Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cytotherapy ; 2024 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-38647505

RESUMO

BACKGROUND AIMS: The production of commercial autologous cell therapies such as chimeric antigen receptor T cells requires complex manual manufacturing processes. Skilled labor costs and challenges in manufacturing scale-out have contributed to high prices for these products. METHODS: We present a robotic system that uses industry-standard cell therapy manufacturing equipment to automate the steps involved in cell therapy manufacturing. The robotic cluster consists of a robotic arm and customized modules, allowing the robot to manipulate a variety of standard cell therapy instruments and materials such as incubators, bioreactors, and reagent bags. This system enables existing manual manufacturing processes to be rapidly adapted to robotic manufacturing, without having to adopt a completely new technology platform. Proof-of-concept for the robotic cluster's expansion module was demonstrated by expanding human CD8+ T cells. RESULTS: The robotic cultures showed comparable cell yields, viability, and identity to those manually performed. In addition, the robotic system was able to maintain culture sterility. CONCLUSIONS: Such modular robotic solutions may support scale-up and scale-out of cell therapies that are developed using classical manual methods in academic laboratories and biotechnology companies. This approach offers a pathway for overcoming manufacturing challenges associated with manual processes, ultimately contributing to the broader accessibility and affordability for personalized immunotherapies.

2.
Front Immunol ; 12: 744763, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34867967

RESUMO

We report on manufacturing outcomes for 41 autologous polyclonal regulatory T cell (PolyTreg) products for 7 different Phase 1 clinical trials over a 10-year period (2011-2020). Data on patient characteristics, manufacturing parameters, and manufacturing outcomes were collected from manufacturing batch records and entered into a secure database. Overall, 88% (36/41) of PolyTreg products met release criteria and 83% (34/41) of products were successfully infused into patients. Of the 7 not infused, 5 failed release criteria, and 2 were not infused because the patient became ineligible due to a change in clinical status. The median fold expansion over the 14-day manufacturing process was 434.8 -fold (range 29.8-2,232), resulting in a median post-expansion cell count of 1,841 x 106 (range 56.9-16,179 x 106). The main correlate of post-expansion cell number was starting cell number, which positively correlates with absolute circulating Treg cell count. Other parameters, including date of PolyTreg production, patient sex, and patient age did not significantly correlate with fold expansion of Treg during product manufacturing. In conclusion, PolyTreg manufacturing outcomes are consistent across trials and dates of production.


Assuntos
Produtos Biológicos , Terapia Baseada em Transplante de Células e Tecidos , Qualidade de Produtos para o Consumidor , Linfócitos T Reguladores , Produtos Biológicos/normas , Terapia Baseada em Transplante de Células e Tecidos/métodos , Terapia Baseada em Transplante de Células e Tecidos/normas , Qualidade de Produtos para o Consumidor/normas , Humanos , Transplante Autólogo/métodos , Transplante Autólogo/normas
3.
MAbs ; 13(1): 1893425, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33706686

RESUMO

G protein-coupled receptors (GPCRs) are a group of seven-transmembrane receptor proteins that have proven to be successful drug targets. Antibodies are becoming an increasingly promising modality to target these receptors due to their unique properties, such as exquisite specificity, long half-life, and fewer side effects, and their improved pharmacokinetic and pharmacodynamic profiles compared to peptides and small molecules, which results from their more favorable biodistribution. To date, there are only two US Food and Drug Administration-approved GPCR antibody drugs, namely erenumab and mogamulizumab, and this highlights the challenges encountered in identifying functional antibodies against GPCRs. Utilizing Twist's precision DNA writing technologies, we have created a GPCR-focused phage display library with 1 × 1010 diversity. Specifically, we mined endogenous GPCR binding ligand and peptide sequences and incorporated these binding motifs into the heavy chain complementarity-determining region 3 in a synthetic antibody library. Glucagon-like peptide-1 receptor (GLP-1 R) is a class B GPCR that acts as the receptor for the incretin GLP-1, which is released to regulate insulin levels in response to food intake. GLP-1 R agonists have been widely used to increase insulin secretion to lower blood glucose levels for the treatment of type 1 and type 2 diabetes, whereas GLP-1 R antagonists have applications in the treatment of severe hypoglycemia associated with bariatric surgery and hyperinsulinomic hypoglycemia. Here we present the discovery and creation of both antagonistic and agonistic GLP-1 R antibodies by panning this GPCR-focused phage display library on a GLP-1 R-overexpressing Chinese hamster ovary cell line and demonstrate their in vitro and in vivo functional activity.


Assuntos
Anticorpos Monoclonais/farmacologia , Glicemia/efeitos dos fármacos , Técnicas de Visualização da Superfície Celular , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 1/antagonistas & inibidores , Controle Glicêmico , Hipoglicemiantes/farmacologia , Incretinas/farmacologia , Biblioteca de Peptídeos , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/farmacocinética , Sítios de Ligação de Anticorpos , Biomarcadores/sangue , Glicemia/metabolismo , Células CHO , Cricetulus , Receptor do Peptídeo Semelhante ao Glucagon 1/genética , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Ensaios de Triagem em Larga Escala , Hipoglicemiantes/metabolismo , Hipoglicemiantes/farmacocinética , Incretinas/genética , Incretinas/metabolismo , Incretinas/farmacocinética , Ligantes , Masculino , Camundongos Endogâmicos C57BL , Domínios e Motivos de Interação entre Proteínas , Ratos Sprague-Dawley
4.
Biol Sex Differ ; 9(1): 49, 2018 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-30400826

RESUMO

BACKGROUND: Chronic stress is a major contributor in the development of metabolic syndrome and associated diseases, such as diabetes. High-fat diet (HFD) and sex are known modifiers of metabolic parameters. Peptide hormones corticotropin-releasing factor (CRF) and urocortins (UCN) mediate stress responses via activation and feedback to the hypothalamic-pituitary-adrenal (HPA) axis. UCN3 is a marker of pancreatic ß-cell differentiation, and UCN2 is known to ameliorate glucose levels in mice rendered diabetic with HFD. CRF receptor 2 (CRF2) is the only known cognate receptor for UCN2/3. Here, we ascertained the role of CRF2 in glucose clearance, insulin sensitivity, and other parameters associated with metabolic syndrome in a mouse model of nutritional stress. METHODS: Wild-type (WT) and Crhr2-/- (null) mice of both sexes were fed either normal chow diet or HFD. After 8 weeks, blood glucose levels in response to glucose and insulin challenge were determined. Change in body and fat mass, plasma insulin, and lipid profile were assessed. Histological evaluation of liver sections was performed. RESULTS: Here, we show that genotype (Crhr2), sex, and diet were all independent variables in the regulation of blood glucose levels, body and fat mass gain/redistribution, and insulin resistance. Surprisingly, CRF2-deficient mice (Crhr2-/-) male mice showed similarly impaired glucose clearance on HFD and chow. HFD-fed female Crhr2-/- mice redistributed their fat depots that were distinct from wild-type females and male mice on either diet. Blood cholesterol and low-density lipoprotein (LDL) levels were elevated significantly in male Crhr2-/- mice; female Crhr2-/- mice were protected. Male, but not female Crhr2-/- mice developed peripheral insulin resistance. HFD, but not chow-fed wild-type male mice developed hepatic macrovesicular steatosis. In contrast, livers of Crhr2-/- male mice showed microvesicular steatosis on either diet, whereas livers of female mice on this 8-week HFD regimen did not develop steatosis. CONCLUSIONS: CRF2 receptor dysregulation is a sexually dimorphic risk factor in development of pre-diabetic and metabolic symptoms.


Assuntos
Dieta Hiperlipídica , Receptores de Hormônio Liberador da Corticotropina/fisiologia , Caracteres Sexuais , Animais , Glicemia/análise , Colesterol/sangue , Dislipidemias/sangue , Ingestão de Alimentos , Feminino , Insulina/sangue , Resistência à Insulina , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Aumento de Peso
5.
PLoS One ; 13(9): e0203704, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30192883

RESUMO

Functional gastrointestinal disorders (FGIDs) are characterized by dysregulated gut-brain interactions. Emerging evidence shows that low-grade mucosal inflammation and immune activation contribute to FGIDs, including functional dyspepsia (FD). Stress plays an important role in the onset of FD symptoms. In human subjects with FD, presence of gastric mast cells has been reported, but factors that influence mast cell infiltration remain uncharacterized. Corticotropin-releasing factor (CRF) initiates the body's stress response and is known to degranulate mast cells. In this study, we delineated the role of the CRF system in the pathogenesis of FD in a rat model. Gastric irritation in neonate rat pups with iodoacetamide (IA) was used to induce FD-like symptoms. RNA interference (RNAi) was used to silence gastric CRF expression. Mast cell infiltrate in the stomach increased by 54% in IA-treated rats compared to controls and CRF-RNAi tended to decrease gastric mast cell infiltrate. Sucrose intake decreased in IA-treated rats and mast cell numbers showed a negative association with sucrose intake. IA treatment and transient silencing of gastric CRF increased hypothalamic CRF levels. In IA-treated rats, gastric levels of CRF receptor 2 (CRF2) decreased by ~76%, whereas hypothalamic CRF receptor 1 (CRF1) levels increased. Plasma levels of TNF-α showed a positive correlation with plasma CRF levels. Levels of phosphorylated p38 and ERK1/2 in the stomach showed a positive correlation with gastric CRF levels. Thus, CRF may contribute to low grade inflammation via modulating mast cell infiltration, cytokine levels, MAPK signaling, and the gut-brain axis.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Dispepsia/imunologia , Dispepsia/metabolismo , Mucosa Gástrica/metabolismo , Mastócitos/citologia , Animais , Comportamento Animal , Contagem de Células , Hormônio Liberador da Corticotropina/deficiência , Hormônio Liberador da Corticotropina/genética , Modelos Animais de Doenças , Dispepsia/patologia , Dispepsia/fisiopatologia , Mucosa Gástrica/efeitos dos fármacos , Trânsito Gastrointestinal/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Iodoacetamida/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Mastócitos/efeitos dos fármacos , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
6.
Am J Physiol Regul Integr Comp Physiol ; 310(11): R1244-57, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27053649

RESUMO

We investigated whether corticotropin-releasing factor receptor 2 (CRF2) and its high-affinity agonist urocortin 1 (Ucn1) mediate sex-specific signaling and immune responses. Intrarectal trinitrobenzene sulfonic acid was used to induce experimental colitis in wild-type, CRF2 knockout (CRF2KO), and heterozygous (CRF2Ht) mice of both sexes. Changes in plasma extravasation, organ weight, survival, immune cell numbers, inflammatory cytokines, and the MAPK signaling pathway were assessed. Stored intestinal biopsies from patients with Crohn's disease (CD) and age- and sex-matched individuals without inflammatory bowel disease (IBD) were examined by immunofluorescence and confocal microscopy to characterize Ucn1 and CRF receptor expression. CRF2Ht mice of both sexes showed decreased survival during colitis compared with other genotypes. Ucn1 improved survival in male mice alone. Ucn1 restored colon length and spleen and adrenal weight and decreased colonic TNF-α, IL-6, and IL-1ß levels in male CRF2Ht mice alone. CRF2Ht mice of both sexes showed decreased phosphorylation of MAPK p38 and heat shock protein 27 (Hsp27) levels. Ucn1 restored p-Hsp27 levels in male CRF2Ht mice alone. Expression of the chaperone protein Hsp90 decreased during colitis, except in male CRF2Ht mice. Taken together, our data indicate that sex shows significant interaction with genotype and Ucn1 during colitis. Human duodenal and colonic biopsies revealed that sex-specific differences exist in levels of CRF receptors and Ucn1 expression in patients with CD compared with the matched non-IBD subjects. To conclude, Ucn1 mediates sex-specific immune and cellular signaling responses via CRF2, emphasizing the need for inclusion of females in preclinical studies.


Assuntos
Colite/imunologia , Citocinas/imunologia , Mediadores da Inflamação/imunologia , Inflamação/imunologia , Receptores de Hormônio Liberador da Corticotropina/imunologia , Urocortinas/imunologia , Animais , Feminino , Masculino , Camundongos , Caracteres Sexuais
7.
Semin Reprod Med ; 30(1): 14-22, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22271290

RESUMO

Estrogens are frequently used in reproductive medicine. The Women's Health Initiative trial found that the risks of menopausal hormone therapy (MHT) exceed the benefits. The estrogens in MHT, however, were introduced prior to our understanding of the mechanism of action of estrogens. Estrogen signaling is highly complex, involving various DNA regulatory elements to which estrogen receptors bind. Numerous transcription factors and co-regulatory proteins modify chromatin structure to further regulate gene transcription. With a greater understanding of estrogen action, the major problem with the current estrogens in MHT appears to be that they are nonselective. This produces beneficial effects in bone, brain, and adipose tissue but increases the risk of breast and endometrial cancer and thromboembolism. Resurrecting MHT for long-term therapy will require the development of more selective estrogens, such as estrogen receptor (ER)ß-selective estrogens and tissue-selective ERα agonists. These compounds will offer the best prospects to expand the indications of MHT and thus prevent the chronic conditions associated with menopause.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação da Expressão Gênica , Animais , Fármacos Antiobesidade/farmacologia , Fármacos Antiobesidade/uso terapêutico , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Diabetes Mellitus Tipo 2/prevenção & controle , Epigênese Genética/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Estrogênios/metabolismo , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Síndrome Metabólica/prevenção & controle , Terapia de Alvo Molecular , Neoplasias/prevenção & controle , Obesidade/prevenção & controle , Especificidade de Órgãos , Elementos de Resposta/efeitos dos fármacos
8.
J Immunol ; 186(7): 4354-60, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21357268

RESUMO

Glucocorticoids exert potent anti-inflammatory effects by repressing proinflammatory genes. We previously demonstrated that estrogens repress numerous proinflammatory genes in U2OS cells. The objective of this study was to determine if cross talk occurs between the glucocorticoid receptor (GR) and estrogen receptor (ER)α. The effects of dexamethasone (Dex) and estradiol on 23 proinflammatory genes were examined in human U2OS cells stably transfected with ERα or GR. Three classes of genes were regulated by ERα and/or GR. Thirteen genes were repressed by both estradiol and Dex (ER/GR-repressed genes). Five genes were repressed by ER (ER-only repressed genes), and another five genes were repressed by GR (GR-only repressed genes). To examine if cross talk occurs between ER and GR at ER/GR-repressed genes, U2OS-GR cells were infected with an adenovirus that expresses ERα. The ER antagonist, ICI 182780 (ICI), blocked Dex repression of ER/GR-repressed genes. ICI did not have any effect on the GR-only repressed genes or genes activated by Dex. These results demonstrate that ICI acts on subset of proinflammatory genes in the presence of ERα but not on GR-activated genes. ICI recruited ERα to the IL-8 promoter but did not prevent Dex recruitment of GR. ICI antagonized Dex repression of the TNF response element by blocking the recruitment of nuclear coactivator 2. These findings indicate that the ICI-ERα complex blocks Dex-mediated repression by interfering with nuclear coactivator 2 recruitment to GR. Our results suggest that it might be possible to exploit ER and GR cross talk for glucocorticoid therapies using drugs that interact with ERs.


Assuntos
Mediadores da Inflamação/fisiologia , Receptor Cross-Talk/imunologia , Receptores de Estrogênio/fisiologia , Receptores de Glucocorticoides/fisiologia , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Linhagem Celular Tumoral , Dexametasona/farmacologia , Estradiol/análogos & derivados , Estradiol/farmacologia , Estradiol/fisiologia , Antagonistas de Estrogênios/farmacologia , Fulvestranto , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/antagonistas & inibidores , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/patologia , Receptor Cross-Talk/efeitos dos fármacos , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Glucocorticoides/antagonistas & inibidores
9.
Breast Cancer Res Treat ; 129(3): 777-84, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21120602

RESUMO

The role of estrogen receptor beta (ERß) in breast cancer is unclear. ERß is considered to have a protective role in breast cancer development based on findings demonstrating that ERß expression inhibits ERα-mediated proliferation of breast cancer cells. We previously demonstrated that ERß causes a ligand independent G2 cell cycle arrest in MCF-7 cells. To study the mechanisms of the ERß-mediated G2 cell cycle arrest, we investigated its effects on the regulatory pathways responsible for the G2/M phase transition. We found that ERß inhibits CDK1 activity, which is the critical determinant of the G2/M progression. CDK1 activity is modulated by both stimulatory and inhibitory factors. Cyclin B1 is the major activator of CDK1. ERß inhibited the cell cycle-dependent stimulation of cyclin B1 mRNA and protein. GADD45A and BTG2 are two major inhibitors of CDK1, which have been implicated in breast tumor formation. Based on these findings, we explored if the expression pattern of GADD45A and BTG2 is affected by ERß. We found that ERß stimulates GADD45A and BTG2 mRNA levels. The induction of these two genes is caused by ERß binding directly to these genes and recruiting c-jun and NCOA2. Our findings demonstrated that unliganded ERß causes a G2 cell cycle arrest by inactivating CDK1 through the repression of cyclin B1 and stimulation of GADD45A and BTG2 expression. These results provide evidence that drugs that stimulate the production of unliganded ERß may be effective new therapies to prevent breast cancer.


Assuntos
Proteína Quinase CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Receptor beta de Estrogênio/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteína Quinase CDC2/genética , Proteínas de Ciclo Celular/genética , Divisão Celular , Linhagem Celular Tumoral , Ciclina B1/genética , Feminino , Pontos de Checagem da Fase G2 do Ciclo Celular , Regulação da Expressão Gênica , Genes jun , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Nucleares/genética , Coativador 2 de Receptor Nuclear/genética , Coativador 2 de Receptor Nuclear/metabolismo , RNA Mensageiro , Proteínas Supressoras de Tumor/genética
10.
Curr Opin Pharmacol ; 10(6): 629-36, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20951642

RESUMO

Estrogenic effects are mediated through two estrogen receptor (ER) subtypes, ERα and ERß. Estrogens are the most commonly prescribed drugs to treat menopausal conditions, but by non-selectively triggering both ERα and ERß pathways in different tissues they can cause serious adverse effects. The different sizes of the binding pockets and sequences of their activation function domains indicate that ERα and ERß should have different specificities for ligands and biological responses that can be exploited for designing safer and more selective estrogens. ERα and ERß regulate different genes by binding to different regulatory elements and recruiting different transcription and chromatin remodeling factors that are expressed in a cell-specific manner. ERα-selective and ERß-selective agonists have been identified that demonstrate that the two ERs produce distinct biological effects. ERα and ERß agonists are a promising new approach for treating specific conditions associated with menopause.


Assuntos
Neoplasias da Mama/prevenção & controle , Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/agonistas , Inflamação/tratamento farmacológico , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular , Montagem e Desmontagem da Cromatina , Estradiol/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Estrogênios/farmacologia , Feminino , Expressão Gênica/efeitos dos fármacos , Redes Reguladoras de Genes , Fogachos/tratamento farmacológico , Humanos , Ligantes , Menopausa , Terapia de Alvo Molecular , Coativadores de Receptor Nuclear/genética , Coativadores de Receptor Nuclear/metabolismo , Ligação Proteica , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Fatores de Transcrição/metabolismo , Aumento de Peso/efeitos dos fármacos
11.
Mol Cell Endocrinol ; 315(1-2): 201-7, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19744542

RESUMO

Tamoxifen can stimulate the growth of some breast tumors and others can become resistant to tamoxifen. We previously showed that unliganded ERbeta inhibits ERalpha-mediated proliferation of MCF-7 cells. We investigated if tamoxifen might have a potential negative effect on some breast cancer cells by blocking the effects of unliganded ERbeta on gene regulation. Gene expression profiles demonstrated that unliganded ERbeta upregulated 196 genes in MCF-7 cells. Tamoxifen significantly inhibited 73 of these genes by greater than 30%, including several growth-inhibitory genes. To explore the mechanism whereby unliganded ERbeta activates genes and how tamoxifen blocks this effect, we used doxycycline-inducible U2OS-ERbeta cells to produce unliganded ERbeta. Doxycycline produced a dose-dependent activation of the NKG2E, MSMB and TUB3A genes, which was abolished by tamoxifen. Unliganded ERbeta recruitment of SRC-2 to the NKG2E gene was blocked by tamoxifen. Our findings suggest that tamoxifen might exert a negative effect on ERbeta expressing tumors due to its antagonistic action on unliganded ERbeta.


Assuntos
Proliferação de Células/efeitos dos fármacos , Antagonistas de Estrogênios/farmacologia , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Tamoxifeno/farmacologia , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Antagonistas de Estrogênios/metabolismo , Receptor beta de Estrogênio/genética , Feminino , Humanos , Subfamília C de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília C de Receptores Semelhantes a Lectina de Células NK/metabolismo , Regiões Promotoras Genéticas , Tamoxifeno/metabolismo
12.
PLoS One ; 4(7): e6271, 2009 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-19609440

RESUMO

Estrogens produce biological effects by interacting with two estrogen receptors, ERalpha and ERbeta. Drugs that selectively target ERalpha or ERbeta might be safer for conditions that have been traditionally treated with non-selective estrogens. Several synthetic and natural ERbeta-selective compounds have been identified. One class of ERbeta-selective agonists is represented by ERB-041 (WAY-202041) which binds to ERbeta much greater than ERalpha. A second class of ERbeta-selective agonists derived from plants include MF101, nyasol and liquiritigenin that bind similarly to both ERs, but only activate transcription with ERbeta. Diarylpropionitrile represents a third class of ERbeta-selective compounds because its selectivity is due to a combination of greater binding to ERbeta and transcriptional activity. However, it is unclear if these three classes of ERbeta-selective compounds produce similar biological activities. The goals of these studies were to determine the relative ERbeta selectivity and pattern of gene expression of these three classes of ERbeta-selective compounds compared to estradiol (E(2)), which is a non-selective ER agonist. U2OS cells stably transfected with ERalpha or ERbeta were treated with E(2) or the ERbeta-selective compounds for 6 h. Microarray data demonstrated that ERB-041, MF101 and liquiritigenin were the most ERbeta-selective agonists compared to estradiol, followed by nyasol and then diarylpropionitrile. FRET analysis showed that all compounds induced a similar conformation of ERbeta, which is consistent with the finding that most genes regulated by the ERbeta-selective compounds were similar to each other and E(2). However, there were some classes of genes differentially regulated by the ERbeta agonists and E(2). Two ERbeta-selective compounds, MF101 and liquiritigenin had cell type-specific effects as they regulated different genes in HeLa, Caco-2 and Ishikawa cell lines expressing ERbeta. Our gene profiling studies demonstrate that while most of the genes were commonly regulated by ERbeta-selective agonists and E(2), there were some genes regulated that were distinct from each other and E(2), suggesting that different ERbeta-selective agonists might produce distinct biological and clinical effects.


Assuntos
Receptor beta de Estrogênio/agonistas , Regulação da Expressão Gênica/efeitos dos fármacos , Western Blotting , Linhagem Celular , Estradiol/farmacologia , Transferência Ressonante de Energia de Fluorescência , Humanos , Lignanas , Nitrilas/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Fenóis/farmacologia , Propionatos/farmacologia , Transcrição Gênica/efeitos dos fármacos
13.
Endocrinology ; 148(2): 538-47, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17095596

RESUMO

Novel estrogenic therapies are needed that ameliorate menopausal symptoms and have the bone-sparing effects of endogenous estrogens but do not promote breast or uterine cancer. Recent evidence suggests that selective activation of the estrogen receptor (ER)-beta subtype inhibits breast cancer cell proliferation. To establish whether ERbeta-selective ligands represent a viable approach to improve hormone therapy, we investigated whether the estrogenic activities present in an herbal extract, MF101, used to treat hot flashes, are ERbeta selective. MF101 promoted ERbeta, but not ERalpha, activation of an estrogen response element upstream of the luciferase reporter gene. MF101 also selectively regulates transcription of endogenous genes through ERbeta. The ERbeta selectivity was not due to differential binding because MF101 binds equally to ERalpha and ERbeta. Fluorescence resonance energy transfer and protease digestion studies showed that MF101 produces a different conformation in ERalpha from ERbeta when compared with the conformations produced by estradiol. The specific conformational change induced by MF101 allows ERbeta to bind to an estrogen response element and recruit coregulatory proteins that are required for gene activation. MF101 did not activate the ERalpha-regulated proliferative genes, c-myc and cyclin D1, or stimulate MCF-7 breast cancer cell proliferation or tumor formation in a mouse xenograft model. Our results demonstrate that herbal ERbeta-selective estrogens may be a safer alternative for hormone therapy than estrogens that nonselectively activate both ER subtypes.


Assuntos
Anemarrhena/química , Receptor beta de Estrogênio/genética , Extratos Vegetais/farmacologia , Ativação Transcricional/efeitos dos fármacos , Animais , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/etiologia , Neoplasias da Mama/patologia , Carcinógenos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Dietilestilbestrol , Receptor alfa de Estrogênio/química , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/química , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Conformação Molecular , Transplante de Neoplasias , Tamanho do Órgão/efeitos dos fármacos , Extratos Vegetais/metabolismo , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/fisiologia , Transcrição Gênica/efeitos dos fármacos , Transplante Heterólogo , Útero/efeitos dos fármacos , Útero/patologia
14.
Mol Cell ; 21(4): 555-64, 2006 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-16483936

RESUMO

The decline in estrogen levels during menopause is associated with increased cytokine production and inflammatory diseases. Estrogens exert anti-inflammatory effects by repressing cytokine genes, such as tumor necrosis factor-alpha (TNFalpha). The mechanisms involved in transcriptional repression by estrogens are virtually unknown. Here, we used chromatin immunoprecipitation to investigate how estrogens repress the autoinduction of the TNFalpha gene. TNFalpha assembled a transcriptional activation complex at the TNFalpha promoter that includes c-jun, p50-NFkappaB, p65-NFkappaB, CBP, Hsp90, and unliganded estrogen receptor (ER). Estradiol repressed TNFalpha gene expression by reversing the ligand-independent activation by ERalpha and the stimulatory actions of c-jun, NFkappaB, and CBP on transcription. Silencing of GRIP1 reversed the repression of TNFalpha and other cytokine genes by estradiol, demonstrating that GRIP1 is required for transcriptional repression and can act as a corepressor. Our study demonstrates that ERalpha is a TNFalpha-induced coactivator that becomes a repressor in the presence of estradiol by recruiting GRIP1.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Regulação da Expressão Gênica , Transcrição Gênica , Fator de Necrose Tumoral alfa/genética , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ligantes , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Regiões Promotoras Genéticas , Fator de Necrose Tumoral alfa/metabolismo
15.
J Med Chem ; 48(19): 5989-6003, 2005 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-16162002

RESUMO

Estrogen receptors (ERs) control transcription of genes important for normal human development and reproduction. The signaling networks are complex, and there is a need for a molecular level understanding of the roles of receptor subtypes ERalpha and ERbeta in normal physiology and as therapeutic targets. We synthesized two series of ER ligands, based on a common indene scaffold, in an attempt to develop compounds that can selectively modulate ER-mediated transcription. The 3-ethyl-1,2-diarylindenes, utilizing an amide linker for the 1-aryl extension, bind weakly to the ERs. The 2,3-diarylindenes bind with high affinity to the ER subtypes and demonstrate a range of different biological activities, both in transcriptional reporter gene assays and inhibition of estradiol-stimulated proliferation of MCF-7 cells. Several ligands differentiate between ERalpha and ERbeta subtypes at an estrogen response element (ERE), displaying various levels of partial to full agonist activity at ERalpha, while antagonizing estradiol action at ERbeta.


Assuntos
Receptor alfa de Estrogênio/agonistas , Receptor beta de Estrogênio/antagonistas & inibidores , Indenos/síntese química , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacologia , Ligação Competitiva , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estradiol/farmacologia , Antagonistas de Estrogênios/síntese química , Antagonistas de Estrogênios/química , Antagonistas de Estrogênios/farmacologia , Receptor alfa de Estrogênio/genética , Receptor beta de Estrogênio/genética , Polarização de Fluorescência , Corantes Fluorescentes , Genes Reporter , Humanos , Indenos/química , Indenos/farmacologia , Ligantes , Elementos de Resposta , Relação Estrutura-Atividade
16.
Mol Endocrinol ; 19(1): 225-36, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15459253

RESUMO

Increasing evidence suggests that fibroblast growth factors (FGFs) are neurotrophic in GnRH neurons. However, the extent to which FGFs are involved in establishing a functional GnRH system in the whole organism has not been investigated. In this study, transgenic mice with the expression of a dominant-negative FGF receptor mutant (FGFRm) targeted to GnRH neurons were generated to examine the consequence of disrupted FGF signaling on the formation of the GnRH system. To first test the effectiveness of this strategy, GT1 cells, a GnRH neuronal cell line, were stably transfected with FGFRm. The transfected cells showed attenuated neurite outgrowth, diminished FGF-2 responsiveness in a cell survival assay, and blunted activation of the signaling pathway in response to FGF-2. Transgenic mice expressing FGFRm in a GnRH neuron-specific manner exhibited a 30% reduction in GnRH neuron number, but the anatomical distribution of GnRH neurons was unaltered. Although these mice were initially fertile, they displayed several reproductive defects, including delayed puberty, reduced litter size, and early reproductive senescence. Overall, our results are the first to show, at the level of the organism, that FGFs are one of the important components involved in the formation and maintenance of the GnRH system.


Assuntos
Fatores de Crescimento de Fibroblastos/farmacologia , Hormônio Liberador de Gonadotropina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Contagem de Células , Linhagem Celular , Sobrevivência Celular , DNA Complementar/genética , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Linhagem , Fenótipo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Transfecção , Transgenes/genética
17.
Neuroendocrinology ; 80(1): 2-10, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15340247

RESUMO

Pharmacologically increasing cyclic adenosine monophosphate (cAMP) levels in GT1 gonadotropin-releasing hormone (GnRH) cell lines increased the secretion of GnRH. Dopamine (DA) increased the GnRH secretion in GT1 cells via a DA receptor positively coupled to adenylate cyclase. We then asked whether inhibition of the DA-induced increase in cAMP would block the stimulatory effect of DA on GnRH release. Expression of the cAMP-specific phosphodiesterase (PDE4D1) was used in a genetic approach to inhibit the DA-induced increase in cAMP levels. Cells were infected with an adenovirus vector (Ad) expressing PDE4D1 (PDE-Ad) or, for controls, with an empty Ad (Null-Ad). Infection with the PDE-Ad completely blocked the forskolin-induced stimulation of GnRH secretion and [Ca2+]i and decreased the majority of the release of cAMP into the culture medium. In contrast, although PDE-Ad infection blocked virtually all of the DA-induced increase in extracellular cAMP, the release of GnRH and the increase in [Ca2+]i were only delayed for approximately 15 min. GT1 cells express the D1 DA receptor which is positively coupled to adenylate cyclase but not the D5 DA receptor. These data suggest that the initial phase of the DA-induced secretion of GnRH is dependent on an increase in cAMP levels. However, it appears that an additional non-cAMP-regulated signaling pathway is involved in the stimulation of GnRH release via the D1 DA receptor.


Assuntos
AMP Cíclico/metabolismo , Dopamina/metabolismo , Hormônio Liberador de Gonadotropina/metabolismo , Receptores de Dopamina D1/metabolismo , Transdução de Sinais/fisiologia , 3',5'-AMP Cíclico Fosfodiesterases/metabolismo , Animais , Linhagem Celular , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Neurônios/metabolismo , Radioimunoensaio , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Cancer Res ; 64(1): 423-8, 2004 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-14729654

RESUMO

Studies indicate that estrogen receptor (ER) alpha mediates breast cancer-promoting effects of estrogens. The role of ERbeta in breast cancer is unknown. Elucidating the role of ERbeta in the pathogenesis of breast cancer is important because many human breast tumors express both ERalpha and ERbeta. We show that adenovirus-mediated expression of ERbeta changes the phenotype of ERalpha-positive MCF-7 cells. Estradiol increases cell proliferation and causes tumor formation of MCF-7 cells expressing only ERalpha. In contrast, introducing ERbeta into MCF-7 cells causes an inhibition of proliferation in vitro and prevents tumor formation in a mouse xenograft model in response to estradiol. ERbeta inhibits proliferation by repressing c-myc, cyclin D1, and cyclin A gene transcription, and increasing the expression of p21(Cip1) and p27(Kip1), which leads to a G(2) cell cycle arrest. These results demonstrate that ERalpha and ERbeta produce opposite effects in MCF-7 cells on cell proliferation and tumor formation. Natural or synthetic ERbeta-selective estrogens may lack breast cancer promoting properties exhibited by estrogens in hormone replacement regimens and may be useful for chemoprevention of breast cancer.


Assuntos
Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Receptores de Estrogênio/genética , Animais , Neoplasias da Mama/prevenção & controle , Primers do DNA , Estradiol/farmacologia , Receptor beta de Estrogênio , Feminino , Fase G2/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Transplante Heterólogo
19.
Neuroendocrinology ; 78(6): 287-93, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14688441

RESUMO

Genetic targeting of the cAMP-specific phosphodiesterase 4D1 (PDE4D1) to gonadotropin-releasing hormone (GnRH) neurons in the GPR-4 transgenic rat resulted in decreased luteinizing hormone (LH) pulse frequency in castrated female and male rats. A similar decrease in the intrinsic GnRH pulse frequency was observed in GT1 GnRH cells expressing the PDE4D1 phosphodiesterase. We have extended these findings in ovariectomized (OVX) GPR-4 rats by asking what effect transgene expression had on pulsatile LH and follicle-stimulating hormone (FSH) secretion, plasma and pituitary levels of LH and FSH, and levels of the alpha-glycoprotein hormone subunit (alpha-GSU), LH-beta and FSH-beta subunit mRNAs. In OVX GPR-4 rats the LH pulse frequency but not pulse amplitude was decreased by 50% compared to wild-type littermate controls. Assaying the same samples for FSH, the FSH pulse frequency and amplitude were unchanged. The plasma and anterior pituitary levels of LH in the GPR-4 rats were significantly decreased by approximately 45%, while the plasma but not anterior pituitary level of FSH was significantly decreased by 25%. As measured by real-time RT-PCR, the mRNA levels for the alpha-GSU in the GPR-4 rats were significantly decreased by 41%, the LH-beta subunit by 38% and the FSH-beta subunit by 28%. We conclude that in the castrated female GPR-4 rats the decreased GnRH pulse frequency results in decreased levels of LH and FSH and in the alpha- and beta-subunit mRNA levels.


Assuntos
Subunidade beta do Hormônio Folículoestimulante/metabolismo , Hormônio Liberador de Gonadotropina/genética , Hormônio Luteinizante Subunidade beta/metabolismo , Ovariectomia , 3',5'-AMP Cíclico Fosfodiesterases/genética , Animais , Animais Geneticamente Modificados , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Feminino , Subunidade beta do Hormônio Folículoestimulante/sangue , Subunidade beta do Hormônio Folículoestimulante/genética , Hormônio Luteinizante Subunidade beta/sangue , Hormônio Luteinizante Subunidade beta/genética , Masculino , Fluxo Pulsátil , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
20.
Proc Natl Acad Sci U S A ; 99(26): 17191-6, 2002 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-12482943

RESUMO

Experiments in the GT1 gonadotropin-releasing hormone (GnRH) cell line have shown that the cAMP signaling pathway plays a central role in regulating the excitability of the cells. Lowering cAMP levels by expressing the constitutively active cAMP-specific phosphodiesterase PDE4D1 in GT1 cells inhibited spontaneous Ca2+ oscillations and intrinsic pulsatile GnRH secretion. To address the role of cAMP levels in endogenous GnRH neurons, we genetically targeted expression of PDE4D1 (P) to GnRH neurons in transgenic rats (R) by using the GnRH gene promoterenhancer regions (G). Three lines of transgenic rats, GPR-2, -4, and -5, were established. In situ hybridization and RT-PCR studies demonstrated that transgene expression was specifically targeted to GnRH neurons. Decreased fertility was observed in female but not in male rats from all three lines. The mean luteinizing hormone (LH) levels in ovariectomized rats were significantly reduced in the GPR-4 and -5 lines but not in the GPR-2 line. In castrated male and female GPR-4 rats, the LH pulse frequency was dramatically reduced. Six of twelve GPR-4 females studied did not ovulate and had polycystic ovaries. The remaining six females ovulated, but the magnitude of the preovulatory LH surge was inhibited by 63%. These findings support the hypothesis that cAMP signaling may play a central role in regulating excitability of GnRH neurons in vivo. The GPR-4 line of transgenic rats provides a genetic model for the understanding of the role of pulsatile gonadotropin release in follicular development.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/fisiologia , Hormônio Liberador de Gonadotropina/fisiologia , Hormônio Luteinizante/sangue , Animais , Animais Geneticamente Modificados , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Feminino , Fertilidade , Hormônio Liberador de Gonadotropina/análise , Hormônio Liberador de Gonadotropina/genética , Hipotálamo/química , Hormônio Luteinizante/metabolismo , Masculino , Síndrome do Ovário Policístico/etiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA