Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Chembiochem ; 23(9): e202200012, 2022 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-35235240

RESUMO

Small-molecule splicing modulators exemplified by an FDA-approved drug, risdiplam, are a new pharmacological modality for regulating the expression and stability of splice isoforms. We report a CRISPR-mediated enzyme fragment complementation (EFC) assay to quantify the splice isoform stability. The EFC assay harnessed a 42 amino acid split of a ß-galactosidase (designate α-tag), which could be fused at the termini of the target genes using CRISPR/cas9. The α-tagged splice isoform would be quantified by measuring the enzymatic activity upon complementation with the rest of ß-galactosidase. This EFC assay retained all the sequences of introns and exons of the target gene in the native genomic environment that recapitulates the cell biology of the diseases of interest. For a proof-of-concept, we developed a CRISPR-mediated EFC assay targeting the exon 7 of the survival of motor neuron 2 (SMN2) gene. The EFC assay is compatible with 384-well plates and robustly quantified the splicing modulation activity of small molecules. In this study, we also discovered that a coumarin derivative, compound 4, potently modulated SMN2 exon 7 splicing at as low as 1.1 nM.


Assuntos
Ensaios Enzimáticos , Éxons/genética , Mutação , Isoformas de Proteínas , beta-Galactosidase
2.
Sci Transl Med ; 10(454)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111643

RESUMO

Pharmacological inhibition of uncontrolled cell growth with small-molecule inhibitors is a potential strategy for treating glioblastoma multiforme (GBM), the most malignant primary brain cancer. We showed that the synthetic small-molecule KHS101 promoted tumor cell death in diverse GBM cell models, independent of their tumor subtype, and without affecting the viability of noncancerous brain cell lines. KHS101 exerted cytotoxic effects by disrupting the mitochondrial chaperone heat shock protein family D member 1 (HSPD1). In GBM cells, KHS101 promoted aggregation of proteins regulating mitochondrial integrity and energy metabolism. Mitochondrial bioenergetic capacity and glycolytic activity were selectively impaired in KHS101-treated GBM cells. In two intracranial patient-derived xenograft tumor models in mice, systemic administration of KHS101 reduced tumor growth and increased survival without discernible side effects. These findings suggest that targeting of HSPD1-dependent metabolic pathways might be an effective strategy for treating GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Metabolismo Energético , Glioblastoma/metabolismo , Glioblastoma/patologia , Tiazóis/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Chaperonina 60/metabolismo , Ciclo do Ácido Cítrico/efeitos dos fármacos , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Glioblastoma/genética , Glicólise/efeitos dos fármacos , Humanos , Redes e Vias Metabólicas/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Invasividade Neoplásica , Estresse Fisiológico/efeitos dos fármacos , Análise de Sobrevida , Transcrição Gênica/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Proc Natl Acad Sci U S A ; 114(13): 3497-3502, 2017 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-28292893

RESUMO

Stauprimide is a staurosporine analog that promotes embryonic stem cell (ESC) differentiation by inhibiting nuclear localization of the MYC transcription factor NME2, which in turn results in down-regulation of MYC transcription. Given the critical role the oncogene MYC plays in tumor initiation and maintenance, we explored the potential of stauprimide as an anticancer agent. Here we report that stauprimide suppresses MYC transcription in cancer cell lines derived from distinct tissues. Using renal cancer cells, we confirmed that stauprimide inhibits NME2 nuclear localization. Gene expression analysis also confirmed the selective down-regulation of MYC target genes by stauprimide. Consistent with this activity, administration of stauprimide inhibited tumor growth in rodent xenograft models. Our study provides a unique strategy for selectively targeting MYC transcription by pharmacological means as a potential treatment for MYC-dependent tumors.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas Proto-Oncogênicas c-myc/genética , Bibliotecas de Moléculas Pequenas/administração & dosagem , Animais , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo
4.
Future Med Chem ; 6(14): 1567-85, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25367391

RESUMO

Cancer stem cells (CSCs) have been identified in a growing list of malignancies and are believed to be responsible for cancer initiation, metastasis and relapse following certain therapies, even though they may only represent a small fraction of the cells in a given cancer. Like somatic stem cells and embryonic stem cells, CSCs are capable of self-renewal and differentiation into more mature, less tumorigenic cells that make up the bulk populations of cancer cells. Elimination of CSCs promises intriguing therapeutic potential and this concept has been adopted in preclinical drug discovery programs. Herein we will discuss the progress of these efforts, general considerations in practice, major challenges and possible solutions.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Descoberta de Drogas , Neoplasias/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Descoberta de Drogas/métodos , Humanos , Terapia de Alvo Molecular/métodos , Neoplasias/metabolismo , Neoplasias/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia
5.
Dev Biol ; 395(2): 255-67, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25238962

RESUMO

Limb development requires the coordinated growth of several tissues and structures including long bones, joints and tendons, but the underlying mechanisms are not wholly clear. Recently, we identified a small drug-like molecule - we named Kartogenin (KGN) - that greatly stimulates chondrogenesis in marrow-derived mesenchymal stem cells (MSCs) and enhances cartilage repair in mouse osteoarthritis (OA) models. To determine whether limb developmental processes are regulated by KGN, we tested its activity on committed preskeletal mesenchymal cells from mouse embryo limb buds and whole limb explants. KGN did stimulate cartilage nodule formation and more strikingly, boosted digit cartilaginous anlaga elongation, synovial joint formation and interzone compaction, tendon maturation as monitored by ScxGFP, and interdigit invagination. To identify mechanisms, we carried out gene expression analyses and found that several genes, including those encoding key signaling proteins, were up-regulated by KGN. Amongst highly up-regulated genes were those encoding hedgehog and TGFß superfamily members, particularly TFGß1. The former response was verified by increases in Gli1-LacZ activity and Gli1 mRNA expression. Exogenous TGFß1 stimulated cartilage nodule formation to levels similar to KGN, and KGN and TGFß1 both greatly enhanced expression of lubricin/Prg4 in articular superficial zone cells. KGN also strongly increased the cellular levels of phospho-Smads that mediate canonical TGFß and BMP signaling. Thus, limb development is potently and harmoniously stimulated by KGN. The growth effects of KGN appear to result from its ability to boost several key signaling pathways and in particular TGFß signaling, working in addition to and/or in concert with the filamin A/CBFß/RUNX1 pathway we identified previously to orchestrate overall limb development. KGN may thus represent a very powerful tool not only for OA therapy, but also limb regeneration and tissue repair strategies.


Assuntos
Anilidas/farmacologia , Condrogênese/efeitos dos fármacos , Extremidades/embriologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Cápsula Articular/embriologia , Mesoderma/efeitos dos fármacos , Ácidos Ftálicos/farmacologia , Animais , Primers do DNA/genética , Processamento de Imagem Assistida por Computador , Immunoblotting , Hibridização In Situ , Cápsula Articular/efeitos dos fármacos , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Luminescentes/metabolismo , Camundongos , Microscopia Confocal , Proteoglicanas/metabolismo , Regeneração/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fator de Crescimento Transformador beta1/metabolismo , Proteína GLI1 em Dedos de Zinco , Proteína Vermelha Fluorescente
6.
Angew Chem Int Ed Engl ; 53(37): 9841-5, 2014 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-25056598

RESUMO

Acute myeloid leukemia (AML), which is the most common acute adult leukemia and the second most common pediatric leukemia, still has a poor prognosis. Human C-type lectin-like molecule-1 (CLL1) is a recently identified myeloid lineage restricted cell surface marker, which is overexpressed in over 90% of AML patient myeloid blasts and in leukemic stem cells. Here, we describe the synthesis of a novel bispecific antibody, αCLL1-αCD3, using the genetically encoded unnatural amino acid, p-acetylphenylalanine. The resulting αCLL1-αCD3 recruits cytotoxic T cells to CLL1 positive cells, and demonstrates potent and selective cytotoxicity against several human AML cell lines and primary AML patient derived cells in vitro. Moreover, αCLL1-αCD3 treatment completely eliminates established tumors in an U937 AML cell line xenograft model. These results validate the clinical potential of CLL1 as an AML-specific antigen for the generation of a novel immunotherapeutic for AML.


Assuntos
Anticorpos Biespecíficos/imunologia , Imunoterapia/métodos , Lectinas Tipo C/imunologia , Leucemia Mieloide Aguda/imunologia , Adulto , Humanos
7.
Annu Rev Pharmacol Toxicol ; 53: 107-25, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23294307

RESUMO

There is considerable interest in the development of stem cell-based strategies for the treatment of a broad range of human diseases, including neurodegenerative, autoimmune, cardiovascular, and musculoskeletal diseases. To date, such regenerative approaches have focused largely on the development of cell transplantation therapies using cells derived from pluripotent embryonic stem cells (ESCs). Although there have been exciting preliminary reports describing the efficacy of ESC-derived replacement therapies, approaches involving ex vivo manipulated ESCs are hindered by issues of mutation, immune rejection, and ethical controversy. An alternative approach involves direct in vivo modulation or ex vivo expansion of endogenous adult stem cell populations using drug-like small molecules. Here we describe chemical approaches to the regulation of somatic stem cell biology that are yielding new biological insights and that may ultimately lead to innovative new medicines.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Células-Tronco Adultas/transplante , Bibliotecas de Moléculas Pequenas/farmacologia , Bibliotecas de Moléculas Pequenas/uso terapêutico , Transplante de Células-Tronco/métodos , Animais , Humanos
8.
Science ; 336(6082): 717-21, 2012 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-22491093

RESUMO

Osteoarthritis (OA) is a degenerative joint disease that involves the destruction of articular cartilage and eventually leads to disability. Molecules that promote the selective differentiation of multipotent mesenchymal stem cells (MSCs) into chondrocytes may stimulate the repair of damaged cartilage. Using an image-based high-throughput screen, we identified the small molecule kartogenin, which promotes chondrocyte differentiation (median effective concentration = 100 nM), shows chondroprotective effects in vitro, and is efficacious in two OA animal models. Kartogenin binds filamin A, disrupts its interaction with the transcription factor core-binding factor ß subunit (CBFß), and induces chondrogenesis by regulating the CBFß-RUNX1 transcriptional program. This work provides new insights into the control of chondrogenesis that may ultimately lead to a stem cell-based therapy for osteoarthritis.


Assuntos
Anilidas/farmacologia , Cartilagem Articular/citologia , Condrócitos/efeitos dos fármacos , Condrogênese , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoartrite/tratamento farmacológico , Ácidos Ftálicos/farmacologia , Anilidas/administração & dosagem , Anilidas/química , Anilidas/uso terapêutico , Animais , Bovinos , Núcleo Celular/metabolismo , Condrócitos/citologia , Condrócitos/metabolismo , Condrócitos/fisiologia , Proteínas Contráteis/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Filaminas , Ensaios de Triagem em Larga Escala , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Osteoartrite/patologia , Osteoartrite/fisiopatologia , Ácidos Ftálicos/administração & dosagem , Ácidos Ftálicos/química , Ácidos Ftálicos/uso terapêutico , Regeneração , Bibliotecas de Moléculas Pequenas , Relação Estrutura-Atividade
10.
Angew Chem Int Ed Engl ; 50(1): 200-42, 2011 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-21184400

RESUMO

Potential applications of stem cells in medicine range from their inclusion in disease modeling and drug discovery to cell transplantation and regenerative therapies. However, before this promise can be realized several obstacles must be overcome, including the control of stem cell differentiation, allogeneic rejection and limited cell availability. This will require an improved understanding of the mechanisms that govern stem cell potential and the development of robust methods to efficiently control their fate. Recently, a number of small molecules have been identified that can be used both in vitro and in vivo as tools to expand stem cells, direct their differentiation, or reprogram somatic cells to a more naive state. These molecules have provided a wealth of insights into the signaling and epigenetic mechanisms that regulate stem cell biology, and are already beginning to contribute to the development of effective treatments for tissue repair and regeneration.


Assuntos
Medicina Regenerativa/métodos , Pesquisa com Células-Tronco , Células-Tronco/fisiologia , Engenharia Tecidual/métodos , Animais , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/fisiologia , Humanos , Medicina Regenerativa/tendências , Transplante de Células-Tronco , Células-Tronco/citologia , Engenharia Tecidual/tendências
11.
Proc Natl Acad Sci U S A ; 107(38): 16542-7, 2010 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-20823227

RESUMO

Adult neurogenesis occurs in mammals and provides a mechanism for continuous neural plasticity in the brain. However, little is known about the molecular mechanisms regulating hippocampal neural progenitor cells (NPCs) and whether their fate can be pharmacologically modulated to improve neural plasticity and regeneration. Here, we report the characterization of a small molecule (KHS101) that selectively induces a neuronal differentiation phenotype. Mechanism of action studies revealed a link of KHS101 to cell cycle exit and specific binding to the TACC3 protein, whose knockdown in NPCs recapitulates the KHS101-induced phenotype. Upon systemic administration, KHS101 distributed to the brain and resulted in a significant increase in neuronal differentiation in vivo. Our findings indicate that KHS101 accelerates neuronal differentiation by interaction with TACC3 and may provide a basis for pharmacological intervention directed at endogenous NPCs.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Tiazóis/farmacologia , Células-Tronco Adultas/citologia , Células-Tronco Adultas/efeitos dos fármacos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Hipocampo/citologia , Masculino , Neurônios/metabolismo , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Tiazóis/química , Tiazóis/farmacocinética
12.
Cell Stem Cell ; 6(1): 37-47, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20085741

RESUMO

Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer associated with a very poor prognosis. Recently, the initiation and growth of GBM has been linked to brain tumor-initiating cells (BTICs), which are poorly differentiated and share features with neural stem cells (NSCs). Here we describe a kinome-wide RNA interference screen to identify factors that control the tumorigenicity of BTICs. We identified several genes whose silencing induces differentiation of BTICs derived from multiple GBM patients. In particular, knockdown of the adaptor protein TRRAP significantly increased differentiation of cultured BTICs, sensitized the cells to apoptotic stimuli, and negatively affected cell cycle progression. TRRAP knockdown also significantly suppressed tumor formation upon intracranial BTIC implantation into mice. Together, these findings support a critical role for TRRAP in maintaining a tumorigenic, stem cell-like state.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/análise , Neoplasias Encefálicas/química , Diferenciação Celular , Transformação Celular Neoplásica/química , Glioblastoma/química , Células-Tronco Neoplásicas/química , Proteínas Nucleares/análise , Interferência de RNA , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/citologia , Proteínas Nucleares/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Future Med Chem ; 2(6): 965-73, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21426114

RESUMO

Embryonic stem cells (ESCs) are a promising cell source for regenerative medicine and transplantation therapy.ESCs are able to self-renew indefinitely in culture; however, the ability to differentiate ESCs into specific cell lineages is key to exploiting their therapeutic potential. Cell-based phenotypic and reporter-based screens have been used to identify small molecules that selectively promote ESC differentiation into a variety of cell lineages. Not only will such molecules facilitate the clinical applications of stem cells, the detailed study of their mechanism is providing new insights into the biology that regulates ESC self-renewal and differentiation. In this article we discuss key issues, challenges and opportunities in the application of this chemical approach to stem cell biology.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Humanos , Medicina Regenerativa/tendências
14.
Biochim Biophys Acta ; 1803(2): 300-10, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19913059

RESUMO

Ezrin/radixin/moesin (ERM) proteins are membrane-cytoskeleton linkers that also have roles in signal transduction. Here we show that G protein-coupled receptor kinase 2 (GRK2) regulates membrane protrusion and cell migration during wound closure in Madin-Darby canine kidney (MDCK) epithelial cell monolayers at least partly through activating phosphorylation of radixin on a conserved, regulatory C-terminal Thr residue. GRK2 phosphorylated radixin exclusively on Thr 564 in vitro. Expression of a phosphomimetic (Thr-564-to-Asp) mutant of radixin resulted in increased Rac1 activity, membrane protrusion and cell motility in MDCK cells, suggesting that radixin functions "upstream" of Rac1, presumably as a scaffolding protein. Phosphorylation of ERM proteins was highest during the most active phase of epithelial cell sheet migration over the course of wound closure. In view of these results, we explored the mode of action of quinocarmycin/quinocarcin analog DX-52-1, an inhibitor of cell migration and radixin function with considerable selectivity for radixin over the other ERM proteins, finding that its mechanism of inhibition of radixin does not appear to involve binding and antagonism at the site of regulatory phosphorylation.


Assuntos
Movimento Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Proteínas do Citoesqueleto/metabolismo , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Proteínas de Membrana/metabolismo , Animais , Linhagem Celular , Proteínas do Citoesqueleto/genética , Cães , Células Epiteliais/efeitos dos fármacos , Quinase 2 de Receptor Acoplado a Proteína G/genética , Inativação Gênica , Isoquinolinas/farmacologia , Proteínas de Membrana/genética , Mutagênese Sítio-Dirigida , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteína cdc42 de Ligação ao GTP/genética , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
15.
Proc Natl Acad Sci U S A ; 106(40): 17025-30, 2009 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-19805117

RESUMO

Malignant melanoma is the most aggressive form of cutaneous carcinoma, accounting for 75% of all deaths caused by skin cancers. Microphthalmia-associated transcription factor (MITF) is a master gene regulating melanocyte development and functions as a "lineage addiction" oncogene in malignant melanoma. We have identified the receptor protein tyrosine kinase TYRO3 as an upstream regulator of MITF expression by a genome-wide gain-of-function cDNA screen and show that TYRO3 induces MITF-M expression in a SOX10-dependent manner in melanoma cells. Expression of TYRO3 is significantly elevated in human primary melanoma tissue samples and melanoma cell lines and correlates with MITF-M mRNA levels. TYRO3 overexpression bypasses BRAF(V600E)-induced senescence in primary melanocytes, inducing transformation of non-tumorigenic cell lines. Furthermore, TYRO3 knockdown represses cellular proliferation and colony formation in melanoma cells, and sensitizes them to chemotherapeutic agent-induced apoptosis; TYRO3 knockdown in melanoma cells also inhibits tumorigenesis in vivo. Taken together, these data indicate that TYRO3 may serve as a target for the development of therapeutic agents for melanoma.


Assuntos
Estudo de Associação Genômica Ampla/métodos , Melanoma/patologia , Fator de Transcrição Associado à Microftalmia/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Análise por Conglomerados , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Melanoma/genética , Melanoma/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Nus , Fator de Transcrição Associado à Microftalmia/genética , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
16.
Cell Stem Cell ; 4(5): 416-26, 2009 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-19427291

RESUMO

Embryonic stem cells (ESCs) are an attractive source of cells for disease modeling in vitro and may eventually provide access to cells/tissues for the treatment of many degenerative diseases. However, applications of ESC-derived cell types are largely hindered by the lack of highly efficient methods for lineage-specific differentiation. Using a high-content screen, we have identified a small molecule, named stauprimide, that increases the efficiency of the directed differentiation of mouse and human ESCs in synergy with defined extracellular signaling cues. Affinity-based methods revealed that stauprimide interacts with NME2 and inhibits its nuclear localization. This, in turn, leads to downregulation of c-Myc, a key regulator of the pluripotent state. Thus, our findings identify a chemical tool that primes ESCs for efficient differentiation through a mechanism that affects c-Myc expression, and this study points to an important role for NME2 in ESC self-renewal.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Inibidores Enzimáticos/farmacologia , Estaurosporina/análogos & derivados , Animais , Diferenciação Celular/efeitos dos fármacos , Imunoprecipitação da Cromatina , Regulação para Baixo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/metabolismo , Genes myc , Humanos , Camundongos , Nucleosídeo NM23 Difosfato Quinases/antagonistas & inibidores , Nucleosídeo NM23 Difosfato Quinases/genética , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Estaurosporina/farmacologia
17.
J Cell Biochem ; 103(3): 972-85, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17668446

RESUMO

Raf kinase inhibitor protein (RKIP) regulates a number of cellular processes, including cell migration. Exploring the role of RKIP in cell adhesion, we found that overexpression of RKIP in Madin-Darby canine kidney (MDCK) epithelial cells increases adhesion to the substratum, while decreasing adhesion of the cells to one another. The level of the adherens junction protein E-cadherin declines profoundly, and there is loss of normal localization of the tight junction protein ZO-1, while expression of the cell-substratum adhesion protein beta1 integrin dramatically increases. The cells also display increased adhesion and spreading on multiple substrata, including collagen, gelatin, fibronectin and laminin. In three-dimensional culture, RKIP overexpression leads to marked cell elongation and extension of long membrane protrusions into the surrounding matrix, and the cells do not form hollow cysts. RKIP-overexpressing cells generate considerably more contractile traction force than do control cells. In contrast, RNA interference-based silencing of RKIP expression results in decreased cell-substratum adhesion in both MDCK and MCF7 human breast adenocarcinoma cells. Treatment of MDCK and MCF7 cells with locostatin, a direct inhibitor of RKIP and cell migration, also reduces cell-substratum adhesion. Silencing of RKIP expression in MCF7 cells leads to a reduction in the rate of wound closure in a scratch-wound assay, although not as pronounced as that previously reported for RKIP-knockdown MDCK cells. These results suggest that RKIP has important roles in the regulation of cell adhesion, positively controlling cell-substratum adhesion while negatively controlling cell-cell adhesion, and underscore the complex functions of RKIP in cell physiology.


Assuntos
Junções Aderentes/metabolismo , Junções Célula-Matriz/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Proteína de Ligação a Fosfatidiletanolamina/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Quinases raf/antagonistas & inibidores , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Movimento Celular/efeitos dos fármacos , Junções Célula-Matriz/efeitos dos fármacos , Cães , Regulação para Baixo , Células Epiteliais/metabolismo , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/química , Humanos , Integrina beta1/metabolismo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Proteínas de Membrana/metabolismo , Oxazolidinonas/farmacologia , Proteína de Ligação a Fosfatidiletanolamina/farmacologia , Fosfoproteínas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Interferência de RNA , Células Tumorais Cultivadas , Regulação para Cima , Cicatrização/efeitos dos fármacos , Proteína da Zônula de Oclusão-1
18.
Proc Natl Acad Sci U S A ; 104(25): 10482-7, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17566101

RESUMO

Previously, a small molecule, reversine, was identified that reverses lineage-committed murine myoblasts to a more primitive multipotent state. Here, we show that reversine can increase the plasticity of C2C12 myoblasts at the single-cell level and that reversine-treated cells gain the ability to differentiate into osteoblasts and adipocytes under lineage-specific inducing conditions. Moreover, reversine is active in multiple cell types, including 3T3E1 osteoblasts and human primary skeletal myoblasts. Biochemical and cellular experiments suggest that reversine functions as a dual inhibitor of nonmuscle myosin II heavy chain and MEK1, and that both activities are required for reversine's effect. Inhibition of MEK1 and nonmuscle myosin II heavy chain results in altered cell cycle and changes in histone acetylation status, but other factors also may contribute to the activity of reversine, including activation of the PI3K signaling pathway.


Assuntos
Linhagem da Célula/efeitos dos fármacos , Morfolinas/farmacologia , Mioblastos Esqueléticos/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Purinas/farmacologia , Células 3T3 , Adipócitos/citologia , Adipócitos/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Histocitoquímica , Humanos , Camundongos , Estrutura Molecular , Morfolinas/química , Mioblastos Esqueléticos/citologia , Purinas/química , Fatores de Tempo
20.
Chem Biol ; 13(9): 973-83, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16984887

RESUMO

In the course of screening for new small-molecule modulators of cell motility, we discovered that quinocarmycin (also known as quinocarcin) analog DX-52-1 is an inhibitor of epithelial cell migration. While it has been assumed that the main target of DX-52-1 is DNA, we identified and confirmed radixin as the relevant molecular target of DX-52-1 in the cell. Radixin is a member of the ezrin/radixin/moesin family of membrane-actin cytoskeleton linker proteins that also participate in signal transduction pathways. DX-52-1 binds specifically and covalently to the C-terminal region of radixin, which contains the domain that interacts with actin filaments. Overexpression of radixin in cells abrogates their sensitivity to DX-52-1's antimigratory activity. Small interfering RNA-mediated silencing of radixin expression reduces the rate of cell migration. Finally, we found that DX-52-1 disrupts radixin's ability to interact with both actin and the cell adhesion molecule CD44.


Assuntos
Actinas/metabolismo , Movimento Celular/efeitos dos fármacos , Proteínas do Citoesqueleto/metabolismo , Receptores de Hialuronatos/metabolismo , Proteínas de Membrana/metabolismo , Animais , Biotinilação , Linhagem Celular , Proteínas do Citoesqueleto/química , Proteínas do Citoesqueleto/genética , Cães , Células Epiteliais/citologia , Humanos , Concentração Inibidora 50 , Insetos , Isoquinolinas/síntese química , Isoquinolinas/metabolismo , Isoquinolinas/farmacocinética , Isoquinolinas/farmacologia , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Ligação Proteica/efeitos dos fármacos , Interferência de RNA , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...