Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Cell ; 137(2): 235-46, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19379691

RESUMO

X-linked myopathy with excessive autophagy (XMEA) is a childhood-onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p it is an essential assembly chaperone of the V-ATPase, the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH, which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids, which upregulates the mTOR pathway and mTOR-dependent macroautophagy, resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge together, and vacuolate the cell. Our results uncover macroautophagic overcompensation leading to cell vacuolation and tissue atrophy as a mechanism of disease.


Assuntos
Genes Ligados ao Cromossomo X , Doenças Musculares/genética , ATPases Vacuolares Próton-Translocadoras/metabolismo , Autofagia , Humanos , Lisossomos/metabolismo , Proteínas de Membrana/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ATPases Vacuolares Próton-Translocadoras/genética
3.
Blood ; 118(24): 6407-17, 2011 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-21963601

RESUMO

Although anemia is common in Shwachman- Diamond syndrome (SDS), the underlying mechanism remains unclear. We asked whether SBDS, which is mutated in most SDS patients, is critical for erythroid development. We found that SBDS expression is high early during erythroid differentiation. Inhibition of SBDS in CD34(+) hematopoietic stem cells and early progenitors (HSC/Ps) and K562 cells led to slow cell expansion during erythroid differentiation. Induction of erythroid differentiation resulted in markedly accelerated apoptosis in the knockdown cells; however, proliferation was only mildly reduced. The percentage of cells entering differentiation was not reduced. Differentiation also increased the oxidative stress in SBDS-knockdown K562 cells, and antioxidants enhanced the expansion capability of differentiating SBDS-knockdown K562 cells and colony production of SDS patient HSC/Ps. Erythroid differentiation also resulted in reduction of all ribosomal subunits and global translation. Furthermore, stimulation of global translation with leucine improved the erythroid cell expansion of SBDS-knockdown cells and colony production of SDS patient HSC/Ps. Leucine did not reduce the oxidative stress in SBDS-deficient K562 cells. These results demonstrate that SBDS is critical for normal erythropoiesis. Erythropoietic failure caused by SBDS deficiency is at least in part related to elevated ROS levels and translation insufficiency because antioxidants and leucine improved cell expansion.


Assuntos
Eritropoese , Proteínas/metabolismo , Antioxidantes/farmacologia , Apoptose , Doenças da Medula Óssea/tratamento farmacológico , Doenças da Medula Óssea/metabolismo , Proliferação de Células , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Insuficiência Pancreática Exócrina/tratamento farmacológico , Insuficiência Pancreática Exócrina/metabolismo , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células K562 , Leucina/metabolismo , Lipomatose/tratamento farmacológico , Lipomatose/metabolismo , Estresse Oxidativo , Biossíntese de Proteínas , Proteínas/antagonistas & inibidores , Proteínas/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/metabolismo , Ribossomos/metabolismo , Síndrome de Shwachman-Diamond
4.
Acta Neuropathol ; 125(3): 439-57, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23315026

RESUMO

X-linked Myopathy with Excessive Autophagy (XMEA) is a childhood onset disease characterized by progressive vacuolation and atrophy of skeletal muscle. We show that XMEA is caused by hypomorphic alleles of the VMA21 gene, that VMA21 is the diverged human ortholog of the yeast Vma21p protein, and that like Vma21p, VMA21 is an essential assembly chaperone of the vacuolar ATPase (V-ATPase), the principal mammalian proton pump complex. Decreased VMA21 raises lysosomal pH which reduces lysosomal degradative ability and blocks autophagy. This reduces cellular free amino acids which leads to downregulation of the mTORC1 pathway, and consequent increased macroautophagy resulting in proliferation of large and ineffective autolysosomes that engulf sections of cytoplasm, merge, and vacuolate the cell. Our results uncover a novel mechanism of disease, namely macroautophagic overcompensation leading to cell vacuolation and tissue atrophy.


Assuntos
Adenosina Trifosfatases/metabolismo , Autofagia/genética , Doenças por Armazenamento dos Lisossomos/genética , Doenças por Armazenamento dos Lisossomos/prevenção & controle , Doenças Musculares/genética , Doenças Musculares/prevenção & controle , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Células Cultivadas , Humanos , Concentração de Íons de Hidrogênio , Leucina/metabolismo , Doenças por Armazenamento dos Lisossomos/patologia , Lisossomos/genética , Lisossomos/metabolismo , Masculino , Camundongos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/ultraestrutura , Doenças Musculares/patologia , Mutação/genética , Interferência de RNA/fisiologia , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Frações Subcelulares/metabolismo , Frações Subcelulares/patologia , Fatores de Tempo , Vacúolos/metabolismo
5.
J Virol ; 85(23): 12529-36, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21917946

RESUMO

Gammaretrovirus receptors have been suggested to contain the necessary determinants to mediate virus binding and entry. Here, we show that murine NIH 3T3 and baby hamster kidney (BHK) cells overexpressing receptors for subgroup A, B, and C feline leukemia viruses (FeLVs) are weakly susceptible (10(1) to 10(2) CFU/ml) to FeLV pseudotype viruses containing murine leukemia virus (MLV) core (Gag-Pol) proteins, whereas FeLV receptor-expressing murine Mus dunni tail fibroblast (MDTF) cells are highly susceptible (10(4) to 10(6) CFU/ml). However, NIH 3T3 cells expressing the FeLV subgroup B receptor PiT1 are highly susceptible to gibbon ape leukemia virus pseudotype virus, which differs from the FeLV pseudotype viruses only in the envelope protein. FeLV resistance is not caused by a defect in envelope binding, low receptor expression levels, or N-linked glycosylation. Resistance is not alleviated by substitution of the MLV core in the FeLV pseudotype virus with FeLV core proteins. Interestingly, FeLV resistance is alleviated by fusion of receptor-expressing NIH 3T3 and BHK cells with MDTF or human TE671 cells, suggesting the absence of an additional cellular component in NIH 3T3 and BHK cells that is required for FeLV infection. The putative FeLV-specific cellular component is not a secreted factor, as MDTF conditioned medium does not alleviate the block to FeLV infection. Together, our findings suggest that FeLV infection requires an additional envelope-dependent cellular component that is absent in NIH 3T3 and BHK cells but that is present in MDTF and TE671 cells.


Assuntos
Vírus da Leucemia Felina/patogenicidade , Leucemia Felina/metabolismo , Leucemia Felina/virologia , Receptores Virais/metabolismo , Proteínas do Core Viral/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Gatos , Células Cultivadas , Cricetinae , Citometria de Fluxo , Glicosilação/efeitos dos fármacos , Humanos , Rim/citologia , Rim/metabolismo , Rim/virologia , Leucemia Felina/patologia , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Células NIH 3T3 , Ligação Proteica , Fator de Transcrição Pit-1/metabolismo , Vírion/fisiologia , Ligação Viral
6.
Transfusion ; 52(2): 332-42, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22239212

RESUMO

BACKGROUND: Xenotropic murine leukemia virus (MLV)-related virus (XMRV) and other related MLVs have been described with chronic fatigue syndrome and certain types of prostate cancer. In addition, prevalence rates as high as 7% have been reported in blood donors, raising the risk of transfusion-related transmission. Several laboratories have utilized microneutralization assays as a surrogate marker for detection of anti-MLV serologic responses--with up to 25% of prostate cancer patients reported to harbor neutralizing antibody responses. STUDY DESIGN AND METHODS: We developed a high-throughput microneutralization assay for research studies on blood donors using retroviral vectors pseudotyped with XMRV-specific envelopes. Infection with these pseudotypes was neutralized by sera from both macaques and mice challenged with XMRV, but not preimmune serum. A total of 354 plasma samples from blood donors in the Reno/Tahoe area were screened for neutralization. RESULTS: A total of 6.5% of donor samples gave moderate neutralization of XMRV, but not control pseudotypes. However, further testing by Western blot revealed no evidence of antibodies against MLVs in any of these samples. Furthermore, no evidence of infectious virus or viral nucleic acid was observed. CONCLUSION: A microneutralization assay was developed for detection of XMRV and can be applied in a high-throughput format for large-scale studies. Although a proportion of blood donors demonstrated the ability to block XMRV envelope-mediated infection, we found no evidence that this inhibition was mediated by specific antibodies elicited by exposure to XMRV or MLV. It is likely that this moderate neutralization is mediated through another, nonspecific mechanism.


Assuntos
Doadores de Sangue , Ensaios de Triagem em Larga Escala/métodos , Vírus da Leucemia Murina/isolamento & purificação , Testes de Neutralização/métodos , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/isolamento & purificação , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais/sangue , Anticorpos Antivirais/isolamento & purificação , Doadores de Sangue/estatística & dados numéricos , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Vírus da Leucemia Murina/imunologia , Macaca mulatta , Masculino , Camundongos , Microquímica/métodos , Células NIH 3T3 , Infecções por Retroviridae/sangue , Infecções por Retroviridae/diagnóstico , Infecções por Retroviridae/epidemiologia , Infecções por Retroviridae/virologia , Vírus Relacionado ao Vírus Xenotrópico da Leucemia Murina/imunologia
7.
J Virol ; 83(13): 6706-16, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19369334

RESUMO

The pathogenic subgroup C feline leukemia virus (FeLV-C) arises in infected cats as a result of mutations in the envelope (Env) of the subgroup A FeLV (FeLV-A). To better understand emergence of FeLV-C and potential FeLV intermediates that may arise, we characterized FeLV Env sequences from the primary FY981 FeLV isolate previously derived from an anemic cat. Here, we report the characterization of the novel FY981 FeLV Env that is highly related to FeLV-A Env but whose variable region A (VRA) receptor recognition sequence partially resembles the VRA sequence from the prototypical FeLV-C/Sarma Env. Pseudotype viruses bearing FY981 Env were capable of infecting feline, human, and guinea pig cells, suggestive of a subgroup C phenotype, but also infected porcine ST-IOWA cells that are normally resistant to FeLV-C and to FeLV-A. Analysis of the host receptor used by FY981 suggests that FY981 can use both the FeLV-C receptor FLVCR1 and the feline FeLV-A receptor THTR1 for infection. However, our results suggest that FY981 infection of ST-IOWA cells is not mediated by the porcine homologue of FLVCR1 and THTR1 but by an alternative receptor, which we have now identified as the FLVCR1-related protein FLVCR2. Together, our results suggest that FY981 FeLV uses FLVCR1, FLVCR2, and THTR1 as receptors. Our findings suggest the possibility that pathogenic FeLV-C arises in FeLV-infected cats through intermediates that are multitropic in their receptor use.


Assuntos
Vírus da Leucemia Felina/genética , Proteínas de Membrana Transportadoras/metabolismo , Receptores Virais/metabolismo , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Animais , Gatos , Linhagem Celular , Cricetinae , Humanos , Vírus da Leucemia Felina/isolamento & purificação , Vírus da Leucemia Felina/patogenicidade , Camundongos , Dados de Sequência Molecular , Alinhamento de Sequência , Suínos
8.
Haematologica ; 93(11): 1617-26, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18815190

RESUMO

BACKGROUND: Diamond-Blackfan anemia is a fatal congenital anemia characterized by a specific disruption in erythroid progenitor cell development. Approximately 25% of patients have mutations in the ribosomal protein RPS19 suggesting that Diamond-Blackfan anemia may be caused by a defect in ribosome biogenesis and translation. However, it is unclear how these defects specifically disrupt early erythropoiesis. Recent studies have shown that the retroviral receptor/heme exporter FLVCR1 is critical for early erythropoiesis. FLVCR1 null mice, despite dying in utero and having reduced myeloid and lymphoid cell growth, show a disruption in early erythropoiesis and have craniofacial and limb deformities similar to those found in some Diamond-Blackfan anemia patients. DESIGN AND METHODS: In this study, we recapitulated the Diamond-Blackfan anemia hematologic features of reduced erythropoiesis but normal myelopoiesis by disrupting FLVCR1 in human hematopoietic stem cells. RESULTS: We found that CD71(high) cells, which are enriched for immature erythroid cells, from Diamond-Blackfan anemia patients negative for RPS19 gene mutations express alternatively spliced isoforms of FLVCR1 transcript which encode proteins whose expression and function are disrupted. More importantly, our results suggest alternative splicing of FLVCR1 is significantly enhanced in Diamond-Blackfan anemia immature erythroid cells. Furthermore, we also observed enhanced FLVCR1 alternative splicing and a dramatic reduction of FLVCR1 protein expression in RPS19 down-regulated human K562 cells, which were used as a model to represent RPS19 gene mutated Diamond-Blackfan anemia. CONCLUSIONS: Taken together, our results suggest enhanced alternative splicing of FLVCR1 transcripts and subsequent FLVCR1 insufficiency as an additional contributing factor to the erythropoietic defect observed in Diamond-Blackfan anemia.


Assuntos
Processamento Alternativo , Anemia de Diamond-Blackfan/genética , Eritropoese/fisiologia , Proteínas de Membrana Transportadoras/genética , Mutação , Receptores Virais/genética , Idade de Início , Medula Óssea/patologia , Primers do DNA , Eritropoese/genética , Feminino , Regulação da Expressão Gênica , Genes env , Vetores Genéticos , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Lactente , Recém-Nascido , Células K562 , Masculino , Núcleo Familiar , Reação em Cadeia da Polimerase , Valores de Referência , Proteínas Ribossômicas/genética
9.
Artigo em Inglês | MEDLINE | ID: mdl-28261277

RESUMO

The use of gene therapy (GT) for the treatment of primary immune deficiencies (PID) including severe combined immune deficiency (SCID) has progressed significantly in the recent years. In particular, long-term studies have shown that adenosine deaminase (ADA) gene delivery into ADA-deficient hematopoietic stem cells that are then transplanted into the patients corrects the abnormal function of the ADA enzyme, which leads to immune reconstitution. In contrast, the outcome was disappointing for patients with X-linked SCID, Wiskott-Aldrich syndrome and chronic granulomatous disease who received GT followed by autologous gene corrected transplantations, as many developed hematological malignancies. The malignancies were attributed to the predilection of the viruses used for gene delivery to integrated at oncogenic areas. The availability of safer and more efficient self-inactivating lentiviruses for gene delivery has reignited the interest in GT for many PID that are now in various stages of pre-clinical studies and clinical trials. Moreover, advances in early diagnosis of PID and gene editing technology coupled with enhanced abilities to generate and manipulate stem cells ex vivo are expected to further contribute to the benefit of GT for PID. Here we review the past, the present and the future of GT for PID, with particular emphasis on the Canadian perspective.

10.
Mol Cell Biol ; 30(22): 5318-24, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20823265

RESUMO

Mutations in FLVCR2, a cell surface protein related by homology and membrane topology to the heme exporter/retroviral receptor FLVCR1, have recently been associated with Fowler syndrome, a vascular disorder of the brain. We previously identified FLVCR2 to function as a receptor for FY981 feline leukemia virus (FeLV). However, the cellular function of FLVCR2 remains unresolved. Here, we report the cellular function of FLVCR2 as an importer of heme, based on the following observations. First, FLVCR2 binds to hemin-conjugated agarose, and binding is competed by free hemin. Second, mammalian cells and Xenopus laevis oocytes expressing FLVCR2 display enhanced heme uptake. Third, heme import is reduced after the expression of FLVCR2-specific small interfering RNA (siRNA) or after the binding of the FY981 FeLV envelope protein to the FLVCR2 receptor. Finally, cells overexpressing FLVCR2 are more sensitive to heme toxicity, a finding most likely attributable to enhanced heme uptake. Tissue expression analysis indicates that FLVCR2 is expressed in a broad range of human tissues, including liver, placenta, brain, and kidney. The identification of a cellular function for FLVCR2 will have important implications in elucidating the pathogenic mechanisms of Fowler syndrome and of phenotypically associated disorders.


Assuntos
Heme/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Receptores Virais/metabolismo , Animais , Transporte Biológico/fisiologia , Encefalopatias/metabolismo , Encefalopatias/patologia , Gatos , Linhagem Celular , Cricetinae , Cricetulus , Feminino , Heme/toxicidade , Humanos , Proteínas de Membrana Transportadoras/genética , Oócitos/citologia , Oócitos/fisiologia , Gravidez , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Virais/genética , Síndrome , Distribuição Tecidual , Xenopus laevis
11.
Virology ; 370(2): 273-84, 2008 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-17945326

RESUMO

The receptor-binding domain (RBD) in the surface (SU) subunit of gammaretrovirus envelope glycoprotein is critical for determining the host receptor specificity of the virus. This domain is separated from the carboxy terminal C domain (Cdom) of SU by a proline-rich region. In this study, we show that the Cdom region in the SU from subgroup C feline leukemia virus (FeLV-C) forms a second receptor-binding domain that is distinct from its RBD, and which can independently bind to its host receptor FLVCR1, in the absence of RBD. Furthermore, our results suggest that residues located in the C2 disulfide-bonded loop in FeLV-C Cdom are critical for SU binding to FLVCR1 and for virus infection. We propose that binding of FeLV-C SU to FLVCR1 involves interaction of two receptor-binding domains (RBD and Cdom) with FLVCR1, and that this mechanism of interaction is conserved for other gammaretroviruses. Our results could have important implications for designing gammaretrovirus vectors that can efficiently infect specific target cells.


Assuntos
Produtos do Gene env/química , Produtos do Gene env/fisiologia , Vírus da Leucemia Felina/fisiologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação/genética , Gatos , Linhagem Celular , Primers do DNA/genética , DNA Viral/genética , Produtos do Gene env/genética , Humanos , Vírus da Leucemia Felina/classificação , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/patogenicidade , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Receptores Virais/genética , Receptores Virais/fisiologia , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Proteínas Oncogênicas de Retroviridae/fisiologia , Homologia de Sequência de Aminoácidos , Virulência
12.
J Virol ; 80(4): 1742-51, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16439531

RESUMO

Infection of cells by the highly anemogenic feline leukemia virus subgroup C (FeLV-C) is mediated by the heme exporter FLVCR1, a cell surface protein containing 12 potential transmembrane segments with six presumptive extracellular loops (ECLs). To identify FLVCR1 residues critical for mediating FeLV-C infection, we first independently isolated a human cDNA encoding the FLVCR2 protein that shares 52% identity to human FLVCR1, and we show that FLVCR2 does not function as a receptor for FeLV-C. Then, by generating specific hybrids between FLVCR1 and FLVCR2 and testing susceptibility of mouse cells expressing these hybrids to beta-galactosidase encoding FeLV-C, we identify FLVCR1 ECLs 1 and 6 as critical for mediating FeLV-C infection. Mouse cells expressing a hybrid protein containing FLVCR2 backbone with the ECL6 sequence from FLVCR1 were highly susceptible to FeLV-C infection. Using site-directed mutagenesis, we show that a single mutation of Asn463 in FLVCR2 ECL6 to an acidic Asp residue (a residue present in the corresponding position 487 in FLVCR1 ECL6) is sufficient to render FLVCR2 functional as an FeLV-C receptor. However, an Asp487Asn mutation in FLVCR1 ECL6 or substitution of the entire FLVCR1 ECL6 sequence for FLVCR2 ECL6 sequence does not disrupt receptor function. Subsequent substitutions show that residues within FLVCR1 ECL1 also contribute to mediating FeLV-C infection. Furthermore, our results suggest that FLVCR1 regions that mediate FeLV-C surface unit binding are distinct from ECL1 and ECL6. Our results are consistent with previous conclusions that infection of cells by gammaretroviruses involves interaction of virus with multiple receptor regions.


Assuntos
Vírus da Leucemia Felina/fisiologia , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/fisiologia , Receptores Virais/química , Receptores Virais/fisiologia , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linhagem Celular , DNA/química , DNA/genética , DNA Complementar/isolamento & purificação , Genes Reporter , Humanos , Proteínas de Membrana Transportadoras/genética , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Estrutura Terciária de Proteína , Receptores Virais/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Recombinação Genética , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , beta-Galactosidase/análise , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA