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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 165(4): 861-873, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37453564

RESUMO

BACKGROUND & AIMS: Small intestinal neuroendocrine tumor (SI-NET) is a rare disease, but its incidence has increased over the past 4 decades. Understanding the genetic risk factors underlying SI-NETs can help in disease prevention and may provide clinically beneficial markers for diagnosis. Here the results of the largest genome-wide association study of SI-NETs performed to date with 405 cases and 614,666 controls are reported. METHODS: Samples from 307 patients with SI-NETs and 287,137 controls in the FinnGen study were used for the identification of SI-NET risk-associated genetic variants. The results were also meta-analyzed with summary statistics from the UK Biobank (n = 98 patients with SI-NET and n = 327,529 controls). RESULTS: We identified 6 genome-wide significant (P < 5 × 10-8) loci associated with SI-NET risk, of which 4 (near SEMA6A, LGR5, CDKAL1, and FERMT2) are novel and 2 (near LTA4H-ELK and in KIF16B) have been reported previously. Interestingly, the top hit (rs200138614; P = 1.80 × 10-19) was a missense variant (p.Cys712Phe) in the LGR5 gene, a bona-fide marker of adult intestinal stem cells and a potentiator of canonical WNT signaling. The association was validated in an independent Finnish collection of 70 patients with SI-NETs, as well as in the UK Biobank exome sequence data (n = 92 cases and n = 392,814 controls). Overexpression of LGR5 p.Cys712Phe in intestinal organoids abolished the ability of R-Spondin1 to support organoid growth, indicating that the mutation perturbed R-Spondin-LGR5 signaling. CONCLUSIONS: Our study is the largest genome-wide association study to date on SI-NETs and reported 4 new associated genome-wide association study loci, including a novel missense mutation (rs200138614, p.Cys712Phe) in LGR5, a canonical marker of adult intestinal stem cells.


Assuntos
Neoplasias Intestinais , Tumores Neuroendócrinos , Adulto , Humanos , Tumores Neuroendócrinos/genética , Tumores Neuroendócrinos/patologia , Mutação de Sentido Incorreto , Estudo de Associação Genômica Ampla , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Receptores Acoplados a Proteínas G/genética , Cinesinas/genética
2.
Gastroenterology ; 158(5): 1389-1401.e10, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31930988

RESUMO

BACKGROUND & AIMS: In addition to the Notch and Wnt signaling pathways, energy metabolism also regulates intestinal stem cell (ISC) function. Tumor suppressor and kinase STK11 (also called LKB1) regulates stem cells and cell metabolism. We investigated whether loss of LKB1 alters ISC homeostasis in mice. METHODS: We deleted LKB1 from ISCs in mice using Lgr5-regulated CRE-ERT2 (Lkb1Lgr5-KO mice) and the traced lineages by using a CRE-dependent TdTomato reporter. Intestinal tissues were collected and analyzed by immunohistochemical and immunofluorescence analyses. We purified ISCs and intestinal progenitors using flow cytometry and performed RNA-sequencing analysis. We measured organoid-forming capacity and ISC percentages using intestinal tissues from Lkb1Lgr5-KO mice. We analyzed human Ls174t cells with knockdown of LKB1 or other proteins by immunoblotting, real-time quantitative polymerase chain reaction, and the Seahorse live-cell metabolic assay. RESULTS: Some intestinal crypts from Lkb1Lgr5-KO mice lost ISCs compared with crypts from control mice. However, most crypts from Lkb1Lgr5-KO mice contained functional ISCs that expressed increased levels of Atoh1 messenger RNA (mRNA), acquired a gene expression signature associated with secretory cells, and generated more cells in the secretory lineage compared with control mice. Knockdown of LKB1 in Ls174t cells induced expression of Atoh1 mRNA and a phenotype of increased mucin production; knockdown of ATOH1 prevented induction of this phenotype. The increased expression of Atoh1 mRNA after LKB1 loss from ISCs or Ls174t cells did not involve Notch or Wnt signaling. Knockdown of pyruvate dehydrogenase kinase 4 (PDK4) or inhibition with dichloroacetate reduced the up-regulation of Atoh1 mRNA after LKB1 knockdown in Ls174t cells. Cells with LKB1 knockdown had a reduced rate of oxygen consumption, which was partially restored by PDK4 inhibition with dichloroacetate. ISCs with knockout of LKB1 increased the expression of PDK4 and had an altered metabolic profile. CONCLUSIONS: LKB1 represses transcription of ATOH1, via PDK4, in ISCs, restricting their differentiation into secretory lineages. These findings provide a connection between metabolism and the fate determination of ISCs.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Metabolismo Energético/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Células-Tronco/fisiologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Ácido Dicloroacético/farmacologia , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Camundongos , Camundongos Knockout , Cultura Primária de Células , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil/antagonistas & inibidores , Piruvato Desidrogenase Quinase de Transferência de Acetil/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , RNA-Seq , Transcrição Gênica , Regulação para Cima/efeitos dos fármacos
3.
J Biol Chem ; 289(23): 16252-61, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24778181

RESUMO

The Cdk8 (cyclin-dependent kinase 8) module of Mediator integrates regulatory cues from transcription factors to RNA polymerase II. It consists of four subunits where Med12 and Med13 link Cdk8 and cyclin C (CycC) to core Mediator. Here we have investigated the contributions of the Cdk8 module subunits to transcriptional regulation using RNA interference in Drosophila cells. Genome-wide expression profiling demonstrated separation of Cdk8-CycC and Med12-Med13 profiles. However, transcriptional regulation by Cdk8-CycC was dependent on Med12-Med13. This observation also revealed that Cdk8-CycC and Med12-Med13 often have opposite transcriptional effects. Interestingly, Med12 and Med13 profiles overlapped significantly with that of the GATA factor Serpent. Accordingly, mutational analyses indicated that GATA sites are required for Med12-Med13 regulation of Serpent-dependent genes. Med12 and Med13 were also found to be required for Serpent-activated innate immunity genes in defense to bacterial infection. The results reveal a novel role for the Cdk8 module in Serpent-dependent transcription and innate immunity.


Assuntos
Quinase 8 Dependente de Ciclina/genética , Perfilação da Expressão Gênica , Imunidade Inata/genética , Transcrição Gênica , Animais , Drosophila , Reação em Cadeia da Polimerase , Interferência de RNA
4.
J Cell Sci ; 126(Pt 1): 263-73, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23132927

RESUMO

Cell migration and spreading is driven by actin polymerization and actin stress fibers. Actin stress fibers are considered to contain α-actinin crosslinkers and nonmuscle myosin II motors. Although several actin stress fiber subtypes have been identified in migrating and spreading cells, the degree of molecular diversity of their composition and the signaling pathways regulating fiber subtypes remain largely uncharacterized. In the present study we identify that dorsal stress fiber assembly requires α-actinin-1. Loss of dorsal stress fibers in α-actinin-1-depleted cells results in defective maturation of leading edge focal adhesions. This is accompanied by a delay in early cell spreading and slower cell migration without noticeable alterations in myosin light chain phosphorylation. In agreement with the unaltered myosin II activity, dorsal stress fiber trunks lack myosin II and are resistant to myosin II ATPase inhibition. Furthermore, the non-contractility of dorsal stress fibers is supported by the finding that Rac1 induces dorsal stress fiber assembly whereas contractile ventral stress fibers are induced by RhoA. Loss of dorsal stress fibers either by depleting α-actinin-1 or Rac1 results in a ß-actin accumulation at the leading edge in migrating and spreading cells. These findings molecularly specify dorsal stress fibers from other actin stress fiber subtypes. Furthermore, we propose that non-contractile dorsal stress fibers promote cell migration and early cell spreading through Rac1-induced actin polymerization.


Assuntos
Actinina/metabolismo , Movimento Celular/fisiologia , Fibras de Estresse/metabolismo , Cicatrização/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/genética , Imunofluorescência , Humanos , Camundongos , Miosinas/metabolismo , Cicatrização/genética
5.
EMBO Rep ; 14(8): 741-7, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23743448

RESUMO

Loss of primary cilia is frequently observed in tumour cells, including glioblastoma cells, and proposed to benefit tumour growth, but a causal link has not been established. Here, we show that CCRK (cell cycle-related kinase) and its substrate ICK (intestinal cell kinase) inhibit ciliogenesis. Depletion of CCRK leads to accumulation of ICK at ciliary tips, altered ciliary transport and inhibition of cell cycle re-entry in NIH3T3 fibroblasts. In glioblastoma cells with deregulated high levels of CCRK, its depletion restores cilia through ICK and an ICK-related kinase MAK, thereby inhibiting glioblastoma cell proliferation. These results indicate that inhibition of ciliogenesis might be a mechanism used by cancer cells to provide a growth advantage.


Assuntos
Neoplasias Encefálicas/genética , Cílios/enzimologia , Quinases Ciclina-Dependentes/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteínas Serina-Treonina Quinases/genética , Animais , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Cílios/patologia , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Camundongos , Células NIH 3T3 , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Quinase Ativadora de Quinase Dependente de Ciclina
6.
Proc Natl Acad Sci U S A ; 109(7): E388-97, 2012 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22308451

RESUMO

Although loss of epithelial integrity is a hallmark of advanced cancer, it remains poorly understood whether genetic alterations corrupting this integrity causally facilitate tumorigenesis. We show that conditional deletion of tumor suppressor gene Lkb1 (Par-4) in the mammary gland compromises epithelial integrity manifested by mislocalization of cell polarity markers, lateralization of tight junctions, deterioration of desmosomes and basement membrane (BM), and hyperbranching of the mammary ductal tree. We identify the desmosomal BM remodelling serine protease Hepsin as a key factor mediating Lkb1 loss-induced structural alterations in mammary epithelium and BM fragmentation. Although loss of Lkb1 alone does not promote mammary tumorigenesis, combination of Lkb1 deficiency with oncogenic c-Myc leads to dramatic acceleration in tumor formation. The results coupling Lkb1 loss-mediated epithelial integrity defects to mislocalization of serine protease Hepsin and to oncogenic synergy with c-Myc imply that Lkb1 loss facilitates oncogenic proliferation by releasing epithelial cells from structural BM boundaries.


Assuntos
Genes Supressores de Tumor , Glândulas Mamárias Animais/enzimologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Quinases Ativadas por AMP , Animais , Células Epiteliais/citologia , Feminino , Deleção de Genes , Genes myc , Glândulas Mamárias Animais/citologia , Camundongos , Proteínas Serina-Treonina Quinases/genética
7.
EMBO J ; 29(2): 469-81, 2010 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-19942859

RESUMO

The mobilization of metabolic energy from adipocytes depends on a tightly regulated balance between hydrolysis and resynthesis of triacylglycerides (TAGs). Hydrolysis is stimulated by beta-adrenergic signalling to PKA that mediates phosphorylation of lipolytic enzymes, including hormone-sensitive lipase (HSL). TAG resynthesis is associated with high-energy consumption, which when inordinate, leads to increased AMPK activity that acts to restrain hydrolysis of TAGs by inhibiting PKA-mediated activation of HSL. Here, we report that in primary mouse adipocytes, PKA associates with and phosphorylates AMPKalpha1 at Ser-173 to impede threonine (Thr-172) phosphorylation and thus activation of AMPKalpha1 by LKB1 in response to lipolytic signals. Activation of AMPKalpha1 by LKB1 is also blocked by PKA-mediated phosphorylation of AMPKalpha1 in vitro. Functional analysis of an AMPKalpha1 species carrying a non-phosphorylatable mutation at Ser-173 revealed a critical function of this phosphorylation for efficient release of free fatty acids and glycerol in response to PKA-activating signals. These results suggest a new mechanism of negative regulation of AMPK activity by PKA that is important for converting a lipolytic signal into an effective lipolytic response.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/enzimologia , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Lipólise , Proteínas Quinases Ativadas por AMP/genética , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Ácidos Graxos/metabolismo , Glicerol/metabolismo , Isoproterenol/farmacologia , Camundongos , Fosforilação , Mutação Puntual , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
8.
Carcinogenesis ; 34(10): 2409-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23722652

RESUMO

Peutz-Jeghers patients develop hamartomatous polyps and carcinomas of the gastrointestinal tract. Cyclooxygenase-2 accelerates polyp growth in Lkb1 (+/-) mice modelling Peutz-Jeghers polyposis. In this study, we aimed to evaluate the effect of the mutagenic carcinogen N-methylnitrosourea (MNU) on gastrointestinal tumourigenesis in Lkb1 (+/-) mice and to investigate the role of cyclooxygenase-2 on the tumourigenesis. We treated 40 Lkb1 (+/-) and 51 wild-type mice with MNU, 10 mice from both groups received the cyclooxygenase-2 inhibitor celecoxib. Carcinogen-treated Lkb1 (+/-) mice displayed worse survival (60%) than treated wild-type (100%, P = 0.028) or untreated Lkb1 (+/-) mice (92%, P = 0.045). Also, the gastrointestinal tumour burden was almost 10-fold higher in carcinogen-treated (2181 mm(3)) than in untreated (237 mm(3), P = 0.00045) Lkb1 (+/-) mice. Celecoxib was much less efficient in reducing tumourigenesis in MNU-treated mice (by 23%; 1686 mm(3)) than in untreated mice (76%; 58 mm(3)). Surprisingly, the increase in tumour burden in MNU-treated mice was not accompanied by consistent histological changes, with only a single focus of epithelial dysplasia noted. This study suggests that MNU promotes Peutz-Jeghers polyposis independently from the acceleration by cyclooxygenase-2.


Assuntos
Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinógenos/toxicidade , Metilnitrosoureia/toxicidade , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/patologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Quinases Ativadas por AMP , Animais , Carcinógenos/administração & dosagem , Celecoxib , Inibidores de Ciclo-Oxigenase 2/farmacologia , Modelos Animais de Doenças , Feminino , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Neoplasias Gastrointestinais/induzido quimicamente , Neoplasias Gastrointestinais/genética , Neoplasias Gastrointestinais/patologia , Metilnitrosoureia/administração & dosagem , Camundongos , Camundongos Knockout , Síndrome de Peutz-Jeghers/mortalidade , Pirazóis/farmacologia , Sulfonamidas/farmacologia
9.
J Cell Sci ; 124(Pt 3): 384-93, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21242312

RESUMO

Actin stress fiber assembly and contractility in nonmuscle motile cells requires phosphorylation of myosin regulatory light chain (MLC). Dephosphorylation and disassembly are mediated by MLC phosphatase, which is targeted to actin fibers by the association of its regulatory subunit MYPT1 with myosin phosphatase Rho-interacting protein (MRIP). In the present study, we identify the kinase NUAK2 as a second protein targeted by MRIP to actin fibers. Association of NUAK2 with MRIP increases MLC phosphorylation and promotes formation of stress fibers. This activity does not require the kinase activity of NUAK2 but is dependent on both MRIP and MYPT1, indicating that the NUAK2-MRIP association inhibits fiber disassembly and MYPT1-mediated MLC dephosphorylation. NUAK2 levels are strongly induced by stimuli increasing actomyosin fiber formation, and NUAK2 is required for fiber maintenance in exponentially growing cells, implicating NUAK2 in a positive-feedback loop regulating actin stress fibers independently of the MLC kinase Rho-associated protein kinase (ROCK). The identified MRIP-NUAK2 association reveals a novel mechanism for the maintenance of actin stress fibers through counteracting MYPT1 and, together with recent results, implicates the NUAK proteins as important regulators of the MLC phosphatase acting in both a kinase-dependent and kinase-independent manner.


Assuntos
Actinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fosfatase de Miosina-de-Cadeia-Leve/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fibras de Estresse/metabolismo , Linhagem Celular Tumoral , Humanos , Contração Muscular , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/metabolismo , Fosforilação , Ligação Proteica , Técnicas do Sistema de Duplo-Híbrido , Quinases Associadas a rho/metabolismo
10.
Nucleic Acids Res ; 39(12): 5025-35, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21385826

RESUMO

The relevance of serine 5 phosphorylation of RNA polymerase II carboxy-terminal domain during initiation has been difficult to determine in mammalian cells as no general in vivo Ser5 kinase has been identified. Here, we demonstrate that deletion of the TFIIH kinase subunit Mat1 in mouse fibroblasts leads to dramatically reduced Pol II Ser5 phosphorylation. This is associated with defective capping and reduced Ser2 phosphorylation, decreased Pol II progression into elongation and severely attenuated transcription detected through analysis of nascent mRNAs, establishing a general requirement for mammalian Mat1 in transcription. Surprisingly, the general defect in Pol II transcription in Mat1(-/-) fibroblasts is not reflected in the majority of steady-state mRNAs. This indicates widespread stabilization of mRNAs and points to the existence of a regulatory mechanism to stabilize mRNAs following transcriptional attenuation, thus revealing a potential caveat in similar studies limited to analysis of steady-state mRNAs.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/fisiologia , RNA Polimerase II/metabolismo , RNA Mensageiro/metabolismo , Serina/metabolismo , Transcrição Gênica , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Proteínas de Ciclo Celular , Fibroblastos/metabolismo , Deleção de Genes , Proteínas de Choque Térmico HSP70/biossíntese , Proteínas de Choque Térmico HSP70/genética , Camundongos , Fosforilação , Proteínas Quinases/química , Estrutura Terciária de Proteína , Subunidades Proteicas/genética , Subunidades Proteicas/fisiologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Proteínas Proto-Oncogênicas c-fos/genética , Capuzes de RNA/análise , RNA Polimerase II/química , Splicing de RNA , Estabilidade de RNA , RNA Mensageiro/biossíntese , Fator de Transcrição TFIIH/metabolismo , Fatores de Transcrição
11.
Dis Model Mech ; 16(3)2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36804687

RESUMO

Intestinal epithelial organoids recapitulate many of the in vivo features of the intestinal epithelium, thus representing excellent research models. Morphology of the organoids based on light-microscopy images is used as a proxy to assess the biological state of the intestinal epithelium. Currently, organoid classification is manual and, therefore, subjective and time consuming, hampering large-scale quantitative analyses. Here, we describe Tellu, an object-detector algorithm trained to classify cultured intestinal organoids. Tellu was trained by manual annotation of >20,000 intestinal organoids to identify cystic non-budding organoids, early organoids, late organoids and spheroids. Tellu can also be used to quantify the relative organoid size, and can classify intestinal organoids into these four subclasses with accuracy comparable to that of trained scientists but is significantly faster and without bias. Tellu is provided as an open, user-friendly online tool to benefit the increasing number of investigations using organoids through fast and unbiased organoid morphology and size analysis.


Assuntos
Mucosa Intestinal , Intestinos , Organoides , Algoritmos
12.
Cell Metab ; 5(2): 129-42, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17276355

RESUMO

The Cdk7/cyclin H/ménage-à-trois 1 (MAT1) heterotrimer has proposed functions in transcription as the kinase component of basal transcription factor TFIIH and is activated in adult hearts by Gq-, calcineurin-, and biomechanical stress-dependent pathways for hypertrophic growth. Using cardiac-specific Cre, we have ablated MAT1 in myocardium. Despite reduced Cdk7 activity, MAT1-deficient hearts grew normally, but fatal heart failure ensued at 6-8 weeks. By microarray profiling, quantitative RT-PCR, and western blotting at 4 weeks, genes for energy metabolism were found to be suppressed selectively, including targets of peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1). Cardiac metabolic defects were substantiated in isolated perfused hearts and isolated mitochondria. In culture, deleting MAT1 with Cre disrupted PGC-1 function: PGC-1alpha failed to activate PGC-1-responsive promoters and nuclear receptors, GAL4-PGC-1alpha was functionally defective, and PGC-1beta was likewise deficient. PGC-1 bound to both MAT1 and Cdk7 in coprecipitation assays. Thus, we demonstrate a requirement for MAT1 in the operation of PGC-1 coactivators that control cell metabolism.


Assuntos
Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Apoptose , Cardiomiopatias/genética , Cardiomiopatias/patologia , Proteínas de Ciclo Celular , Sobrevivência Celular , Quinases Ciclina-Dependentes/metabolismo , Receptor com Domínio Discoidina 1 , Deleção de Genes , Regulação da Expressão Gênica , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Miocárdio/enzimologia , Miocárdio/patologia , Fosforilação , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Estrogênio/metabolismo , Fatores de Transcrição/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
13.
Dev Biol ; 357(1): 259-68, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21736876

RESUMO

ß-catenin has well-established functions in cell growth and differentiation as part of the Wnt signaling pathway and in regulation of cellular adhesion with E-cadherin. Here we studied its significance in midbrain development by temporally controlled deletion of ß-catenin allowing simultaneous analysis of complete (ß-cat-null) and partial (ß-cat-low) loss-of-function phenotypes in progenitor cells. ß-cat-null cells did not contain centrosomes or a microtubule network and were unpolarized forming delaminated bulges. ß-cat-low cells displayed defects in the orientation of the mitotic spindle, increased asymmetric cell divisions and premature differentiation in absence of alterations in polarity or adhesion. The spindle defect was associated with decreased centrosomal S33/S34/T41 phosphorylated ß-catenin (p-ß-cat) and centrosomal and microtubule defects. Interestingly, neural progenitor cells in mice expressing only unphosphorylatable ß-catenin share several phenotypes with ß-catenin loss-of-function mice with defects in microtubules and polarity. The results demonstrate a novel function for p-ß-cat in maintaining neuroepithelial integrity and suggest that centrosomal p-ß-cat is required to maintain symmetric cleavages and polarity in neural progenitors.


Assuntos
Centrossomo/metabolismo , Mesencéfalo/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Neurônios/metabolismo , beta Catenina/metabolismo , Animais , Polaridade Celular/fisiologia , Cães , Embrião de Mamíferos/metabolismo , Feminino , Mesencéfalo/metabolismo , Camundongos , Camundongos Endogâmicos , Células-Tronco Neurais/citologia , Neurônios/citologia , Fosforilação , beta Catenina/análise
14.
Int J Cancer ; 131(5): 1032-41, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22034055

RESUMO

Cyclooxygenase-2 (Cox-2) expression is a marker of reduced survival in gastric cancer patients, and inhibition of Cox-2 suppresses gastrointestinal carcinogenesis in experimental animal models. To investigate the role of Cox-2 in gastric carcinogenesis in vivo, we utilized trefoil factor 1 (Tff1) deficient mice, which model the neoplastic process of the stomach by developing gastric adenomas with full penetrance. These tumors express Cox-2 protein and mRNA, and we have now investigated the effects of genetic deletion of the mouse Cox-2 gene [also known as prostaglandin-endoperoxide synthase 2 (Ptgs2)] and a Cox-2 selective drug celecoxib. Our results show that genetic deletion of Cox-2 in the Tff1 deleted background resulted in reduced adenoma size and ulceration with a chronic inflammatory reaction at the site of the adenoma. To characterize the effect of Cox-2 inhibition in more detail, mice that had already developed an adenoma were fed with celecoxib for 8-14 weeks, which resulted in disruption of the adenoma that ranged from superficial erosion to deep ulcerated destruction accompanied with chronic inflammation. Importantly, mice fed with celecoxib for 16 weeks, followed by control food for 9 weeks, redeveloped a complete adenoma with no detectable inflammatory process. Finally, we determined the identity of the Cox-2 expressing cells and found them to be fibroblasts. Our results show that inhibition of Cox-2 is sufficient to reversibly disrupt gastric adenomas in mice.


Assuntos
Adenoma/prevenção & controle , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/fisiologia , Peptídeos/fisiologia , Pirazóis/uso terapêutico , Neoplasias Gástricas/prevenção & controle , Sulfonamidas/uso terapêutico , Adenoma/metabolismo , Adenoma/patologia , Animais , Apoptose , Western Blotting , Celecoxib , Proliferação de Células , Feminino , Imunofluorescência , Mucosa Gástrica/metabolismo , Técnicas Imunoenzimáticas , Masculino , Camundongos , Camundongos Knockout , Estômago/patologia , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Fator Trefoil-1
15.
Nat Methods ; 6(1): 75-7, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19079255

RESUMO

There is an increasing demand for network analysis of protein-protein interactions (PPIs). We introduce a web-based protein interaction network analysis platform (PINA), which integrates PPI data from six databases and provides network construction, filtering, analysis and visualization tools. We demonstrated the advantages of PINA by analyzing two human PPI networks; our results suggested a link between LKB1 and TGFbeta signaling, and revealed possible competitive interactors of p53 and c-Jun.


Assuntos
Mapeamento de Interação de Proteínas/métodos , Proteínas/análise , Proteínas/metabolismo , Bases de Dados Genéticas , Humanos , Ligação Proteica , Especificidade por Substrato
16.
Clin Cancer Res ; 28(1): 227-237, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34667030

RESUMO

PURPOSE: Mutations in STK11 (LKB1) occur in 17% of lung adenocarcinoma (LUAD) and drive a suppressive (cold) tumor immune microenvironment (TIME) and resistance to immunotherapy. The mechanisms underpinning the establishment and maintenance of a cold TIME in LKB1-mutant LUAD remain poorly understood. In this study, we investigated the role of the LKB1 substrate AMPK in immune evasion in human non-small cell lung cancer (NSCLC) and mouse models and explored the mechanisms involved. EXPERIMENTAL DESIGN: We addressed the role of AMPK in immune evasion in NSCLC by correlating AMPK phosphorylation and immune-suppressive signatures and by deleting AMPKα1 (Prkaa1) and AMPKα2 (Prkaa2) in a KrasG12D -driven LUAD. Furthermore, we dissected the molecular mechanisms involved in immune evasion by comparing gene-expression signatures, AMPK activity, and immune infiltration in mouse and human LUAD and gain or loss-of-function experiments with LKB1- or AMPK-deficient cell lines. RESULTS: Inactivation of both AMPKα1 and AMPKα2 together with Kras activation accelerated tumorigenesis and led to tumors with reduced infiltration of CD8+/CD4+ T cells and gene signatures associated with a suppressive TIME. These signatures recapitulate those in Lkb1-deleted murine LUAD and in LKB1-deficient human NSCLC. Interestingly, a similar signature is noted in human NSCLC with low AMPK activity. In mechanistic studies, we find that compromised LKB1 and AMPK activity leads to attenuated antigen presentation in both LUAD mouse models and human NSCLC. CONCLUSIONS: The results provide evidence that the immune evasion noted in LKB1-inactivated lung cancer is due to subsequent inactivation of AMPK and attenuation of antigen presentation.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas Quinases Ativadas por AMP/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adenocarcinoma de Pulmão/genética , Animais , Apresentação de Antígeno , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Evasão da Resposta Imune , Neoplasias Pulmonares/patologia , Camundongos , Microambiente Tumoral
17.
Chromosoma ; 119(4): 415-24, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20237935

RESUMO

During mitosis, all chromosomes must attach to microtubules of the mitotic spindle to ensure correct chromosome segregation. Microtubule attachment occurs at specialized structures at the centromeric region of chromosomes, called kinetochores. These kinetochores can generate microtubule attachments through capture of centrosome-derived microtubules, but in addition, they can generate microtubules themselves, which are subsequently integrated with centrosome-derived microtubules to form the mitotic spindle. Here, we have performed a large scale RNAi screen and identify cyclin G-associated kinase (GAK) as a novel regulator of microtubule generation at kinetochores/chromatin. This function of GAK requires its C-terminal J-domain, which is essential for clathrin recycling from endocytic vesicles. Consistently, cells lacking GAK show strongly reduced levels of clathrin on the mitotic spindle, and reduction of clathrin levels also inhibits microtubule generation at kinetochores/chromosomes. Finally, we present evidence that association of clathrin with the spindle is promoted by a signal coming from the chromosomes. These results identify a role for GAK and clathrin in microtubule outgrowth from kinetochores/chromosomes and suggest that GAK acts through clathrin to control microtubule outgrowth around chromosomes.


Assuntos
Clatrina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/metabolismo , Linhagem Celular Tumoral , Centrômero/metabolismo , Segregação de Cromossomos/fisiologia , Cromossomos Humanos/genética , Cromossomos Humanos/fisiologia , Cromossomos Humanos/ultraestrutura , Clatrina/genética , Clatrina/fisiologia , Células HeLa , Humanos , Cinetocoros/fisiologia , Microtúbulos/ultraestrutura , Mitose , RNA Interferente Pequeno , Fuso Acromático/genética , Tubulina (Proteína)/metabolismo
18.
Am J Pathol ; 176(5): 2467-76, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20363912

RESUMO

Gastrointestinal hamartomatous polyps in the Peutz-Jeghers cancer predisposition syndrome and its mouse model (Lkb1(+/-)) are presumed to contain all cell types native to the site of their occurrence. This study aimed to explore the pathogenesis of Peutz-Jeghers syndrome polyposis by characterizing cell types and differentiation of the epithelium of gastric polyps and predisposed mucosa. Both antral and fundic polyps were characterized by a deficit of pepsinogen C-expressing differentiated gland cells (antral gland, mucopeptic, and chief cells); in large fundic polyps, parietal cells were also absent. Gland cell loss was associated with an increase in precursor neck cells, an expansion of the proliferative zone, and an increase in smooth muscle alpha-actin expressing myofibroblasts in the polyp stroma. Lack of pepsinogen C-positive gland cells identified incipient polyps, and even the unaffected mucosa of young predisposed mice displayed an increase in pepsinogen C negative glands (25%; P = 0045). In addition, in small intestinal polyps, gland cell differentiation was defective, with the absence of Paneth cells. There were no signs of metaplastic differentiation in any of the tissues studied, and both the gastric and small intestinal defects were seen in Lkb1(+/-) mice, as well as polyps from patients with Peutz-Jeghers syndrome. These results identify impaired epithelial differentiation as the earliest pathological sign likely to contribute to tumorigenesis in individuals with inherited Lkb1 mutations.


Assuntos
Mucosa Gástrica/patologia , Mutação , Síndrome de Peutz-Jeghers/metabolismo , Animais , Diferenciação Celular , Proliferação de Células , Humanos , Imuno-Histoquímica , Pólipos Intestinais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Anatômicos , Análise de Sequência com Séries de Oligonucleotídeos , Pepsinogênio C/química
19.
Exp Cell Res ; 316(5): 762-74, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20036235

RESUMO

p27Kip1 (p27) tumour suppressor protein is regulated by multiple mechanisms including its turnover, localization and complex formation with its key targets, cyclin-dependent kinases (CDK) and cyclins. We have earlier shown that p27 exists in cells in a form that lacks cyclin/CDK interactions (hence non-CDK, p27(NCDK)) but the nature of p27(NCDK) has remained unresolved. Here we demonstrate that the epitope recognized by the p27(NCDK)-specific antibody resides in the p27 CDK-interaction domain and that p27(NCDK) is regulated by the balance of CDK inhibitors and cyclin-CDK complexes. We find that signalling by cellular growth promoting pathways, like phosphoinositol 3-kinase (PI3K) and specifically Akt/PKB kinase, inversely correlates with p27(NCDK) levels whereas total p27 levels are unaffected. p27(NCDK), but not total p27, is increased by cellular perturbations such as hyperosmotic and metabolic stress and activation of AMP-activated protein kinase (AMPK). By using AMPK catalytic subunit proficient and deficient cells we further demonstrate that the AMPK pathway governs p27(NCDK) responses to metabolic stress and PI3K inhibition. These results indicate that p27(NCDK) is a sensitive marker for both cell stress and proliferation over and above p27 and is regulated by Akt/PKB and AMPK pathways.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Biomarcadores/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Estresse Fisiológico , Proteínas Quinases Ativadas por AMP/genética , Sequência de Aminoácidos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/metabolismo , Animais , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p27/genética , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/metabolismo , Humanos , Hipoglicemiantes/metabolismo , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Peptídeos/genética , Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Ribonucleotídeos/metabolismo
20.
Nat Commun ; 12(1): 7186, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34893605

RESUMO

How tubular organs elongate is poorly understood. We found that attenuated ciliary Hedgehog signaling in the gut wall impaired patterning of the circumferential smooth muscle and inhibited proliferation and elongation of developing intestine and esophagus. Similarly, ablation of gut-wall smooth muscle cells reduced lengthening. Disruption of ciliary Hedgehog signaling or removal of smooth muscle reduced residual stress within the gut wall and decreased activity of the mechanotransductive effector YAP. Removing YAP in the mesenchyme also reduced proliferation and elongation, but without affecting smooth muscle formation, suggesting that YAP interprets the smooth muscle-generated force to promote longitudinal growth. Additionally, we developed an intestinal culture system that recapitulates the requirements for cilia and mechanical forces in elongation. Pharmacologically activating YAP in this system restored elongation of cilia-deficient intestines. Thus, our results reveal that ciliary Hedgehog signaling patterns the circumferential smooth muscle to generate radial mechanical forces that activate YAP and elongate the gut.


Assuntos
Trato Gastrointestinal , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Transdução de Sinais/fisiologia , Animais , Proliferação de Células , Cílios/metabolismo , Mesoderma/metabolismo , Camundongos , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA