Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(6): 2325-30, 2014 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-24449858

RESUMO

Small-fiber neuropathy (SFN) is a disorder of peripheral nerves commonly found in patients with diabetes mellitus, HIV infection, and those receiving chemotherapy. The complexity of disease etiology has led to a scarcity of effective treatments. Using two models of progressive SFN, we show that overexpression of glial cell line-derived neurotrophic factor (GDNF) in skin keratinocytes or topical application of XIB4035, a reported nonpeptidyl agonist of GDNF receptor α1 (GFRα1), are effective treatments for SFN. We also demonstrate that XIB4035 is not a GFRα1 agonist, but rather it enhances GFRα family receptor signaling in conjunction with ligand stimulation. Taken together, our results indicate that topical application of GFRα/RET receptor signaling modulators may be a unique therapy for SFN, and we have identified XIB4035 as a candidate therapeutic agent.


Assuntos
Eritromelalgia/tratamento farmacológico , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Quinolinas/uso terapêutico , Transdução de Sinais , Administração Tópica , Animais , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Ligantes , Camundongos , Camundongos Transgênicos , Quinolinas/administração & dosagem , Bibliotecas de Moléculas Pequenas
2.
Proc Natl Acad Sci U S A ; 107(39): 17005-10, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20837532

RESUMO

Recent studies indicate that molecules released by glia can induce synapse formation. However, what induces glia to produce such signals, their identity, and their in vivo relevance remain poorly understood. Here we demonstrate that supporting cells of the vestibular organ--cells that have many characteristics of glia--promote synapse formation only when induced by neuron-derived signals. Furthermore, we identify BDNF as the synaptogenic signal produced by these nonneuronal cells. Mice in which erbB signaling has been eliminated in supporting cells have vestibular dysfunction caused by failure of synapse formation between hair cells and sensory neurons. This phenotype correlates with reduced BDNF expression in supporting cells and is rescued by reexpression of BDNF in these cells. Furthermore, knockdown of BDNF expression in supporting cells postnatally phenocopies the loss of erbB signaling. These results indicate that vestibular supporting cells contribute in vivo to vestibular synapse formation and that this is mediated by reciprocal signals between sensory neurons and supporting cells involving erbB receptors and BDNF.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Epitélio/fisiologia , Receptores ErbB/metabolismo , Neuroglia/fisiologia , Sinapses/fisiologia , Vestíbulo do Labirinto/fisiologia , Animais , Receptores ErbB/genética , Proteína Glial Fibrilar Ácida , Células Ciliadas Vestibulares/metabolismo , Células Ciliadas Vestibulares/fisiologia , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Receptor ErbB-4 , Vestíbulo do Labirinto/citologia
3.
J Med Chem ; 65(6): 4600-4615, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35293760

RESUMO

Inhibition of the S-adenosyl methionine (SAM)-producing metabolic enzyme, methionine adenosyltransferase 2A (MAT2A), has received significant interest in the field of medicinal chemistry due to its implication as a synthetic lethal target in cancers with the deletion of the methylthioadenosine phosphorylase (MTAP) gene. Here, we report the identification of novel MAT2A inhibitors with distinct in vivo properties that may enhance their utility in treating patients. Following a high-throughput screening, we successfully applied the structure-based design lessons from our first-in-class MAT2A inhibitor, AG-270, to rapidly redesign and optimize our initial hit into two new lead compounds: a brain-penetrant compound, AGI-41998, and a potent, but limited brain-penetrant compound, AGI-43192. We hope that the identification and first disclosure of brain-penetrant MAT2A inhibitors will create new opportunities to explore the potential therapeutic effects of SAM modulation in the central nervous system (CNS).


Assuntos
Metionina Adenosiltransferase , Neoplasias , Encéfalo/metabolismo , Desenho de Fármacos , Humanos , Neoplasias/tratamento farmacológico , S-Adenosilmetionina/metabolismo
4.
J Med Chem ; 64(8): 4430-4449, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33829783

RESUMO

The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was recently implicated as a synthetic lethal target in cancers with deletion of the methylthioadenosine phosphorylase (MTAP) gene, which is adjacent to the CDKN2A tumor suppressor and codeleted with CDKN2A in approximately 15% of all cancers. Previous attempts to target MAT2A with small-molecule inhibitors identified cellular adaptations that blunted their efficacy. Here, we report the discovery of highly potent, selective, orally bioavailable MAT2A inhibitors that overcome these challenges. Fragment screening followed by iterative structure-guided design enabled >10 000-fold improvement in potency of a family of allosteric MAT2A inhibitors that are substrate noncompetitive and inhibit release of the product, S-adenosyl methionine (SAM), from the enzyme's active site. We demonstrate that potent MAT2A inhibitors substantially reduce SAM levels in cancer cells and selectively block proliferation of MTAP-null cells both in tissue culture and xenograft tumors. These data supported progressing AG-270 into current clinical studies (ClinicalTrials.gov NCT03435250).


Assuntos
Inibidores Enzimáticos/química , Metionina Adenosiltransferase/antagonistas & inibidores , Purina-Núcleosídeo Fosforilase/genética , Sítios de Ligação , Cristalografia por Raios X , Desenho de Fármacos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/uso terapêutico , Homozigoto , Humanos , Metionina Adenosiltransferase/metabolismo , Simulação de Dinâmica Molecular , Neoplasias/tratamento farmacológico , Purina-Núcleosídeo Fosforilase/metabolismo , S-Adenosilmetionina/metabolismo , Relação Estrutura-Atividade
5.
Cancer Cell ; 39(2): 209-224.e11, 2021 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-33450196

RESUMO

The methylthioadenosine phosphorylase (MTAP) gene is located adjacent to the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor-suppressor gene and is co-deleted with CDKN2A in approximately 15% of all cancers. This co-deletion leads to aggressive tumors with poor prognosis that lack effective, molecularly targeted therapies. The metabolic enzyme methionine adenosyltransferase 2α (MAT2A) was identified as a synthetic lethal target in MTAP-deleted cancers. We report the characterization of potent MAT2A inhibitors that substantially reduce levels of S-adenosylmethionine (SAM) and demonstrate antiproliferative activity in MTAP-deleted cancer cells and tumors. Using RNA sequencing and proteomics, we demonstrate that MAT2A inhibition is mechanistically linked to reduced protein arginine methyltransferase 5 (PRMT5) activity and splicing perturbations. We further show that DNA damage and mitotic defects ensue upon MAT2A inhibition in HCT116 MTAP-/- cells, providing a rationale for combining the MAT2A clinical candidate AG-270 with antimitotic taxanes.


Assuntos
Dano ao DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Metionina Adenosiltransferase/antagonistas & inibidores , Proteína-Arginina N-Metiltransferases/genética , Purina-Núcleosídeo Fosforilase/genética , Splicing de RNA/efeitos dos fármacos , RNA Mensageiro/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Inibidor p16 de Quinase Dependente de Ciclina , Dano ao DNA/genética , Deleção de Genes , Células HCT116 , Células HEK293 , Humanos , Metionina Adenosiltransferase/genética , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Neoplasias/tratamento farmacológico , Neoplasias/genética , Splicing de RNA/genética , S-Adenosilmetionina/metabolismo
6.
ACS Med Chem Lett ; 11(10): 1899-1904, 2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-33062171

RESUMO

Hepatocellular carcinoma (HCC) accounts for a majority of primary liver cancer and is one of the most common forms of cancer worldwide. Aberrant signaling of the FGF19-FGFR4 pathway leads to HCC in mice and is hypothesized to be a driver in FGF19 amplified HCC in humans. Multiple small molecule inhibitors have been pursued as targeted therapies for HCC in recent years, including several selective FGFR4 inhibitors that are currently being evaluated in clinical trials. Herein, we report a novel series of highly selective, covalent 2-amino-6,8-dimethyl-pyrido[2,3-d]pyrimidin-7(8H)-ones that potently and selectively inhibit FGFR4 signaling through covalent modification of Cys552, which was confirmed by X-ray crystallography. Correlative target occupancy and pFGFR4 inhibition were observed in vivo, as well as tumor regression in preclinical models of orthotopic and sorafenib-resistant HCC.

7.
Nat Commun ; 10(1): 96, 2019 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-30626880

RESUMO

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Assuntos
Antineoplásicos/farmacologia , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Esqualeno Mono-Oxigenase/antagonistas & inibidores , Esqualeno Mono-Oxigenase/metabolismo , Antineoplásicos/química , Linhagem Celular Tumoral , Colesterol/biossíntese , Deleção de Genes , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos
8.
Cell Chem Biol ; 25(6): 705-717.e11, 2018 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-29628435

RESUMO

Activating KRAS mutations are major oncogenic drivers in multiple tumor types. Synthetic lethal screens have previously been used to identify targets critical for the survival of KRAS mutant cells, but their application to drug discovery has proven challenging, possibly due in part to a failure of monolayer cultures to model tumor biology. Here, we report the results of a high-throughput synthetic lethal screen for small molecules that selectively inhibit the growth of KRAS mutant cell lines in soft agar. Chemoproteomic profiling identifies the target of the most KRAS-selective chemical series as dihydroorotate dehydrogenase (DHODH). DHODH inhibition is shown to perturb multiple metabolic pathways. In vivo preclinical studies demonstrate strong antitumor activity upon DHODH inhibition in a pancreatic tumor xenograft model.


Assuntos
Oxirredutases atuantes sobre Doadores de Grupo CH-CH/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Pirimidinas/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Di-Hidro-Orotato Desidrogenase , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Feminino , Humanos , Camundongos , Camundongos SCID , Mutação , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/antagonistas & inibidores , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/genética , Pirimidinas/química , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Células Tumorais Cultivadas
9.
J Comp Neurol ; 501(1): 30-7, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17206617

RESUMO

Recent studies indicate that neurotrophin 3 (NT3) may be important for the maintenance and function of the adult inner ear, but the pattern of postnatal NT3 expression in this organ has not been characterized. We used a reporter mouse in which cells expressing NT3 also express beta-galactosidase, allowing for their histochemical visualization, to determine the pattern of NT3 expression in cochlear and vestibular organs. We analyzed animals from birth (P0) to adult (P135). At P0, NT3 was strongly expressed in supporting cells and hair cells of all vestibular and cochlear sense organs, Reissner's membrane, saccular membrane, and the dark cells adjacent to canal organs. With increasing age, staining disappeared in most cell types but remained relatively high in inner hair cells (IHCs) and to a lesser extent in IHC supporting cells. In the cochlea, by P0 there is a longitudinal gradient (apex > base) that persists into adulthood. In vestibular maculae, staining gradients are: striolar > extrastriolar regions and supporting cells > hair cells. By P135, cochlear staining is restricted to IHCs and their supporting cells, with stronger expression in the apex than the base. By the same age, in the vestibular organs, NT3 expression is weak and restricted to saccular and utricular supporting cells. These results suggest that NT3 might play a long-term role in the maintenance and functioning of the adult auditory and vestibular systems and that supporting cells are the main source of this factor in the adult.


Assuntos
Envelhecimento/metabolismo , Animais Recém-Nascidos/crescimento & desenvolvimento , Animais Recém-Nascidos/metabolismo , Orelha Interna/metabolismo , Neurotrofina 3/metabolismo , Animais , Cóclea/metabolismo , Células Ciliadas Auditivas Internas/metabolismo , Imuno-Histoquímica , Óperon Lac , Camundongos , Camundongos Transgênicos , Sáculo e Utrículo/metabolismo , Coloração e Rotulagem , Distribuição Tecidual , beta-Galactosidase/metabolismo
10.
PLoS One ; 12(9): e0185092, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28950000

RESUMO

Tumor cells display fundamental changes in metabolism and nutrient uptake in order to utilize additional nutrient sources to meet their enhanced bioenergetic requirements. Glutamine (Gln) is one such nutrient that is rapidly taken up by tumor cells to fulfill this increased metabolic demand. A vital step in the catabolism of glutamine is its conversion to glutamate by the mitochondrial enzyme glutaminase (GLS). This study has identified GLS a potential therapeutic target in breast cancer, specifically in the basal subtype that exhibits a deregulated glutaminolysis pathway. Using inducible shRNA mediated gene knockdown, we discovered that loss of GLS function in triple-negative breast cancer (TNBC) cell lines with a deregulated glutaminolysis pathway led to profound tumor growth inhibition in vitro and in vivo. GLS knockdown had no effect on growth and metabolite levels in non-TNBC cell lines. We rescued the anti-tumor effect of GLS knockdown using shRNA resistant cDNAs encoding both GLS isoforms and by addition of an α-ketoglutarate (αKG) analog thus confirming the critical role of GLS in TNBC. Pharmacological inhibition of GLS with the small molecule inhibitor CB-839 reduced cell growth and led to a decrease in mammalian target of rapamycin (mTOR) activity and an increase in the stress response pathway driven by activating transcription factor 4 (ATF4). Finally, we found that GLS inhibition synergizes with mTOR inhibition, which introduces the possibility of a novel therapeutic strategy for TNBC. Our study revealed that GLS is essential for the survival of TNBC with a deregulated glutaminolysis pathway. The synergistic activity of GLS and mTOR inhibitors in TNBC cell lines suggests therapeutic potential of this combination for the treatment of vulnerable subpopulations of TNBC.


Assuntos
Glutaminase/metabolismo , Glutamina/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/enzimologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia
11.
Cell Rep ; 17(3): 876-890, 2016 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-27732861

RESUMO

Although aberrant metabolism in tumors has been well described, the identification of cancer subsets with particular metabolic vulnerabilities has remained challenging. Here, we conducted an siRNA screen focusing on enzymes involved in the tricarboxylic acid (TCA) cycle and uncovered a striking range of cancer cell dependencies on OGDH, the E1 subunit of the alpha-ketoglutarate dehydrogenase complex. Using an integrative metabolomics approach, we identified differential aspartate utilization, via the malate-aspartate shuttle, as a predictor of whether OGDH is required for proliferation in 3D culture assays and for the growth of xenograft tumors. These findings highlight an anaplerotic role of aspartate and, more broadly, suggest that differential nutrient utilization patterns can identify subsets of cancers with distinct metabolic dependencies for potential pharmacological intervention.


Assuntos
Ácido Aspártico/metabolismo , Complexo Cetoglutarato Desidrogenase/metabolismo , Neoplasias/metabolismo , Animais , Linhagem Celular Tumoral , Respiração Celular/efeitos dos fármacos , Ciclo do Ácido Cítrico/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Humanos , RNA Interferente Pequeno/metabolismo
12.
Cell Rep ; 15(3): 574-587, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-27068473

RESUMO

Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.


Assuntos
Adenosina/análogos & derivados , Antígenos de Neoplasias/metabolismo , Deleção de Genes , Metionina Adenosiltransferase/metabolismo , Neoplasias/enzimologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Purina-Núcleosídeo Fosforilase/metabolismo , Transdução de Sinais , Tionucleosídeos/metabolismo , Adenosina/metabolismo , Genômica , Células HCT116 , Humanos , Complexos Multiproteicos/metabolismo , Neoplasias/metabolismo , Purina-Núcleosídeo Fosforilase/deficiência , RNA Interferente Pequeno/metabolismo
13.
Cancer Res ; 73(19): 6024-35, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23928993

RESUMO

HER2/HER3 dimerization resulting from overexpression of HER2 or neuregulin (NRG1) in cancer leads to HER3-mediated oncogenic activation of phosphoinositide 3-kinase (PI3K) signaling. Although ligand-blocking HER3 antibodies inhibit NRG1-driven tumor growth, they are ineffective against HER2-driven tumor growth because HER2 activates HER3 in a ligand-independent manner. In this study, we describe a novel HER3 monoclonal antibody (LJM716) that can neutralize multiple modes of HER3 activation, making it a superior candidate for clinical translation as a therapeutic candidate. LJM716 was a potent inhibitor of HER3/AKT phosphorylation and proliferation in HER2-amplified and NRG1-expressing cancer cells, and it displayed single-agent efficacy in tumor xenograft models. Combining LJM716 with agents that target HER2 or EGFR produced synergistic antitumor activity in vitro and in vivo. In particular, combining LJM716 with trastuzumab produced a more potent inhibition of signaling and cell proliferation than trastuzumab/pertuzumab combinations with similar activity in vivo. To elucidate its mechanism of action, we solved the structure of LJM716 bound to HER3, finding that LJM716 bound to an epitope, within domains 2 and 4, that traps HER3 in an inactive conformation. Taken together, our findings establish that LJM716 possesses a novel mechanism of action that, in combination with HER2- or EGFR-targeted agents, may leverage their clinical efficacy in ErbB-driven cancers.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/patologia , Neuregulina-1/metabolismo , Conformação Proteica/efeitos dos fármacos , Receptor ErbB-3/antagonistas & inibidores , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/mortalidade , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Feminino , Humanos , Immunoblotting , Imunoprecipitação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Fosforilação/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/imunologia , Receptor ErbB-2/metabolismo , Receptor ErbB-3/química , Receptor ErbB-3/imunologia , Receptor ErbB-3/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
14.
J Assoc Res Otolaryngol ; 11(1): 19-26, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19820996

RESUMO

Recent studies indicate that supporting cells play important roles in inner ear development, function, and regeneration after injury, but the molecular mechanisms underlying these processes remain poorly understood. Inducible cell-specific gene recombination in supporting cells could be a powerful tool to study the roles of specific molecules in these cells. Here we tested the feasibility, effectiveness, and cell specificity of inducible Cre-mediated gene recombination in the postnatal inner ear using mice that express an inducible form of Cre (CreER(T)) under the transcriptional control of the proteolipid protein (PLP) promoter. We assessed the pattern of tamoxifen-induced gene recombination in the inner ear using the ROSA26-LacZ reporter line, in which the beta-galactosidase gene is expressed only after Cre-mediated excision of a loxP-flanked stop cassette. Recombination was detected in cochlear inner phalangeal cells, supporting cells surrounding hair cells in vestibular maculae and cristae. Recombination also occurred in Schwann cells. We also found that this CreER(T) line can be used to increase and decrease the levels of expression of a trophic factor, brain-derived neurotrophic factor, specifically in supporting cells. These results show that PLP/CreER(T) mice are a powerful tool to dissect gene function in inner ear supporting cells.


Assuntos
Orelha Interna , Regulação da Expressão Gênica no Desenvolvimento , Neuroglia/fisiologia , Recombinação Genética/fisiologia , Células de Schwann/fisiologia , Animais , Antineoplásicos Hormonais , Fator Neurotrófico Derivado do Encéfalo/genética , Orelha Interna/citologia , Orelha Interna/embriologia , Orelha Interna/fisiologia , Genes Reporter , Células Ciliadas Vestibulares/fisiologia , Integrases/genética , Óperon Lac , Camundongos , Camundongos Transgênicos , Proteína Proteolipídica de Mielina/genética , Regiões Promotoras Genéticas/genética , Tamoxifeno , Transgenes/genética
15.
Nat Med ; 16(12): 1429-33, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21076395

RESUMO

Aberrant activation of the Hedgehog (Hh) pathway can drive tumorigenesis. To investigate the mechanism by which glioma-associated oncogene family zinc finger-1 (GLI1), a crucial effector of Hh signaling, regulates Hh pathway activation, we searched for GLI1-interacting proteins. We report that the chromatin remodeling protein SNF5 (encoded by SMARCB1, hereafter called SNF5), which is inactivated in human malignant rhabdoid tumors (MRTs), interacts with GLI1. We show that Snf5 localizes to Gli1-regulated promoters and that loss of Snf5 leads to activation of the Hh-Gli pathway. Conversely, re-expression of SNF5 in MRT cells represses GLI1. Consistent with this, we show the presence of a Hh-Gli-activated gene expression profile in primary MRTs and show that GLI1 drives the growth of SNF5-deficient MRT cells in vitro and in vivo. Therefore, our studies reveal that SNF5 is a key mediator of Hh signaling and that aberrant activation of GLI1 is a previously undescribed targetable mechanism contributing to the growth of MRT cells.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Tumor Rabdoide/genética , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas Cromossômicas não Histona/genética , Primers do DNA/genética , Proteínas de Ligação a DNA/genética , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Hibridização In Situ , Espectrometria de Massas , Camundongos , Análise em Microsséries , Proteína SMARCB1 , Fatores de Transcrição/genética , Proteína GLI1 em Dedos de Zinco
16.
J Neurosci Res ; 85(10): 2080-6, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17492793

RESUMO

Oligodendrocytes (OLs), the myelinating cells of the central nervous system, have specialized morphologies that subserve their function. Numerous qualitative studies suggest that OLs in different brain regions can differ in their morphological characteristics, including number of branches and internodes, internode length, etc. However, progress in identifying and characterizing the diverse types of OLs and their distribution in the brain has been made difficult by several technical constraints. Here we report a new strategy to analyze OL morphology with a high degree of quantitative power and throughput. We used confocal microscopy and three-dimensional cell tracing software to study OLs in the frontal cortex of mice expressing enhanced green fluorescent protein (eGFP) under the control of the proteolipid protein (Plp) gene promoter. Three-dimensional reconstructions were then used to analyze and quantify cell morphology, including total process length, total process surface area, total internode length, number of primary processes, number of branch points, and number of internodes. In addition, these reconstructions were subjected to Sholl analysis, which allows for the quantitative measure of OL arbor complexity. By using this approach, we identified and characterized a previously undescribed population of small OLs with a compact but complex morphology that includes numerous branching processes and a large number of short internodes. Our data suggest that other populations of OLs remain to be identified and characterized and that the tools we have developed could help in the process of characterizing them.


Assuntos
Lobo Frontal/citologia , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Microscopia Confocal , Oligodendroglia/ultraestrutura , Animais , Dendritos/ultraestrutura , Corantes Fluorescentes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Oligodendroglia/metabolismo
17.
Proc Natl Acad Sci U S A ; 104(19): 8131-6, 2007 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-17483467

RESUMO

Several psychiatric disorders are associated with white matter defects, suggesting that oligodendrocyte (OL) abnormalities underlie some aspects of these diseases. Neuregulin 1 (NRG1) and its receptor, erbB4, are genetically linked with susceptibility to schizophrenia and bipolar disorder. In vitro studies suggest that NRG1-erbB signaling is important for OL development. To test whether erbB signaling contributes to psychiatric disorders by regulating the structure or function of OLs, we analyzed transgenic mice in which erbB signaling is blocked in OLs in vivo. Here we show that loss of erbB signaling leads to changes in OL number and morphology, reduced myelin thickness, and slower conduction velocity in CNS axons. Furthermore, these transgenic mice have increased levels of dopamine receptors and transporters and behavioral alterations consistent with neuropsychiatric disorders. These results indicate that defects in white matter can cause alterations in dopaminergic function and behavior relevant to neuropsychiatric disorders.


Assuntos
Dopamina/fisiologia , Receptores ErbB/fisiologia , Transtornos Mentais/etiologia , Bainha de Mielina/fisiologia , Oligodendroglia/fisiologia , Transdução de Sinais/fisiologia , Anfetamina/farmacologia , Animais , Ansiedade/etiologia , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1 , Camundongos , Camundongos Transgênicos , Atividade Motora , Proteínas do Tecido Nervoso/fisiologia , Condução Nervosa , Neuregulina-1 , Diester Fosfórico Hidrolases/genética , Regiões Promotoras Genéticas , Receptor ErbB-4 , Comportamento Social
18.
Glia ; 54(6): 578-90, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16921523

RESUMO

Fibroblast growth factor 2 (FGF2) inhibits oligodendrocyte progenitor cell (OPC) differentiation during development and limits remyelination following chronic demyelination. The current study examines the mechanism underlying this effect of FGF2 expression on OPC differentiation. Retroviral lineage tracing demonstrates a direct in vivo effect of FGF receptor (FGFR) signaling on OPC differentiation. Retrovirus expressing a dominant negative FGFR construct (FGFRdn) and green fluorescent protein (GFP) was injected into the dorsal columns of postnatal day 7 (P7) mice followed by perfusion at P28. Among the GFP-labeled cells, FGFRdn retrovirus generated a higher proportion of oligodendrocytes than did control infections. This result from FGFRdn expression in OPCs was similar to the result obtained in our previous study using control retrovirus in FGF2 null mice. Further, in vitro retroviral siRNA expression distinguishes the function of specific FGFR isoforms in OPC responses to FGF2. FGF2 inhibition of OPC differentiation was effectively blocked by siRNA targeted to FGFR1, but not FGFR2 or FGFR3. We propose a model of direct FGF2 activation of FGFR1 leading to inhibition of OPC differentiation. This signaling pathway may be an important regulator of oligodendrocyte generation during myelination in development and may perturb OPC generation of remyelinating oligodendrocytes in demyelinating disease.


Assuntos
Diferenciação Celular/fisiologia , Linhagem da Célula/fisiologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Oligodendroglia/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Células-Tronco/metabolismo , Animais , Animais Recém-Nascidos , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Células Cultivadas , Fator 2 de Crescimento de Fibroblastos/metabolismo , Vetores Genéticos/genética , Proteínas de Fluorescência Verde , Inibidores do Crescimento/metabolismo , Inibidores do Crescimento/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/metabolismo , Regeneração Nervosa/fisiologia , Oligodendroglia/efeitos dos fármacos , RNA Interferente Pequeno/genética , Ratos , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Retroviridae/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Células-Tronco/efeitos dos fármacos , Transfecção/métodos
19.
Cell ; 127(1): 185-97, 2006 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-17018285

RESUMO

Embryonic multipotent neural precursors are exposed to extracellular signals instructing them to adopt different fates, neuronal or glial. However, the mechanisms by which precursors integrate these signals to make timely fate choices remained undefined. Here we show that direct nuclear signaling by a receptor tyrosine kinase inhibits the responses of precursors to astrocyte differentiation factors while maintaining their neurogenic potential. Upon neuregulin-induced activation and presenilin-dependent cleavage of ErbB4, the receptor's intracellular domain forms a complex with TAB2 and the corepressor N-CoR. This complex undergoes nuclear translocation and binds promoters of astrocytic genes, repressing their expression. Consistent with this observation, astrogenesis occurs precociously in ErbB4 knockout mice. Our studies define how presenilin-dependent nuclear signaling by a receptor tyrosine kinase directly regulates gene transcription and cell fate. This pathway could be of importance for neural stem cell biology and for understanding the pathogenesis of Alzheimer's disease.


Assuntos
Astrócitos/fisiologia , Encéfalo , Núcleo Celular/metabolismo , Receptores ErbB/metabolismo , Presenilinas/metabolismo , Transdução de Sinais/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Astrócitos/citologia , Encéfalo/citologia , Encéfalo/embriologia , Diferenciação Celular/fisiologia , Linhagem Celular , Receptores ErbB/genética , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Camundongos Knockout , Complexos Multiproteicos , Neuregulina-1/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , Presenilinas/genética , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Ratos , Receptor ErbB-4 , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Fatores de Tempo , Técnicas do Sistema de Duplo-Híbrido
20.
Neurobiol Dis ; 19(1-2): 171-82, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15837572

RESUMO

Repair of myelin damage in the adult CNS requires oligodendrocyte progenitor (OP) proliferation and subsequent differentiation into remyelinating oligodendrocytes. Platelet-derived growth factor (PDGF) and fibroblast growth factor-2 (FGF2) have been predicted to act individually and/or cooperatively to generate remyelinating oligodendrocytes. Analysis of PDGF alpha receptor (PDGF alpha R) heterozygous (+/-) mice indicates that PDGF alpha R expression modulates oligodendrocyte density in non-lesioned adult CNS. Analysis of cuprizone demyelination and recovery in PDGF alpha R+/- mice, FGF2 knockout (-/-) mice, and PDGF alpha R+/- FGF2-/- mice demonstrated that: (1) OP proliferation and oligodendrocyte regeneration is impaired in PDGF alpha R heterozygotes, (2) PDGF alpha R+/- and FGF2-/- deletions do not act cooperatively to impair OP amplification, (3) oligodendrocyte differentiation is more frequent in FGF2-/- mice, and (4) FGF2 deletion in combination with the PDGF alpha R+/- genotype rescues impaired oligodendrocyte regeneration of PDGF alpha R heterozygotes. These findings demonstrate distinct roles for PDGF and FGF2 in vivo in the context of a demyelinating disease with spontaneous remyelination.


Assuntos
Doenças Desmielinizantes/metabolismo , Fator 2 de Crescimento de Fibroblastos/fisiologia , Fibras Nervosas Mielinizadas/fisiologia , Oligodendroglia/fisiologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Transdução de Sinais/fisiologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/fisiologia , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Doenças Desmielinizantes/patologia , Fator 2 de Crescimento de Fibroblastos/deficiência , Fator 2 de Crescimento de Fibroblastos/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Fibras Nervosas Mielinizadas/patologia , Oligodendroglia/patologia , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/deficiência , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA