333

RESUMO

Interferon gamma (IFN-gamma), a potent cytokine inducing a wide range of immunologic activities, is increased in the airway secondary to viral infection or during an inflammatory response. This increase in IFN-gamma concentration may alter the expression of specific airway epithelial cell genes that regulate adaptation of airway inflammatory responses. One protein induced by IFN-gamma is Clara cell secretory protein (CCSP), which may contribute to the attenuation of airway inflammation. This study was done to investigate the molecular mechanism by which IFN-gamma stimulates the expression of the CCSP gene in mouse transformed Clara cells and transgenic mice. Deletion mapping and linker-scanning mutations demonstrated that IFN-gamma-induced expression of CCSP was regulated, in part, at the level of transcription. In vitro and in vivo studies verified that the minimal IFN-gamma-responsive segment was localized to the proximal 166 bp of the 5'-flanking region. Additionally, IFN-gamma-induced expression of CCSP was mediated indirectly through an interferon regulatory factor-1-mediated increase in hepatocyte nuclear factor-3beta.


Assuntos
Regulação da Expressão Gênica/fisiologia , Interferon gama/fisiologia , Proteínas/genética , Fatores de Transcrição , Transcrição Gênica/fisiologia , Uteroglobina , Animais , Sítios de Ligação/fisiologia , Linhagem Celular Transformada , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Expressão Gênica/fisiologia , Fator 3-beta Nuclear de Hepatócito , Fator Regulador 1 de Interferon , Camundongos , Proteínas Nucleares/metabolismo , Fosfoproteínas/fisiologia , Regiões Promotoras Genéticas , Proteínas/química , Mucosa Respiratória/citologia , Mucosa Respiratória/metabolismo , Estereoisomerismo , Transativadores/metabolismo
19.
Trends Endocrinol Metab ; 12(8): 348-53, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11551808

RESUMO

Investigations of the mechanisms involved in appropriate, developmentally regulated tissue-specific gene transcription have laid the foundations for transgenic and gene-therapy technologies directing specific induction or ablation of genes of interest in a tissue-restricted manner. This technology has further evolved to allow for temporal control of gene expression and ablation. Genes can now be switched on and off or be ablated by administering exogenous compounds. These technologies are based on the development of ligand-inducible transcription factors or recombinases that regulate gene expression or ablation by the administration of specific ligands and should lead to animal models that are better suited for investigating the molecular basis of human disease. This review describes the evolution, components and applications of systems that are currently being employed in transgenic and mutant-mouse technology for the conditional regulation of gene expression and ablation.


Assuntos
Regulação da Expressão Gênica , Marcação de Genes , Técnicas Genéticas , Animais , Humanos , Camundongos , Camundongos Transgênicos
20.
Circulation ; 104(7): 826-31, 2001 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-11502710

RESUMO

BACKGROUND: The mechanisms responsible for tumor necrosis factor (TNF)-induced LV structural remodeling in the adult heart are not known. METHODS AND RESULTS: We generated a line of transgenic mice (MHCsTNF) with cardiac restricted overexpression of TNF that develop progressive LV dilation/remodeling from 4 to 12 weeks of age. During the early phases of LV structural remodeling, there was a significant increase in total matrix metalloproteinase (MMP) activity that corresponded to a decrease in total myocardial fibrillar collagen content. As the MHCsTNF mice aged, there was a significant decrease in total MMP zymographic activity that was accompanied by an increase in total fibrillar collagen content. The changes in total MMP activity and myocardial fibrillar collagen content were related to a time- dependent increase in myocardial tissue inhibitor of metalloproteinases (TIMP)-1 levels, resulting in a significant time-dependent decrease in the MMP activity/TIMP level ratio in the MHCsTNF mice. To determine a possible mechanism for the increase in myocardial fibrosis, we also measured levels of TGF-beta(1) and TGF-beta(2) protein levels, which were shown to be significantly elevated in the hearts of the MHCsTNF mice. CONCLUSIONS: Our results suggest that progressive time-dependent changes in the balance between MMP activity and TIMP activity are responsible, at least in part, for the spectrum of TNF-induced changes in the myofibrillar collagen content that occur during LV structural remodeling in the MHCsTNF mice.


Assuntos
Cardiomegalia/metabolismo , Expressão Gênica/fisiologia , Miocárdio/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Remodelação Ventricular , Envelhecimento/metabolismo , Animais , Northern Blotting , Cardiomegalia/genética , Cardiomegalia/patologia , Colágeno/metabolismo , Citocinas/genética , Citocinas/metabolismo , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocárdio/ultraestrutura , Miofibrilas/metabolismo , Miofibrilas/ultraestrutura , Especificidade de Órgãos/fisiologia , RNA Mensageiro/metabolismo , Inibidores Teciduais de Metaloproteinases/metabolismo , Fator de Necrose Tumoral alfa/genética , Remodelação Ventricular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA