Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Mol Reprod Dev ; 88(7): 482-489, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33973295

RESUMO

Glands of the uterus are essential for the establishment of pregnancy in mice and their products regulate embryo implantation and stromal cell decidualization critical for pregnancy establishment. Forkhead box A2 (FOXA2) is expressed specifically in the glands and a critical regulator of their differentiation, development and function. Progesterone and FOXA2 regulate members of a serine proteinase gene family (Prss28 and Prss29). Here, CRISPR-Cas9 genome-editing was used to create mice with a heterozygous or homozygous deletion of Prss28 or/and Prss29 to determine their biological roles in uterine function. Female mice lacking Prss28 and Prss29 or both developed normally and were fertile without alterations in uterine histoarchitecture, uterine gland number, or and gene expression. Thus, Prss28 and Prss29 are dispensable for female fertility and do not impact endometrial gland development or uterine function mice.


Assuntos
Sistemas CRISPR-Cas/genética , Técnicas de Inativação de Genes/métodos , Serina Endopeptidases/genética , Animais , Endométrio/metabolismo , Feminino , Edição de Genes/métodos , Masculino , Camundongos , Camundongos Knockout , Gravidez , Deleção de Sequência/fisiologia , Serina Endopeptidases/metabolismo , Útero/metabolismo
2.
Nat Commun ; 9(1): 4394, 2018 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-30349055

RESUMO

Heterozygous deletion of chromosome 17p (17p) is one of the most frequent genomic events in human cancers. Beyond the tumor suppressor TP53, the POLR2A gene encoding the catalytic subunit of RNA polymerase II (RNAP2) is also included in a ~20-megabase deletion region of 17p in 63% of metastatic castration-resistant prostate cancer (CRPC). Using a focused CRISPR-Cas9 screen, we discovered that heterozygous loss of 17p confers a selective dependence of CRPC cells on the ubiquitin E3 ligase Ring-Box 1 (RBX1). RBX1 activates POLR2A by the K63-linked ubiquitination and thus elevates the RNAP2-mediated mRNA synthesis. Combined inhibition of RNAP2 and RBX1 profoundly suppress the growth of CRPC in a synergistic manner, which potentiates the therapeutic effectivity of the RNAP2 inhibitor, α-amanitin-based antibody drug conjugate (ADC). Given the limited therapeutic options for CRPC, our findings identify RBX1 as a potentially therapeutic target for treating human CRPC harboring heterozygous deletion of 17p.


Assuntos
Cromossomos Humanos Par 17/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , RNA Polimerase II/metabolismo , Apoptose/genética , Apoptose/fisiologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Cromossomos Humanos Par 17/genética , Humanos , Immunoblotting , Imuno-Histoquímica , Imunoprecipitação , Masculino , Neoplasias de Próstata Resistentes à Castração/genética , RNA Polimerase II/genética , Deleção de Sequência/genética , Deleção de Sequência/fisiologia , Transcrição Gênica/genética , Transcrição Gênica/fisiologia , Ubiquitina-Proteína Ligases
3.
Mitochondrial DNA A DNA Mapp Seq Anal ; 27(4): 2520-2, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26017042

RESUMO

The COII/tRNA(Lys) intergenic 9-bp deletion is one of the most commonly studied human mitochondrial DNA (mtDNA) polymorphisms. It consists of the loss of one of two tandemly repeated copies of the sequence CCCCCTCTA from a non-coding region located between cytochrome oxidase II (COII) and tRNA(Lys) gene. Most recently, case-control studies have shown a positive association between this deletion with hepatocellular cancer. In this study, we first performed a detailed analysis between this deletion and clinical diseases; moreover, we took the phylogenetic approach to examine the pathogenicity status of 9-bp deletion.


Assuntos
Carcinoma Hepatocelular/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , Genoma Mitocondrial/genética , Neoplasias Hepáticas/genética , RNA de Transferência de Lisina/genética , Deleção de Sequência/genética , DNA Mitocondrial/genética , Humanos , Conformação de Ácido Nucleico , Filogenia , Deleção de Sequência/fisiologia
4.
Biochim Biophys Acta ; 1852(8): 1561-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25912737

RESUMO

A pathological hallmark of Alzheimer disease (AD) is the accumulation of misfolded hyperphosphorylated microtubule-associated protein Tau within neurons, forming neurofibrillary tangles and leading to synaptic dysfunction and neuronal death. Here we study sequence-dependent abnormal aggregation of human fragment Tau244-372 in an inducible cell model. As evidenced by confocal laser scanning microscopy, Western blot, and immunogold electron microscopy, fibril-forming motifs are essential and sufficient for abnormal aggregation of Tau244-372 in SH-SY5Y neuroblastoma cells induced by Congo red: when its two fibril-forming segments PHF6 and PHF6* are deleted, Tau244-372 does lose its ability to form fibrils in SH-SY5Y cells, and the replacement of PHF6 and PHF6* with an unrelated amyloidogenic sequence IFQINS from human lysozyme does rescue the fibril-forming ability of Tau244-372 in SH-SY5Y cells. By contrast, insertion of a non-fibril forming peptide GGGGGG does not drive the disabled Tau244-372 to misfold in SH-SY5Y cells. Furthermore, as revealed by quantum dots based probes combined with annexin V staining, annexin V-FITC apoptosis detection assay, and immunofluorescence, fibril-forming motifs are essential and sufficient for early apoptosis of living SH-SY5Y cells induced by abnormal aggregation of Tau244-372. Our results suggest that fibril-forming motifs could be the determinants of Tau protein tending to misfold in living cells, thereby inducing neuronal apoptosis and causing the initiation and development of AD.


Assuntos
Agregação Patológica de Proteínas , Domínios e Motivos de Interação entre Proteínas , Proteínas tau/química , Proteínas tau/genética , Proteínas tau/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Sequência de Aminoácidos/fisiologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Modelos Biológicos , Mutagênese Insercional/fisiologia , Emaranhados Neurofibrilares/genética , Emaranhados Neurofibrilares/metabolismo , Neurônios/metabolismo , Neurônios/patologia , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Agregação Patológica de Proteínas/genética , Agregação Patológica de Proteínas/metabolismo , Dobramento de Proteína , Domínios e Motivos de Interação entre Proteínas/genética , Deleção de Sequência/fisiologia , Transfecção , Regulação para Cima/genética
5.
Andrologia ; 47(10): 1160-5, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25597551

RESUMO

Kallmann syndrome (KS) is a genetically heterogeneous disease characterised by hypogonadotrophic hypogonadism in association with anosmia or hyposmia. This condition affects 1 in 10 000 men and 1 in 50,000 women. Defects in seventeen genes including KAL1 gene contribute to the molecular basis of KS. We report the clinical characteristics, molecular causes and treatment outcome of two Chinese brothers with KS and X-linked ichthyosis. The phenotypes of the patients were characterised by bilateral cryptorchidism, unilateral renal agenesis in one patient but normal kidney development in another. The patients had low serum testosterone, follicle-stimulating hormone and luteinising hormone levels and a blunt response to the gonadotrophin-releasing hormone stimulation test. After human chorionic gonadotrophin treatment, the serum testosterone levels were normalized, and the pubic hair, penis length and testicular volumes were greatly improved in both of the patients. The two affected siblings had the same novel deletion at Xp22.3 including exons 9-14 of KAL1 gene and entire STS gene. Our study broadens the mutation spectrum in the KAL1 gene associated with KS and facilitates the genetic diagnosis and counselling for KS.


Assuntos
Proteínas da Matriz Extracelular/genética , Ictiose Ligada ao Cromossomo X/genética , Síndrome de Kallmann/genética , Proteínas do Tecido Nervoso/genética , Deleção de Sequência/genética , Esteril-Sulfatase/genética , Proteínas da Matriz Extracelular/fisiologia , Homozigoto , Humanos , Masculino , Proteínas do Tecido Nervoso/fisiologia , Linhagem , Deleção de Sequência/fisiologia , Irmãos , Esteril-Sulfatase/fisiologia
6.
Oncogene ; 32(7): 827-36, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-22484427

RESUMO

The tumor suppressor protein, p53 is one of the most important cellular defences against malignant transformation. In response to cellular stressors p53 can induce apoptosis, cell cycle arrest or senescence as well as aid in DNA repair. Which p53 function is required for tumor suppression is unclear. The proline-rich domain (PRD) of p53 (residues 58-101) has been reported to be essential for the induction of apoptosis. To determine the importance of the PRD in tumor suppression in vivo we previously generated a mouse containing a 33-amino-acid deletion (residues 55-88) in p53 (mΔpro). We showed that mΔpro mice are protected from T-cell tumors but not late-onset B-cell tumors. Here, we characterize the functionality of the PRD and show that it is important for mediating the p53 response to DNA damage induced by γ-radiation, but not the p53-mediated responses to Ha-Ras expression or oxidative stress. We conclude that the PRD is important for receiving incoming activating signals. Failure of PRD mutants to respond to the activating signaling produced by DNA damage leads to impaired downstream signaling, accumulation of mutations, which potentially leads to late-onset tumors.


Assuntos
Domínios Proteicos Ricos em Prolina/fisiologia , Radiação Ionizante , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo , Animais , Linfócitos B/metabolismo , Linfócitos B/fisiologia , Linfócitos B/efeitos da radiação , Transformação Celular Neoplásica/genética , Células Cultivadas , Dano ao DNA/genética , Dano ao DNA/efeitos da radiação , Embrião de Mamíferos , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Células-Tronco Embrionárias/efeitos da radiação , Camundongos , Camundongos Knockout , Modelos Biológicos , Prolina/química , Prolina/fisiologia , Domínios Proteicos Ricos em Prolina/genética , Domínios Proteicos Ricos em Prolina/efeitos da radiação , Deleção de Sequência/fisiologia , Estresse Fisiológico/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/efeitos da radiação
7.
J Endocrinol Invest ; 35(2): 124-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22490989

RESUMO

AIM: To perform a genetic screening for the multiple endocrine neoplasia type 1 (MEN1) gene mutations in patients affected by an apparently sporadic form of the disease, referred to an internal medicine unit of a large general hospital. SUBJECTS AND METHODS: In a group of 12 consecutive patients presenting clinical features of MEN type 1 syndrome, we performed a genetic screening for germline MEN1 gene mutations, including complete sequencing of the coding region (exons 2 to 10) and multiplex ligation-dependent probe amplification analysis for large deletion detection. RESULTS: Among these patients affected by apparently sporadic MEN type 1 syndrome, a targeted clinical history could detect indirect support for a diagnosis of familial condition only in 2 cases. The genetic screening identified pathogenic germline MEN1 gene mutations in 3 patients (25%). A previously unknown 18 base-pair deletion within exon 3, c.564_581delCAATGGGGAGCAGACAGC, resulting in loss of 6 amino acids (pAsp189_Ala194del), was found in heterozygosis in a woman affected by primary hyperparathyroidism and multifocal pancreatic neoplasia. CONCLUSIONS: Our results underscore the importance of performing genetic testing also in apparently sporadic MEN1 patients and extend the list of molecular variants leading to inactivation of the MEN1 gene.


Assuntos
Neoplasia Endócrina Múltipla Tipo 1/genética , Proteínas Proto-Oncogênicas/genética , Deleção de Sequência , Adenoma/etiologia , Adenoma/genética , Adulto , Estudos de Coortes , Análise Mutacional de DNA , Feminino , Humanos , Insulinoma/etiologia , Insulinoma/genética , Itália , Masculino , Pessoa de Meia-Idade , Neoplasia Endócrina Múltipla Tipo 1/complicações , Neoplasias Pancreáticas/etiologia , Neoplasias Pancreáticas/genética , Neoplasias das Paratireoides/etiologia , Neoplasias das Paratireoides/genética , Deleção de Sequência/fisiologia , Síndrome
8.
Traffic ; 13(2): 218-33, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21995724

RESUMO

In vertebrates, the nuclear pore complex (NPC), the gate for transport of macromolecules between the nucleus and the cytoplasm, consists of approximately 30 different nucleoporins (Nups). The Nup and SUMO E3-ligase Nup358/RanBP2 are the major components of the cytoplasmic filaments of the NPC. In this study, we perform a structure-function analysis of Nup358 and describe its role in nuclear import of specific proteins. In a screen for nuclear proteins that accumulate in the cytoplasm upon Nup358 depletion, we identified proteins that were able to interact with Nup358 in a receptor-independent manner. These included the importin α/ß-cargo DBC-1 (deleted in breast cancer 1) and DMAP-1 (DNA methyltransferase 1 associated protein 1). Strikingly, a short N-terminal fragment of Nup358 was sufficient to promote import of DBC-1, whereas DMAP-1 required a larger portion of Nup358 for stimulated import. Neither the interaction of RanGAP with Nup358 nor its SUMO-E3 ligase activity was required for nuclear import of all tested cargos. Together, Nup358 functions as a cargo- and receptor-specific assembly platform, increasing the efficiency of nuclear import of proteins through various mechanisms.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Carioferinas/metabolismo , Chaperonas Moleculares/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/metabolismo , Transporte Proteico/fisiologia , Proteínas Adaptadoras de Transdução de Sinal , Núcleo Celular/metabolismo , Citoplasma/metabolismo , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Células HeLa , Humanos , Carioferinas/genética , Chaperonas Moleculares/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Membrana Nuclear/metabolismo , Sinais de Localização Nuclear/genética , Poro Nuclear/metabolismo , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Mutação Puntual/fisiologia , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , RNA Interferente Pequeno/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Deleção de Sequência/fisiologia , Transfecção , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Fatores Estimuladores Upstream/genética , Fatores Estimuladores Upstream/metabolismo , alfa Carioferinas/genética , alfa Carioferinas/metabolismo , beta Carioferinas/genética , beta Carioferinas/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana/metabolismo
9.
Blood ; 119(13): 2981-90, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22174151

RESUMO

Deletion of chromosomal region 13q14 represents the most common genetic aberration in B-cell chronic lymphocytic leukemia (CLL). 13q14 deletions are commonly large and heterogeneous in size and affect multiple genes. We recently found that targeted deletion in mice of the 0.11 megabase (mb)-long minimal deleted region (MDR) encompassing the DLEU2/miR-15a/16-1 cluster recapitulates the spectrum of CLL-associated lymphoproliferations in humans, including CLL, CD5(+) monoclonal B-cell lymphocytosis, and CD5(-) non-Hodgkin lymphomas. In the present study, we demonstrate that additional deletion of the 0.69-mb large genomic region telomeric to the MDR called the common deleted region (CDR) changed the spectrum of lymphoproliferations developing in CDR- versus MDR-deleted mice in that the number of CLL among B-cell lymphoproliferations was significantly elevated in the former. In addition, CDR-deleted mice seemed to succumb to their disease faster than MDR-deleted mice. Comparing HCDR3 regions of CD5(+) lymphoproliferations derived from this and published CLL mouse models, 44% (29 of 66) of junctions could be assigned to 8 sets of highly similar HCDR3 regions, demonstrating that CLL developing in mice frequently expresses almost identical, stereotypic Ag receptors. These results suggest that the size of 13q14 deletions influences the phenotype of the developing lymphoproliferations and potentially the severity of disease, suggesting a tumor-suppressor function for genetic elements in addition to DLEU2/miR-15a/16-1.


Assuntos
Cromossomos Humanos Par 13/genética , Loci Gênicos , Proteínas Supressoras de Tumor/genética , Animais , Mapeamento Cromossômico , Clonagem Molecular , Embrião de Mamíferos , Técnicas de Transferência de Genes , Genes Letais/fisiologia , Loci Gênicos/genética , Loci Gênicos/fisiologia , Humanos , Leucemia Linfocítica Crônica de Células B/genética , Leucemia Linfocítica Crônica de Células B/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Deleção de Sequência/fisiologia , Proteínas Supressoras de Tumor/fisiologia
10.
PLoS One ; 6(8): e24178, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21904612

RESUMO

Central Asian Strain 1 (CAS1) is the prevalent Mycobacterium tuberculosis genogroup in South Asia. CAS1 strains carry deletions in RD149 and RD152 regions. Significance of these deletions is as yet unknown. We compared CAS1 strains with RD149 and concurrent RD149-RD152 deletions with CAS1 strains without deletions and with the laboratory reference strain, M. tuberculosis H37Rv for growth and for induction of TNFα, IL6, CCL2 and IL10 in THP-1 cells. Growth of CAS1 strains with deletions was slower in broth (RD149; p = 0.024 and RD149-RD152; p = 0.025) than that of strains without deletions. CAS1 strains with RD149 deletion strains further showed reduced intracellular growth (p = 0.013) in THP-1 cells as compared with strains without deletions, and also as compared with H37Rv (p = 0.007) and with CAS1 RD149-RD152 deletion strains (p = 0.029). All CAS1 strains induced higher levels of TNFα and IL10 secretion in THP-1 cells than H37Rv. Additionally, CAS1 strains with RD149 deletions induced more TNFα secretion than those without deletions (p = 0.013). CAS1 RD149 deletion strains from extrapulmonary sources showed more rapid growth and induced lower levels of TNFα and IL6 secretion in THP-1 cells than isolates from pulmonary sources. This data suggests that presence of RD149 reduces growth and increases the induction of TNFα in host cells by CAS1 strains. Differences observed for extrapulmonary strains may indicate an adaptation which increases potential for dissemination and tropism outside the lung. Overall, we hypothesise that RD149 deletions generate genetic diversity within strains and impact interactions of CAS1 strains with host cells with important clinical consequences.


Assuntos
Monócitos/metabolismo , Monócitos/microbiologia , Mycobacterium tuberculosis/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Linhagem Celular , Quimiocina CCL2/metabolismo , Variação Genética , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Mycobacterium tuberculosis/genética , Deleção de Sequência/genética , Deleção de Sequência/fisiologia
11.
J Cell Biol ; 194(5): 779-87, 2011 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-21875948

RESUMO

GCC185 is a long coiled-coil protein localized to the trans-Golgi network (TGN) that functions in maintaining Golgi structure and tethering mannose 6-phosphate receptor (MPR)-containing transport vesicles en route to the Golgi. We report the identification of two distinct domains of GCC185 needed either for Golgi structure maintenance or transport vesicle tethering, demonstrating the independence of these two functions. The domain needed for vesicle tethering binds to the clathrin adaptor AP-1, and cells depleted of GCC185 accumulate MPRs in transport vesicles that are AP-1 decorated. This study supports a previously proposed role of AP-1 in retrograde transport of MPRs from late endosomes to the Golgi and indicates that docking may involve the interaction of vesicle-associated AP-1 protein with the TGN-associated tethering protein GCC185.


Assuntos
Complexo 1 de Proteínas Adaptadoras/metabolismo , Complexo de Golgi/fisiologia , Proteínas de Membrana/metabolismo , Transporte Proteico/fisiologia , Vesículas Transportadoras/fisiologia , Animais , Bovinos , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/metabolismo , Complexo de Golgi/patologia , Proteínas da Matriz do Complexo de Golgi , Células HeLa , Humanos , Proteínas de Membrana/genética , Microscopia de Fluorescência , Proteínas Associadas aos Microtúbulos/metabolismo , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , RNA Interferente Pequeno/genética , Receptor IGF Tipo 2/metabolismo , Deleção de Sequência/fisiologia , Transfecção , Proteínas de Transporte Vesicular/metabolismo , Rede trans-Golgi/fisiologia
12.
Exp Cell Res ; 317(16): 2321-32, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21749867

RESUMO

Hypoxia has been shown to be one of the major events involved in EPO expression. Accordingly, EPO might be expressed by cerebral neoplastic cells, especially in glioblastoma, known to be highly hypoxic tumours. The expression of EPOR has been described in glioma cells. However, data from the literature remain descriptive and controversial. On the basis of an endogenous source of EPO in the brain, we have focused on a potential role of EPOR in brain tumour growth. In the present study, with complementary approaches to target EPO/EPOR signalling, we demonstrate the presence of a functional EPO/EPOR system on glioma cells leading to the activation of the ERK pathway. This EPO/EPOR system is involved in glioma cell proliferation in vitro. In vivo, we show that the down-regulation of EPOR expression on glioma cells reduces tumour growth and enhances animal survival. Our results support the hypothesis that EPOR signalling in tumour cells is involved in the control of glioma growth.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Glioma/tratamento farmacológico , Receptores da Eritropoetina/antagonistas & inibidores , Transdução de Sinais/fisiologia , Animais , Astrócitos/metabolismo , Neoplasias Encefálicas/metabolismo , Núcleo Caudado/patologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Eritropoetina/antagonistas & inibidores , Eritropoetina/genética , Eritropoetina/metabolismo , Eritropoetina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/genética , Glioma/metabolismo , Glioma/patologia , Células Hep G2 , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , RNA Interferente Pequeno/uso terapêutico , Ratos , Ratos Endogâmicos F344 , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Deleção de Sequência/fisiologia , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Exp Cell Res ; 317(16): 2333-41, 2011 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-21726550

RESUMO

The voltage-gated potassium channel, Kv1.3, plays an important role in regulating membrane excitability in diverse cell types ranging from T-lymphocytes to neurons. In the present study, we test the hypothesis that the C-terminal PDZ binding domain modulates the function and localization of Kv1.3. We created a mutant form of Kv1.3 that lacked the last three amino acids of the C-terminal PDZ-binding domain (Kv1.3ΔTDV). This form of Kv1.3 did not bind the PDZ domain containing protein, PSD95. We transfected wild type and mutant Kv1.3 into HEK293 cells and determined if the mutation affected current, Golgi localization, and surface expression of the channel. We found that cells transfected with Kv1.3ΔTDV had greater current and lower Golgi localization than those transfected with Kv1.3. Truncation of the C-terminal PDZ domain did not affect surface expression of Kv1.3. These findings suggest that PDZ-dependent interactions affect both Kv1.3 localization and function. The finding that current and Golgi localization changed without a corresponding change in surface expression suggests that PDZ interactions affect localization and function via independent mechanisms.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Canal de Potássio Kv1.3/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Transporte Proteico/fisiologia , Animais , Animais Recém-Nascidos , Artérias/citologia , Artérias/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Complexo de Golgi/metabolismo , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Músculo Liso Vascular/metabolismo , Neurônios/metabolismo , Oligopeptídeos , Peptídeos/genética , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência/fisiologia , Sistema Nervoso Simpático/citologia , Sistema Nervoso Simpático/metabolismo , Cauda/irrigação sanguínea , Transfecção
14.
J Physiol ; 589(Pt 11): 2719-31, 2011 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-21486785

RESUMO

The cystic fibrosis transmembrane conductance regulator (CFTR) is a chloride channel that belongs to the ATP binding cassette (ABC) superfamily. The deletion of the phenylalanine 508 (ΔF508-CFTR) is the most common mutation among cystic fibrosis (CF) patients. The mutant channels present a severe trafficking defect, and the few channels that reach the plasma membrane are functionally impaired. Interestingly, an ATP analogue, N6-(2-phenylethyl)-2'-deoxy-ATP (P-dATP), can increase the open probability (Po) to ∼0.7, implying that the gating defect of ΔF508 may involve the ligand binding domains, such as interfering with the formation or separation of the dimeric states of the nucleotide-binding domains (NBDs). To test this hypothesis, we employed two approaches developed for gauging the stability of the NBD dimeric states using the patch-clamp technique. We measured the locked-open time induced by pyrophosphate (PPi), which reflects the stability of the full NBD dimer state, and the ligand exchange time for ATP/N6-(2-phenylethyl)-ATP (P-ATP), which measures the stability of the partial NBD dimer state wherein the head of NBD1 and the tail of NBD2 remain associated. We found that both the PPi-induced locked-open time and the ATP/P-ATP ligand exchange time of ΔF508-CFTR channels are dramatically shortened, suggesting that the ΔF508 mutation destabilizes the full and partial NBD dimer states. We also tested if mutations that have been shown to improve trafficking of ΔF508-CFTR, namely the solubilizing mutation F494N/Q637R and ΔRI (deletion of the regulatory insertion), exert any effects on these newly identified functional defects associated with ΔF508-CFTR. Our results indicate that although these mutations increase the membrane expression and function of ΔF508-CFTR, they have limited impact on the stability of both full and partial NBD dimeric states for ΔF508 channels. The structure-function insights gained from this mechanism may provide clues for future drug design.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/química , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Fibrose Cística/genética , Nucleotídeos/metabolismo , Deleção de Sequência/fisiologia , Trifosfato de Adenosina/análogos & derivados , Trifosfato de Adenosina/metabolismo , Substituição de Aminoácidos/fisiologia , Animais , Sítios de Ligação/fisiologia , Células CHO , Cricetinae , Cricetulus , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Difosfatos/metabolismo , Fenômenos Eletrofisiológicos/fisiologia , Humanos , Ativação do Canal Iônico/fisiologia , Técnicas de Patch-Clamp , Fenilalanina/genética , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico/fisiologia , Transfecção
15.
J Clin Endocrinol Metab ; 96(4): E685-90, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21289259

RESUMO

BACKGROUND AND AIMS: Correct gene dosage of SOX3 is critical for the development of the hypothalamo-pituitary axis. Both overdosage of SOX3, as a result of gene duplication, and loss of function resulting from expansion of the first polyalanine (PA) tract are associated with variable degrees of hypopituitarism, with or without mental retardation. The aim of this study was to further investigate the contribution of SOX3 in the etiology of hypopituitarism and the mechanisms involved in the phenotypic variability. METHODS: We screened 154 patients with congenital hypopituitarism and an undescended posterior pituitary for mutations in SOX3 and variability in the length of the first PA tract. In addition, 300 patients with variable septooptic dysplasia were screened for variability of the PA tract. RESULTS: We report a novel 18-base pair deletion (p.A243_A248del6, del6PA) in a female patient with hypopituitarism resulting in a 2-fold increase in transcriptional activation in vitro, compared with wild-type SOX3. We also identified a previously reported seven-alanine expansion (p.A240_A241ins7, +7PA) in two male siblings with isolated GH deficiency and a distinct phenotype, in addition to the nonsynonymous variant p.R5Q in an unrelated individual; this appears to have no functional effect on the protein. In contrast to +7PA, del6PA maintained its ability to repress ß-catenin mediated transcription in vitro. CONCLUSION: This is the first study to report that PA tract deletions associated with hypopituitarism have functional consequences in vitro, possibly due to increased activation of SOX3 target genes. In addition, we have expanded the phenotypic spectrum associated with PA tract expansion (+7PA) mutations to include panhypopituitarism or isolated GH deficiency, with or without mental retardation.


Assuntos
Hipopituitarismo/genética , Peptídeos/genética , Fatores de Transcrição SOXB1/genética , Deleção de Sequência/fisiologia , Criança , Pré-Escolar , Feminino , Testes Genéticos , Humanos , Hipopituitarismo/diagnóstico por imagem , Lactente , Imageamento por Ressonância Magnética , Masculino , Radiografia , Ativação Transcricional , Repetições de Trinucleotídeos/genética , Regulação para Cima
16.
PLoS One ; 6(2): e16627, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21311766

RESUMO

Several mammalian proteins involved in chromatin and DNA modification contain CXXC zinc finger domains. We compared the structure and function of the CXXC domains in the DNA methyltransferase Dnmt1 and the methylcytosine dioxygenase Tet1. Sequence alignment showed that both CXXC domains have a very similar framework but differ in the central tip region. Based on the known structure of a similar MLL1 domain we developed homology models and designed expression constructs for the isolated CXXC domains of Dnmt1 and Tet1 accordingly. We show that the CXXC domain of Tet1 has no DNA binding activity and is dispensable for catalytic activity in vivo. In contrast, the CXXC domain of Dnmt1 selectively binds DNA substrates containing unmethylated CpG sites. Surprisingly, a Dnmt1 mutant construct lacking the CXXC domain formed covalent complexes with cytosine bases both in vitro and in vivo and rescued DNA methylation patterns in dnmt1⁻/⁻ embryonic stem cells (ESCs) just as efficiently as wild type Dnmt1. Interestingly, neither wild type nor ΔCXXC Dnmt1 re-methylated imprinted CpG sites of the H19a promoter in dnmt1⁻/⁻ ESCs, arguing against a role of the CXXC domain in restraining Dnmt1 methyltransferase activity on unmethylated CpG sites.


Assuntos
DNA (Citosina-5-)-Metiltransferases/química , Proteínas de Ligação a DNA/química , Proteínas Proto-Oncogênicas/química , Dedos de Zinco/fisiologia , Sequência de Aminoácidos/fisiologia , Animais , Células Cultivadas , Ilhas de CpG/genética , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , DNA (Citosina-5-)-Metiltransferases/fisiologia , Metilação de DNA/fisiologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Ativação Enzimática/genética , Ativação Enzimática/fisiologia , Humanos , Camundongos , Oxigenases de Função Mista , Modelos Moleculares , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/genética , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas/fisiologia , Deleção de Sequência/fisiologia , Homologia de Sequência de Aminoácidos , Dedos de Zinco/genética
17.
Ann Neurol ; 69(1): 152-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21280085

RESUMO

OBJECTIVE: The fused in sarcoma/translated in liposarcoma (FUS/TLS) protein was recently identified as a cause of familial amyotrophic lateral sclerosis (ALS), as well as a major component of the inclusion bodies found in subtypes of frontotemporal lobar degeneration (FTLD). These diseases now are collectively known as the novel clinical spectrum, FUS proteinopathy. ALS-linked mutations of FUS are clustered in the C-terminal region; however, the molecular properties of mutant FUS remain unclear. To gain insight into the pathogenesis of FUS proteinopathy, we examined the biochemical and cellular characteristics of mutant FUS in expressing cells. METHODS AND RESULTS: Expression of ALS-linked FUS mutations resulted in their assembly into cytoplasmic stress granules (SGs), cellular structures that package mRNA and RNA-binding proteins during cell stress. A deletion mutant series revealed that the C-terminal region in FUS is critical for nuclear retention via Ran guanosine triphosphatase-dependent transport machinery. A parallel study of subcellular distribution revealed that ALS-linked mutants additively disturb the function of the C-terminus for nuclear traffic, resulting in cytoplasmic accumulation and the formation of SGs. INTERPRETATION: This study demonstrates that mutant FUS, which is missing the nuclear traffic activity of the C-terminus, is dislocated to cytoplasm and assembled into SGs, indicating that disruption of translational regulation and metabolism of mRNA via inappropriate/excessive SGs may be crucial for FUS proteinopathies. Our findings provide new biological and pathological insights into the FUS protein that should help our understanding of the pathogenesis of ALS/FTLD.


Assuntos
Transporte Ativo do Núcleo Celular/genética , Transporte Ativo do Núcleo Celular/fisiologia , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/fisiopatologia , Proteína FUS de Ligação a RNA/fisiologia , Deleção de Sequência/fisiologia , Esclerose Lateral Amiotrófica/metabolismo , Animais , Células Cultivadas , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , DNA Complementar/metabolismo , Imunofluorescência , Regulação da Expressão Gênica , Células HeLa/metabolismo , Humanos , Corpos de Inclusão/genética , Corpos de Inclusão/metabolismo , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína FUS de Ligação a RNA/genética , Proteínas de Ligação a RNA/genética , Deleção de Sequência/genética , Frações Subcelulares/metabolismo , Transfecção
18.
FEBS J ; 278(2): 354-70, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21134130

RESUMO

We previously identified a novel interaction between tuberous sclerosis-2 (TSC2) and death-associated protein kinase-1 (DAPK), the consequence being that DAPK catalyses the inactivating phosphorylation of TSC2 to stimulate mammalian target of rapamycin complex 1 (mTORC1) activity. We now report that TSC2 binding to DAPK promotes the degradation of DAPK. We show that DAPK protein levels, but not gene expression, inversely correlate with TSC2 expression. Furthermore, altering mTORC1 activity does not affect DAPK levels, excluding indirect effects of TSC2 on DAPK protein levels through changes in mTORC1 translational control. We provide evidence that the C-terminus regulates TSC2 stability and is required for TSC2 to reduce DAPK protein levels. Importantly, using a GTPase-activating protein-dead missense mutation of TSC2, we demonstrate that the effect of TSC2 on DAPK is independent of GTPase-activating protein activity. TSC2 binds to the death domain of DAPK and we show that this interaction is required for TSC2 to reduce DAPK protein levels and half-life. Finally, we show that DAPK is regulated by the lysosome pathway and that lysosome inhibition blocks TSC2-mediated degradation of DAPK. Our study therefore establishes important functions of TSC2 and the lysosomal-degradation pathway in the control of DAPK stability, which taken together with our previous findings, reveal a regulatory loop between DAPK and TSC2 whose balance can either promote: (a) TSC2 inactivation resulting in mTORC1 stimulation, or (b) DAPK degradation via TSC2 signalling under steady-state conditions. The fine balance between DAPK and TSC2 in this regulatory loop may have subtle but important effects on mTORC1 steady-state function.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Lisossomos/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas Reguladoras de Apoptose/genética , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/genética , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Cloroquina/farmacologia , Proteínas Quinases Associadas com Morte Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Expressão Gênica/genética , Células HEK293 , Humanos , Leupeptinas/farmacologia , Lisossomos/efeitos dos fármacos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Modelos Biológicos , Proteínas Monoméricas de Ligação ao GTP/genética , Complexos Multiproteicos , Mutação de Sentido Incorreto/fisiologia , Neuropeptídeos/genética , Fosforilação/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma , Ligação Proteica/fisiologia , Domínios e Motivos de Interação entre Proteínas/fisiologia , Proteínas/antagonistas & inibidores , RNA Interferente Pequeno/genética , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Proteína Regulatória Associada a mTOR , Proteínas Quinases S6 Ribossômicas/metabolismo , Deleção de Sequência/fisiologia , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Transfecção , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/genética
19.
FEBS J ; 278(2): 282-94, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21126317

RESUMO

MUC5B is one of the major mucin genes expressed in the respiratory tract. Previous studies in our laboratory have demonstrated that MUC5B is expressed in human lung adenocarcinomas and during lung morphogenesis. Moreover, in human lung adenocarcinoma tissues, a converse correlation between MUC5B and thyroid transcription factor-1 (TTF-1) expression, a lung-specific transcription factor, has been established. However, the molecular mechanisms that govern the regulation of MUC5B expression in the lung are largely unknown. In order to better understand the biological role of MUC5B in lung pathophysiology, we report the characterization of the promoter region of the mouse Muc5b mucin gene. The promoter is flanked by a TATA box (TACATAA) identical to that in the human gene. Human and murine promoters share 67.5% similarity over the first 170 nucleotides. By RT-PCR, co-transfection studies and gel-shift assays, we show that Muc5b promoter activity is completely inhibited by TTF-1, whereas factors of the GATA family (GATA-4/GATA-5/GATA-6) are activators. Together, these results demonstrate, for the first time, that Muc5b is a target gene of transcription factors (TTF-1, GATA-6) involved in lung differentiation programs during development and carcinogenesis, and identify TTF-1 as a strong repressor of Muc5b. The characterization of the structural and functional features of the Muc5b mucin gene will provide us with a strong base to develop studies in murine models aimed at the identification of its biological role in lung pathophysiology.


Assuntos
Fator de Transcrição GATA6/metabolismo , Regulação da Expressão Gênica/fisiologia , Mucina-5B/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma de Pulmão , Adenocarcinoma Bronquioloalveolar/metabolismo , Adenocarcinoma Papilar/metabolismo , Animais , Sequência de Bases/genética , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , DNA/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA5/genética , Fator de Transcrição GATA5/metabolismo , Fator de Transcrição GATA6/genética , Expressão Gênica/genética , Humanos , Neoplasias Pulmonares/metabolismo , Camundongos , Dados de Sequência Molecular , Mucina-5B/metabolismo , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/fisiologia , Ligação Proteica/genética , Deleção de Sequência/fisiologia , Homologia de Sequência do Ácido Nucleico , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/genética , Transfecção
20.
Cancer Cell ; 17(6): 560-73, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20541701

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) exert their anticancer effects through cyclooxygenase-2 (COX-2)-dependent and independent mechanisms. Here, we report that Sulindac, an NSAID, induces apoptosis by binding to retinoid X receptor-alpha (RXRalpha). We identified an N-terminally truncated RXRalpha (tRXRalpha) in several cancer cell lines and primary tumors, which interacted with the p85alpha subunit of phosphatidylinositol-3-OH kinase (PI3K). Tumor necrosis factor-alpha (TNFalpha) promoted tRXRalpha interaction with the p85alpha, activating PI3K/AKT signaling. When combined with TNFalpha, Sulindac inhibited TNFalpha-induced tRXRalpha/p85alpha interaction, leading to activation of the death receptor-mediated apoptotic pathway. We designed and synthesized a Sulindac analog K-80003, which has increased affinity to RXRalpha but lacks COX inhibitory activity. K-80003 displayed enhanced efficacy in inhibiting tRXRalpha-dependent AKT activation and tRXRalpha tumor growth in animals.


Assuntos
Anti-Inflamatórios não Esteroides/metabolismo , Anti-Inflamatórios não Esteroides/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor X Retinoide alfa/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Sulindaco/análogos & derivados , Sulindaco/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Citoplasma/metabolismo , Dinoprostona/metabolismo , Desenho de Fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Moleculares , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Prostaglandina-Endoperóxido Sintases/efeitos dos fármacos , Prostaglandina-Endoperóxido Sintases/metabolismo , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Deleção de Sequência/fisiologia , Transdução de Sinais/fisiologia , Sulindaco/metabolismo , Sulindaco/uso terapêutico , Ativação Transcricional/efeitos dos fármacos , Transfecção , Tretinoína/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA