Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nucleic Acids Res ; 51(17): 9356-9368, 2023 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-37486777

RESUMO

RIG-I (retinoic acid inducible gene-I) can sense subtle differences between endogenous and viral RNA in the cytoplasm, triggering an anti-viral immune response through induction of type I interferons (IFN) and other inflammatory mediators. Multiple crystal and cryo-EM structures of RIG-I suggested a mechanism in which the C-terminal domain (CTD) is responsible for the recognition of viral RNA with a 5'-triphoshate modification, while the CARD domains serve as a trigger for downstream signaling, leading to the induction of type I IFN. However, to date contradicting conclusions have been reached around the role of ATP in the mechanism of the CARD domains ejection from RIG-I's autoinhibited state. Here we present an application of NMR spectroscopy to investigate changes induced by the binding of 5'-triphosphate and 5'-OH dsRNA, both in the presence and absence of nucleotides, to full length RIG-I with all its methionine residues selectively labeled (Met-[ϵ-13CH3]). With this approach we were able to identify residues on the CTD, helicase domain, and CARDs that served as probes to sense RNA-induced conformational changes in those respective regions. Our results were analyzed in the context of either agonistic or antagonistic RNAs, by and large supporting a mechanism proposed by the Pyle Lab in which CARD release is primarily dependent on the RNA binding event.


Assuntos
Transativadores , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Interferon Tipo I/genética , Estrutura Terciária de Proteína , RNA de Cadeia Dupla , RNA Viral/genética , RNA Viral/metabolismo , Transdução de Sinais , Transativadores/metabolismo
2.
Oncotarget ; 13: 1350-1358, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36537914

RESUMO

One of the central challenges for cancer therapy is the identification of factors in the tumor microenvironment that increase tumor progression and immune tolerance. In breast cancer, fibrosis is a histopathologic criterion for invasive cancer and poor survival that results from inflammatory factors and remodeling of the extracellular matrix to produce an immune tolerant microenvironment. To determine whether tolerance is associated with the immune checkpoint, Programmed Cell Death 1 (PD-1), NeuT/ATTAC mice, a conditional model of mammary fibrosis that we recently developed, were administered a murine-specific anti-PD-1 mAb related to pembrolizumab, and drug response was monitored by tumor development, imaging mass cytometry, immunohistochemistry and tumor gene expression by RNAseq. Tumor progression in NeuT/ATTAC mice was unaffected by weekly injection of anti-PD-1 over four months. Insensitivity to anti-PD-1 was associated with several processes, including increased tumor-associated macrophages (TAM), epithelial to mesenchymal transition (EMT), fibroblast proliferation, an enhanced extracellular matrix and the Wnt signaling pathway, including increased expression of Fzd5, Wnt5a, Vimentin, Mmp3, Col2a1 and Tgfß1. These results suggest potential therapeutic avenues that may enhance PD-1 immune checkpoint sensitivity, including the use of tumor microenvironment targeted agents and Wnt pathway inhibitors.


Assuntos
Antineoplásicos , Neoplasias , Camundongos , Animais , Via de Sinalização Wnt , Transição Epitelial-Mesenquimal , Antineoplásicos/farmacologia , Macrófagos , Microambiente Tumoral , Linhagem Celular Tumoral
3.
Front Immunol ; 12: 605930, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33854495

RESUMO

Detection of DNA is an important determinant of host-defense but also a driver of autoinflammatory and autoimmune diseases. Failure to degrade self-DNA in DNAseII or III(TREX1)-deficient mice results in activation of the cGAS-STING pathway. Deficiency of cGAS or STING in these models ameliorates disease manifestations. However, the contribution of the cGAS-STING pathway, relative to endosomal TLRs, in systemic lupus erythematosus (SLE) is controversial. In fact, STING deficiency failed to rescue, and actually exacerbated, disease manifestations in Fas-deficient SLE-prone mice. We have now extended these observations to a chronic model of SLE induced by the i.p. injection of TMPD (pristane). We found that both cGAS- and STING-deficiency not only failed to rescue mice from TMPD-induced SLE, but resulted in increased autoantibody production and higher proteinuria levels compared to cGAS STING sufficient mice. Further, we generated cGASKOFaslpr mice on a pure MRL/Faslpr background using Crispr/Cas9 and found slightly exacerbated, and not attenuated, disease. We hypothesized that the cGAS-STING pathway constrains TLR activation, and thereby limits autoimmune manifestations in these two models. Consistent with this premise, mice lacking cGAS and Unc93B1 or STING and Unc93B1 developed minimal systemic autoimmunity as compared to cGAS or STING single knock out animals. Nevertheless, TMPD-driven lupus in B6 mice was abrogated upon AAV-delivery of DNAse I, implicating a DNA trigger. Overall, this study demonstrated that the cGAS-STING pathway does not promote systemic autoimmunity in murine models of SLE. These data have important implications for cGAS-STING-directed therapies being developed for the treatment of systemic autoimmunity.


Assuntos
Autoimunidade , Suscetibilidade a Doenças , Lúpus Eritematoso Sistêmico/etiologia , Proteínas de Membrana/metabolismo , Nucleotidiltransferases/metabolismo , Transdução de Sinais , Animais , Autoanticorpos/imunologia , Autoimunidade/genética , Biomarcadores , Modelos Animais de Doenças , Expressão Gênica , Imunofenotipagem , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Nucleotidiltransferases/genética
4.
JAMA Netw Open ; 3(2): e1920833, 2020 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-32049290

RESUMO

Importance: Because cancer drugs given in combination have the potential for increased tumor-cell killing, finding the best combination partners for programmed cell death 1 (PD-1) checkpoint inhibitors could improve clinical outcomes for patients with cancer. Objective: To identify optimal strategies for combining PD-1 immune checkpoint inhibitors with other cancer therapies. Design, Setting, and Participants: This cross-sectional study compiled 319 results from 98 clinical trials testing PD-1 pathway inhibitors alone or in combination with other agents among 24 915 patients with metastatic cancer. All clinical trials had a primary completion date before September 16, 2018. Data analysis was conducted from November 2018 to August 2019. Exposures: Patients with metastatic cancer were treated with PD-1 immune checkpoint inhibitors alone or with other cancer therapies. Main Outcomes and Measures: Clinical activity was measured as objective response rates (ORRs). Combination measures included fold change from monotherapy to combination ORR, comparison of observed combination ORRs with estimated combination ORRs based on independent additivity, and a computational model to assess clinical synergy. To assess whether the ORRs of various combinations may be greater than the independent contribution of each agent, a Bliss independent activity model was used to analyze observed combination ORRs, and a Z score, measuring the difference between observed and calculated ORRs, was generated. Results: In 319 results from 98 clinical trials among 24 915 patients, ORRs for monotherapy were compared with combination data by indication and line of therapy, demonstrating an increased ORR in 105 of 127 results (82.7%) where ORRs were available for both PD-1 pathway inhibitor monotherapy and combination therapy. A few combinations showed increases above the Bliss-estimated activity, possibly identifying limited clinical synergy. The mean (SD) Z score for all trials was 0.0430 (0.0243). The mean (SD) Z score was 0.0923 (0.0628) for platinum chemotherapy regimen combinations, 0.0547 (0.0821) for vascular endothelial growth factor or vascular endothelial growth factor receptor tyrosine kinase inhibitor combinations, 0.0893 (0.086) for indoleamine 2,3-dioxygenase inhibitor combinations, and 0.0558 (0.0849) for cytotoxic T-lymphocyte-associated protein 4 inhibitor combinations. Conclusions and Relevance: In this cross-sectional study, most combination trials showed the expected benefit of combining 2 active anticancer agents, but few combination trials showed clinical synergy according to the Bliss independent activity model.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia/métodos , Neoplasias/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Ensaios Clínicos como Assunto , Estudos Transversais , Humanos , Neoplasias/imunologia , Receptor de Morte Celular Programada 1/imunologia , Resultado do Tratamento
5.
J Pharmacol Exp Ther ; 369(2): 223-233, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30804001

RESUMO

We leveraged a clinical pharmacokinetic (PK)/pharmacodynamics (PD)/efficacy relationship established with an oral phosphatidylinositol 3-kinase (PI3K)δ inhibitor (Idelalisib) in a nasal allergen challenge study to determine whether a comparable PK/PD/efficacy relationship with PI3Kδ inhibitors was observed in preclinical respiratory models of type 2 T helper cell (TH2) and type 1 T helper cell (TH1) inflammation. Results from an in vitro rat blood basophil (CD63) activation assay were used as a PD biomarker. IC50 values for PI3Kδ inhibitors, MSD-496486311, MSD-126796721, Idelalisib, and Duvelisib, were 1.2, 4.8, 0.8, and 0.5 µM. In the ovalbumin Brown Norway TH2 pulmonary inflammation model, all PI3Kδ inhibitors produced a dose-dependent inhibition of bronchoalveolar lavage eosinophils (maximum effect between 80% and 99%). In a follow-up experiment designed to investigate PK attributes [maximum (or peak) plasma concentration (Cmax), area under the curve (AUC), time on target (ToT)] that govern PI3Kδ efficacy, MSD-496486311 [3 mg/kg every day (QD) and 100 mg/kg QD] produced 16% and 93% inhibition of eosinophils, whereas doses (20 mg/kg QD, 10 mg/kg twice per day, and 3 mg/kg three times per day) produced 54% to 66% inhibition. Our profiling suggests that impact of PI3Kδ inhibitors on eosinophils is supported by a PK target with a ToT over the course of treatment close to the PD IC50 rather than strictly driven by AUC, Cmax, or Cmin (minimum blood plasma concentration) coverage. Additional studies in an Altenaria alternata rat model, a sheep Ascaris-sensitive sheep model, and a TH1-driven rat ozone exposure model did not challenge our hypothesis, suggesting that an IC50 level of TE (target engagement) sustained for 24 hours is required to produce efficacy in these traditional models. We conclude that the PK/PD observations in our animal models appear to align with clinical results associated with a TH2 airway disease.


Assuntos
Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase/farmacocinética , Doenças Respiratórias/tratamento farmacológico , Doenças Respiratórias/imunologia , Células Th1/efeitos dos fármacos , Células Th2/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Humanos , Inflamação/tratamento farmacológico , Inflamação/imunologia , Inflamação/metabolismo , Masculino , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Ratos , Doenças Respiratórias/metabolismo
6.
ACS Med Chem Lett ; 10(1): 92-97, 2019 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-30655953

RESUMO

Drugging large protein pockets is a challenge due to the need for higher molecular weight ligands, which generally possess undesirable physicochemical properties. In this communication, we highlight a strategy leveraging small molecule active site dimers to inhibit the large symmetric binding pocket in the STING protein. By taking advantage of the 2:1 binding stoichiometry, maximal buried interaction with STING protein can be achieved while maintaining the ligand physicochemical properties necessary for oral exposure. This mode of binding requires unique considerations for potency optimization including simultaneous optimization of protein-ligand as well as ligand-ligand interactions. Successful implementation of this strategy led to the identification of 18, which exhibits good oral exposure, slow binding kinetics, and functional inhibition of STING-mediated cytokine release.

7.
Cell Rep ; 17(12): 3206-3218, 2016 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-28009290

RESUMO

Recent studies have elucidated the molecular mechanism of RORγT transcriptional regulation of Th17 differentiation and function. RORγT was initially identified as a transcription factor required for thymopoiesis by maintaining survival of CD4+CD8+ (DP) thymocytes. While RORγ antagonists are currently being developed to treat autoimmunity, it remains unclear how RORγT inhibition may impact thymocyte development. In this study, we show that in addition to regulating DP thymocytes survival, RORγT also controls genes that regulate thymocyte migration, proliferation, and T cell receptor (TCR)α selection. Strikingly, pharmacological inhibition of RORγ skews TCRα gene rearrangement, limits T cell repertoire diversity, and inhibits development of autoimmune encephalomyelitis. Thus, targeting RORγT not only inhibits Th17 cell development and function but also fundamentally alters thymic-emigrant recognition of self and foreign antigens. The analysis of RORγ inhibitors has allowed us to gain a broader perspective of the diverse function of RORγT and its impact on T cell biology.


Assuntos
Encefalomielite Autoimune Experimental/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Timócitos/imunologia , Animais , Antígenos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/genética , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/terapia , Regulação da Expressão Gênica/imunologia , Rearranjo Gênico/genética , Humanos , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/antagonistas & inibidores , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Células Th17/efeitos dos fármacos , Células Th17/imunologia
8.
Br J Pharmacol ; 173(21): 3080-3087, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27417329

RESUMO

BACKGROUND AND PURPOSE: Asthma presents as a heterogeneous syndrome characterized by airway obstruction, inflammation and hyper-reactivity (AHR). Spleen tyrosine kinase (Syk) mediates allergen-induced mast cell degranulation, a central component of allergen-induced inflammation and AHR. However, the role of Syk in IgE-mediated constriction of human small airways remains unknown. In this study, we addressed whether selective inhibition of Syk attenuates IgE-mediated constriction and mast cell mediator release in human small airways. EXPERIMENTAL APPROACH: Human precision cut lung slices (hPCLS) ex vivo derived from non-asthmatic donors were incubated overnight with human IgE, dexamethasone, montelukast, antihistamines or a selective Syk inhibitor (SYKi). High-affinity IgE receptor (FcεRI) activation by anti-IgE cross-linking was performed, and constriction and mediator release measured. Airway constriction was normalized to that induced by maximal carbachol stimulation. Syk expression (determined by qPCR and immunoblot) was also evaluated in human primary airway smooth muscle (HASM) cells to determine whether Syk directly modulates HASM function. KEY RESULTS: While dexamethasone had little effect on FcεR-mediated contraction, montelukast or antihistamines partially attenuated the response. SYKi abolished anti-IgE-mediated contraction and suppressed the release of mast cell or basophil mediators from the IgE-treated hPCLS. In contrast, SYKi had little effect on the non-allergic contraction induced by carbachol. Syk mRNA and protein were undetectable in HASM cells. CONCLUSIONS AND IMPLICATIONS: A selective Syk inhibitor, but not corticosteroids, abolished FcεR-mediated contraction in human small airways ex vivo. The mechanism involved FcεRI receptor activation on mast cells or basophils that degranulate causing airway constriction, rather than direct actions on HASM.


Assuntos
Imunoglobulina E/imunologia , Pulmão/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Baço/enzimologia , Células Cultivadas , Humanos , Técnicas In Vitro , Pulmão/citologia , Pulmão/enzimologia , Pulmão/imunologia , Contração Muscular/efeitos dos fármacos , Contração Muscular/imunologia , Músculo Liso/enzimologia , Músculo Liso/imunologia , Inibidores de Proteínas Quinases/química , Proteínas Tirosina Quinases/metabolismo
9.
Eur J Pharmacol ; 724: 102-11, 2014 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-24374007

RESUMO

Glucocorticoids are used widely in the treatment of inflammatory diseases, but use is accompanied by a significant burden of adverse effects. It has been hypothesized that gene- and cell-specific regulation of the glucocorticoid receptor by small molecule ligands could be translated into a therapeutic with an improved risk-benefit profile. MK-5932 is a highly selective glucocorticoid receptor modulator that is anti-inflammatory in vivo with an improved profile on glucose metabolism: Bungard et al. (2011). Bioorg. Med. Chem. 19, 7374-7386. Here we describe the full biological profile of MK-5932. Cytokine production following lipopolysaccharide (LPS) challenge was blocked by MK-5932 in both rat and human whole blood. Oral administration reduced inflammatory cytokine levels in the serum of rats challenged with LPS. MK-5932 was anti-inflammatory in a rat contact dermatitis model, but was differentiated from 6-methylprednisolone by a lack of elevation of fasting insulin or glucose levels after 7 days of dosing, even at high exposure levels. In fact, animals in the vehicle group were consistently hyperglycemic at the end of the study, and MK-5932 normalized glucose levels in a dose-dependent manner. MK-5932 was also anti-inflammatory in the rat collagen-induced arthritis and adjuvant-induced arthritis models. In healthy dogs, oral administration of MK-5932 exerted acute pharmacodynamic effects with potency comparable to prednisone, but with important differences on neutrophil counts, again suggestive of a dissociated profile. Important gaps in our understanding of mechanism of action remain, but MK-5932 will be a useful tool in dissecting the mechanisms of glucose dysregulation by therapeutic glucocortiocids.


Assuntos
Anti-Inflamatórios/uso terapêutico , Artrite Experimental/tratamento farmacológico , Benzamidas/uso terapêutico , Dermatite de Contato/tratamento farmacológico , Edema/tratamento farmacológico , Indazóis/uso terapêutico , Receptores de Glucocorticoides/metabolismo , Animais , Anti-Inflamatórios/sangue , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Benzamidas/sangue , Benzamidas/farmacocinética , Benzamidas/farmacologia , Linhagem Celular Tumoral , Colágeno , Citocinas/sangue , Cães , Feminino , Células HeLa , Humanos , Indazóis/sangue , Indazóis/farmacocinética , Indazóis/farmacologia , Insulina , Lipopolissacarídeos , Masculino , Metilprednisolona/farmacologia , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley
10.
Stem Cells ; 27(9): 2175-84, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19591226

RESUMO

Embryonic stem (ES) cells require a coordinated network of transcription factors to maintain pluripotency or trigger lineage specific differentiation. Central to these processes are the proteins Oct4, Nanog, and Sox2. Although the transcriptional targets of these factors have been extensively studied, very little is known about how the proteins themselves are regulated, especially at the post-translational level. Post-translational modifications are well documented to have broad effects on protein stability, activity, and cellular distribution. Here, we identify a key lysine residue in the nuclear export signal of Sox2 that is acetylated, and demonstrate that blocking acetylation at this site retains Sox2 in the nucleus and sustains expression of its target genes under hyperacetylation or differentiation conditions. Mimicking acetylation at this site promotes association of Sox2 with the nuclear export machinery. In addition, increased cellular acetylation leads to reduction in Sox2 levels by ubiquitination and proteasomal degradation, thus abrogating its ability to drive transcription of its target genes. Acetylation-mediated nuclear export may be a commonly used regulatory mechanism for many Sox family members, as this lysine is conserved across species and in orthologous proteins.


Assuntos
Núcleo Celular/metabolismo , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Acetilação , Transporte Ativo do Núcleo Celular/fisiologia , Animais , Imunoprecipitação da Cromatina , Cromatografia Líquida , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/metabolismo , Ligação Proteica , Processamento de Proteína Pós-Traducional/genética , Processamento de Proteína Pós-Traducional/fisiologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição SOXB1/genética , Espectrometria de Massas em Tandem , Fatores de Transcrição de p300-CBP/metabolismo
11.
J Biol Chem ; 284(11): 6998-7006, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19139101

RESUMO

Large networks of proteins govern embryonic stem (ES) cell pluripotency. Recent analysis of the critical pluripotency factors Oct4 and Nanog has identified their interaction with multiple transcriptional repression complexes, including members of the mSin3A-HDAC complex, suggesting that these factors could be involved in the regulation of Oct4/Nanog function. mSin3A is critical for embryonic development, but the mechanism by which the mSin3A-HDAC complex is able to regulate ES cell pluripotency is undefined. Herein we show that the mSin3A-HDAC complex positively regulates Nanog expression in ES cells through Sox2, a critical ES cell transcription factor and regulator of Nanog. We have identified the mSin3A-HDAC complex to be present at the Nanog promoter only under proliferating conditions concurrent with histone acetylation. We find that Sox2 associates with mSin3A-HDAC complex members both in vitro and in vivo, similar to the interactions found between Oct4/Nanog and the mSin3A-HDAC complex. Knockdown of mSin3A-HDAC complex members or HDAC inhibitor treatment reduces Nanog expression, and overexpression of mSin3A-HDAC complex subunits stimulates Nanog expression. Our data demonstrate that the mSin3A-HDAC complex can positively regulate Nanog expression under proliferating conditions and that this activity is complementary to mSin3A-mediated p53-dependent silencing of Nanog during differentiation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Homeodomínio/biossíntese , Complexos Multiproteicos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Técnicas de Silenciamento de Genes , Histona Desacetilases/genética , Proteínas de Homeodomínio/genética , Camundongos , Complexos Multiproteicos/genética , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas/fisiologia , Proteínas Repressoras/genética , Fatores de Transcrição SOXB1/genética , Complexo Correpressor Histona Desacetilase e Sin3
12.
J Biol Chem ; 284(6): 3709-18, 2009 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-19036726

RESUMO

Nanog, Oct4, and Sox2 form the core of a transcription factor network that maintains embryonic stem cells in the pluripotent state in both humans and mice. These critical factors have been implicated as both positive and negative regulators of transcription, varying by promoter and differentiation state of the cell. The Mediator complex, a ubiquitous conserved complex of approximately 30 subunits, facilitates transcription by coordinating RNA polymerase II binding to target promoters via gene-specific activators and can be divided into several functional subcomplexes. Med12 is part of a subcomplex of four proteins associated with the core Mediator complex and has been found to function both in repressing and activating transcription when recruited to target promoters. We identified an interaction between Med12 and Nanog and present evidence of involvement of Med12 in regulation of Nanog function. Gene expression analysis of embryonic stem cells knocked down for Med12 showed a similarity to Nanog knockdown, with increased expression of Nanog-repressed targets and decreased expression of Nanog-activated targets. Using chromatin immunoprecipitation, we found that Med12 and Nanog co-occupied Nanog target promoters in embryonic stem cells and that Med12 dissociated from target promoters upon differentiation with kinetics similar to Nanog. Our results indicate that Nanog and Med12 function in concert to regulate Nanog target genes and identify a novel role for Med12 in embryonic stem cell regulation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/metabolismo , Complexos Multiproteicos/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Técnicas de Silenciamento de Genes , Proteínas de Homeodomínio/genética , Complexo Mediador , Camundongos , Complexos Multiproteicos/genética , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição/genética
13.
PLoS Genet ; 4(9): e1000190, 2008 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-18787701

RESUMO

Dot1 is an evolutionarily conserved histone methyltransferase specific for lysine 79 of histone H3 (H3K79). In Saccharomyces cerevisiae, Dot1-mediated H3K79 methylation is associated with telomere silencing, meiotic checkpoint control, and DNA damage response. The biological function of H3K79 methylation in mammals, however, remains poorly understood. Using gene targeting, we generated mice deficient for Dot1L, the murine Dot1 homologue. Dot1L-deficient embryos show multiple developmental abnormalities, including growth impairment, angiogenesis defects in the yolk sac, and cardiac dilation, and die between 9.5 and 10.5 days post coitum. To gain insights into the cellular function of Dot1L, we derived embryonic stem (ES) cells from Dot1L mutant blastocysts. Dot1L-deficient ES cells show global loss of H3K79 methylation as well as reduced levels of heterochromatic marks (H3K9 di-methylation and H4K20 tri-methylation) at centromeres and telomeres. These changes are accompanied by aneuploidy, telomere elongation, and proliferation defects. Taken together, these results indicate that Dot1L and H3K79 methylation play important roles in heterochromatin formation and in embryonic development.


Assuntos
Embrião de Mamíferos/enzimologia , Heterocromatina/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , Proteínas Metiltransferases/metabolismo , Aneuploidia , Animais , Centrômero/metabolismo , Células-Tronco Embrionárias/metabolismo , Feminino , Heterocromatina/química , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Histonas/genética , Lisina/genética , Lisina/metabolismo , Metiltransferases/genética , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Mutação , Fenótipo , Telômero/metabolismo
14.
Curr Opin Drug Discov Devel ; 9(2): 169-75, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16566287

RESUMO

Current stem-cell research has the potential to lead to new approaches for the treatment of cardiovascular, neurodegenerative and musculoskeletal diseases, as well as diabetes and cancer. Stem-cell-based approaches could be employed in cell-replacement therapy or in drug treatments that encourage adult stem cells to migrate and activate at a site of injury or disease. For such therapeutic approaches to be successful, a greater understanding of the signaling pathways that determine the diverse developmental fates of these cells is needed. From a drug-discovery perspective, efforts are being deployed in developing cell-based assays to screen for small molecules that can modulate stem-cell fate. Such compounds will provide new insights into stem-cell biology, and may ultimately contribute to effective disease treatments.


Assuntos
Transplante de Células-Tronco , Células-Tronco/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Humanos , Neoplasias/patologia , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Toxicologia/métodos
15.
Mol Biol Cell ; 13(7): 2533-46, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12134088

RESUMO

The Ena/vasodilator-stimulated phosphoprotein (VASP) protein family is implicated in the regulation of a number of actin-based cellular processes, including lamellipodial protrusion necessary for whole cell translocation. A growing body of evidence derived largely from in vitro biochemical experiments using purified proteins, cell-free extracts, and pathogen motility has begun to suggest various mechanistic roles for Ena/VASP proteins in the control of actin dynamics. Using complementation of phenotypes in Ena/VASP-deficient cells and overexpression in normal fibroblasts, we have assayed the function of a panel of mutants in one member of this family, Mena, by mutating highly conserved sequence elements found in this protein family. Surprisingly, deletion of sites required for binding of the actin monomer-binding protein profilin, a known ligand of Ena/VASP proteins, has no effect on the ability of Mena to regulate random cell motility. Our analysis revealed two features essential for Ena/VASP function in cell movement, cyclic nucleotide-dependent kinase phosphorylation sites and an F-actin binding motif. Interestingly, expression of the C-terminal EVH2 domain alone is sufficient to complement loss of Ena/VASP function in random cell motility.


Assuntos
Actinas/metabolismo , Moléculas de Adesão Celular/metabolismo , Movimento Celular/fisiologia , Proteínas do Citoesqueleto , Fosfoproteínas/metabolismo , Prolina/metabolismo , Motivos de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Genes Reporter , Teste de Complementação Genética , Proteínas de Fluorescência Verde , Indicadores e Reagentes/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Camundongos , Proteínas dos Microfilamentos , Proteínas do Tecido Nervoso/metabolismo , Fenótipo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilação , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina/metabolismo , Treonina/metabolismo , Vinculina/metabolismo , Proteína Neuronal da Síndrome de Wiskott-Aldrich
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...