Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
N Engl J Med ; 371(6): 507-518, 2014 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-25029335

RESUMO

BACKGROUND: The study of autoinflammatory diseases has uncovered mechanisms underlying cytokine dysregulation and inflammation. METHODS: We analyzed the DNA of an index patient with early-onset systemic inflammation, cutaneous vasculopathy, and pulmonary inflammation. We sequenced a candidate gene, TMEM173, encoding the stimulator of interferon genes (STING), in this patient and in five unrelated children with similar clinical phenotypes. Four children were evaluated clinically and immunologically. With the STING ligand cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), we stimulated peripheral-blood mononuclear cells and fibroblasts from patients and controls, as well as commercially obtained endothelial cells, and then assayed transcription of IFNB1, the gene encoding interferon-ß, in the stimulated cells. We analyzed IFNB1 reporter levels in HEK293T cells cotransfected with mutant or nonmutant STING constructs. Mutant STING leads to increased phosphorylation of signal transducer and activator of transcription 1 (STAT1), so we tested the effect of Janus kinase (JAK) inhibitors on STAT1 phosphorylation in lymphocytes from the affected children and controls. RESULTS: We identified three mutations in exon 5 of TMEM173 in the six patients. Elevated transcription of IFNB1 and other gene targets of STING in peripheral-blood mononuclear cells from the patients indicated constitutive activation of the pathway that cannot be further up-regulated with stimulation. On stimulation with cGAMP, fibroblasts from the patients showed increased transcription of IFNB1 but not of the genes encoding interleukin-1 (IL1), interleukin-6 (IL6), or tumor necrosis factor (TNF). HEK293T cells transfected with mutant constructs show elevated IFNB1 reporter levels. STING is expressed in endothelial cells, and exposure of these cells to cGAMP resulted in endothelial activation and apoptosis. Constitutive up-regulation of phosphorylated STAT1 in patients' lymphocytes was reduced by JAK inhibitors. CONCLUSIONS: STING-associated vasculopathy with onset in infancy (SAVI) is an autoinflammatory disease caused by gain-of-function mutations in TMEM173. (Funded by the Intramural Research Program of the National Institute of Arthritis and Musculoskeletal and Skin Diseases; ClinicalTrials.gov number, NCT00059748.).


Assuntos
Inflamação/genética , Proteínas de Membrana/genética , Mutação , Dermatopatias Vasculares/genética , Idade de Início , Citocinas/genética , Citocinas/metabolismo , Feminino , Fibroblastos/metabolismo , Genes Dominantes , Humanos , Lactente , Recém-Nascido , Inflamação/metabolismo , Interferon gama/genética , Interferon gama/metabolismo , Janus Quinases/antagonistas & inibidores , Pneumopatias/genética , Masculino , Linhagem , Fosforilação , Fator de Transcrição STAT1/metabolismo , Análise de Sequência de DNA , Dermatopatias Vasculares/metabolismo , Síndrome , Transcrição Gênica , Regulação para Cima
2.
Gene Ther ; 23(11): 815-818, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27487944

RESUMO

Integrating vectors based on γ-retroviruses and containing full-length long terminal repeats (LTRs) have been associated with activation of oncogene expression and leukemogenesis in human gene therapy trials. Identification of the specific molecular elements of the LTRs that have a role in insertional oncogenesis events is important as it can lead to the development of safer gene transfer vectors. The negative control region (NCR) of the LTR is a particularly well-conserved sequence among mammalian γ-retroviruses with demonstrated regulatory activity of gene transcription in hematopoietic cells, which led us to hypothesize that this region may have a role in insertional oncogenesis after γ-retroviral vector (GV)-mediated gene transfer into hematopoietic progenitors. We used an in vitro assay of murine bone marrow cell immortalization to compare the immortalization capabilities of a series of GVs carrying murine leukemia virus (MLV) LTR deletion mutants. Compared with GV carrying the full-length MLV LTR, deletion of the complete LTR enhancer sequence showed significant reduction of immortalization rates. However, the use of a mutant LTR deleted of the enhancer sequence, with exception of the NCR, did not affect immortalization. Importantly, the inclusion of an LTR mutant devoid only of the NCR did show significant reduction of immortalization rates compared with the full LTR sequence. Therefore, our data point to the NCR as a key element for immortalization and justify additional studies to evaluate its specific role in MLV-mediated insertional oncogenesis.


Assuntos
Transformação Celular Viral , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Vírus da Leucemia Murina de Moloney/genética , Sequências Repetidas Terminais , Animais , Células Cultivadas , Técnicas de Transferência de Genes , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional
3.
Clin Immunol ; 142(2): 160-6, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22079330

RESUMO

Wiskott-Aldrich syndrome (WAS) is a rare X-linked disorder caused by mutations in the WAS gene. Glomerulonephritis is a frequent complication, however, histopathological data from affected patients is scarce because the thrombocytopenia that affects most patients is a contraindication to renal biopsies. We found that WASp-deficient mice develop proliferative glomerulonephritis reminiscent of human IgA nephropathy (IgAN). We examined whether increased aberrant IgA production is associated with the development of glomerulonephritis in WASp-deficient mice. Serum IgA and IgA production by splenic B cells was increased in WASp-deficient mice compared to wild-type (WT) mice. A lectin-binding study revealed a reduced ratio of sialylated and galactosylated IgA in the sera from old WASp-deficient mice. Circulating IgA-containing immune complexes showed significantly higher titers in WASp-deficient mice compared to WT mice. These results indicate that the increased IgA production and aberrant glycosylation of IgA may be critically involved in the pathogenesis of glomerulonephritis in WAS.


Assuntos
Glomerulonefrite por IGA/imunologia , Imunoglobulina A/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/deficiência , Síndrome de Wiskott-Aldrich/imunologia , Animais , Linfócitos B/imunologia , Modelos Animais de Doenças , Glomerulonefrite por IGA/metabolismo , Glomerulonefrite por IGA/patologia , Glicosilação , Humanos , Imunoglobulina A/sangue , Camundongos , Camundongos Knockout , Baço/imunologia , Trombocitopenia/metabolismo
4.
Scand J Immunol ; 74(5): 471-81, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21671975

RESUMO

Patients with adenosine deaminase (ADA) deficiency exhibit spontaneous and partial clinical remission associated with somatic reversion of inherited mutations. We report a child with severe combined immunodeficiency (T-B- SCID) due to ADA deficiency diagnosed at the age of 1 month, whose lymphocyte counts including CD4+ and CD8+ T and NK cells began to improve after several months with normalization of ADA activity in Peripheral blood lymphocytes (PBL), as a result of somatic mosaicism caused by monoallelic reversion of the causative mutation in the ADA gene. He was not eligible for haematopoietic stem cell transplantation (HSCT) or gene therapy (GT); therefore he was placed on enzyme replacement therapy (ERT) with bovine PEG-ADA. The follow-up of metabolic and immunologic responses to ERT included gradual improvement in ADA activity in erythrocytes and transient expansion of most lymphocyte subsets, followed by gradual stabilization of CD4+ and CD8+ T (with naïve phenotype) and NK cells, and sustained expansion of TCRγδ+ T cells. This was accompanied by the disappearance of the revertant T cells as shown by DNA sequencing from PBL. Although the patient's clinical condition improved marginally, he later developed a germinal cell tumour and eventually died at the age of 67 months from sepsis. This case adds to our current knowledge of spontaneous reversion of mutations in ADA deficiency and shows that the effects of the ERT may vary among these patients, suggesting that it could depend on the cell and type in which the somatic mosaicism is established upon reversion.


Assuntos
Adenosina Desaminase/metabolismo , Terapia de Reposição de Enzimas , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Neoplasias Primárias Desconhecidas/genética , Neoplasias Primárias Desconhecidas/terapia , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Adenosina Desaminase/administração & dosagem , Adenosina Desaminase/genética , Adenosina Desaminase/imunologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Bovinos , Contagem de Células , Criança , Pré-Escolar , Análise Mutacional de DNA , Evolução Fatal , Humanos , Imunofenotipagem , Lactente , Células Matadoras Naturais/patologia , Neoplasias Pulmonares/complicações , Neoplasias Pulmonares/fisiopatologia , Neoplasias Pulmonares/secundário , Masculino , Mosaicismo/efeitos dos fármacos , Mutação/genética , Neoplasias Primárias Desconhecidas/complicações , Neoplasias Primárias Desconhecidas/patologia , Neoplasias Primárias Desconhecidas/fisiopatologia , Receptores de Antígenos de Linfócitos T/metabolismo , Imunodeficiência Combinada Severa/complicações , Imunodeficiência Combinada Severa/patologia , Imunodeficiência Combinada Severa/fisiopatologia , Choque Séptico
5.
J Exp Med ; 183(6): 2687-92, 1996 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-8676091

RESUMO

Mutations affecting the expression of the Janus family kinase JAK3 were recently shown to be responsible for autosomal recessive severe combined immunodeficiency (SCID). JAK3-deficient patients present with a clinical phenotype virtually indistinguishable from boys affected by X-linked SCID, a disease caused by genetic defects of the common gamma chain (gamma c) that is a shared component of the receptors for IL-2, IL-4, IL-7, IL-9, and IL-15. The specific interaction of JAK3 and gamma c represents the biochemical basis for the similarities between these two immunodeficiencies. Both forms of SCID are characterized by recurrent, severe infections leading to death in infancy unless successfully treated by allogeneic bone marrow transplantation. Because of the potentially lethal complications associated with allogeneic bone marrow transplantation and the frequent lack of suitable marrow donors, the development of alternative forms of therapy is highly desirable. To this end, we investigated a retroviral-mediated gene correction approach for JAK3-deficiency. A vector carrying a copy of JAK3 cDNA was constructed and used to transduce B cell lines derived from patients with JAK3-deficient SCID. We demonstrate restoration of JAK3 expression and phosphorylation upon IL-2 and IL-4 stimulation. Furthermore, patients' cells transduced with JAK3 acquired the ability to proliferate normally in response to IL-2. These data indicate that the biological defects of JAK3-deficient cells can be efficiently corrected in vitro by retroviral-mediated gene transfer, thus providing the basis for future investigation of gene therapy as treatment for JAK3-deficient SCID.


Assuntos
Terapia Genética , Proteínas Tirosina Quinases/deficiência , Proteínas Tirosina Quinases/genética , Receptores de Interleucina/genética , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia , Linfócitos B/imunologia , Western Blotting , Células Cultivadas , Criança , Clonagem Molecular , Consanguinidade , Genes Recessivos , Vetores Genéticos , Humanos , Interleucina-2/farmacologia , Janus Quinase 3 , Ativação Linfocitária/efeitos dos fármacos , Substâncias Macromoleculares , Masculino , Proteínas Tirosina Quinases/biossíntese , Proteínas Recombinantes/biossíntese , Retroviridae , Imunodeficiência Combinada Severa/enzimologia , Transfecção , Cromossomo X
6.
Mol Cell Biol ; 20(12): 4371-80, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10825200

RESUMO

Interleukin-2 (IL-2) activates several different families of tyrosine kinases, but precisely how these kinases interact is not completely understood. We therefore investigated the functional relationships among Jak3, Lck, and Syk in IL-2 signaling. We first observed that in the absence of Jak3, both Lck and Syk had the capacity to phosphorylate Stat3 and Stat5a. However, neither supported IL-2-induced STAT activation, nor did dominant negative alleles of these kinases inhibit. Moreover, pharmacological abrogation of Lck activity did not inhibit IL-2-mediated phosphorylation of Jak3 and Stat5a. Importantly, ligand-dependent Syk activation was dependent on the presence of catalytically active Jak3, whereas Lck activation was not. Interestingly, Syk functioned as a direct substrate of Jak1 but not Jak3. Additionally, Jak3 phosphorylated Jak1, whereas the reverse was not the case. Taken together, our data support a model in which Lck functions in parallel with Jak3, while Syk functions as a downstream element of Jaks in IL-2 signaling. Jak3 may regulate Syk catalytic activity indirectly via Jak1. However, IL-2-mediated Jak3/Stat activation is not dependent on Lck or Syk. While the essential roles of Jak1 and Jak3 in signaling by gammac-utilizing cytokines are clear, it will be important to dissect the exact contributions of Lck and Syk in mediating the effects of IL-2 and related cytokines.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Interleucina-2/fisiologia , Proteínas do Leite , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais , Transativadores/fisiologia , Animais , Linhagem Celular , Ativação Enzimática , Precursores Enzimáticos/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Janus Quinase 3 , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/fisiologia , Fosforilação , Receptores de Interleucina-2/fisiologia , Fator de Transcrição STAT5 , Quinase Syk , Proteínas Supressoras de Tumor
7.
Mol Cell Biol ; 20(3): 947-56, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10629052

RESUMO

The structure of Janus kinases (JAKs) is unique among protein tyrosine kinases in having tandem, nonidentical kinase and pseudokinase domains. Despite its conservation in evolution, however, the function of the pseudokinase domain remains poorly understood. Lack of JAK3 expression results in severe combined immunodeficiency (SCID). In this study, we analyze two SCID patients with mutations in the JAK3 pseudokinase domain, which allows for protein expression but disrupts the regulation of the kinase activity. Specifically, these mutant forms of JAK3 had undetectable kinase activity in vitro but were hyperphosphorylated both in patients' Epstein-Barr virus-transformed B cells and when overexpressed in COS7 cells. Moreover, reconstitution of cells with these mutants demonstrated that, although they were constitutively phosphorylated basally, they were unable to transmit cytokine-dependent signals. Further analysis showed that the isolated catalytic domain of JAK3 was functional whereas either the addition of the pseudokinase domain or its deletion from the full-length molecule reduced catalytic activity. Through coimmunoprecipitation of the isolated pseudokinase domain with the isolated catalytic domain, we provide the first evidence that these two domains interact. Furthermore, whereas the wild-type pseudokinase domain modestly inhibited kinase domain-mediated STAT5 phosphorylation, the patient-derived mutants markedly inhibited this phosphorylation. We thus conclude that the JAK3 pseudokinase domain is essential for JAK3 function by regulating its catalytic activity and autophosphorylation. We propose a model in which this occurs via intramolecular interaction with the kinase domain and that increased inhibition of kinase activity by the pseudokinase domain likely contributes to the disease pathogenesis in these two patients.


Assuntos
Mutação , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Imunodeficiência Combinada Severa/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Células COS , Domínio Catalítico , Linhagem Celular Transformada , Clonagem Molecular , Humanos , Interleucina-2/farmacologia , Interleucina-2/fisiologia , Janus Quinase 3 , Mutação Puntual , Proteínas Tirosina Quinases/química , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Imunodeficiência Combinada Severa/enzimologia , Transdução de Sinais , Transfecção
8.
Cancer Res ; 59(20): 5233-8, 1999 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-10537302

RESUMO

The goal of this study was to improve the therapeutic index of the herpes simplex virus-thymidine kinase/ganciclovir (HSV-tk/GCV) system by the addition of thymidylate synthase (TS) inhibitors. For this, we assessed the potential of GCV to synergistically interact with 5-fluorouracil (5-FU), ZD1694 (Tomudex), and (E)-5-(2-bromovinyl)-2'-deoxyuridine in HSV-tk-expressing murine MC38 STK and human HT-29 STK colon carcinoma cell lines. Synergistic cell killing was observed in a clonogenic assay over most of the cytotoxic dose range by the median-effect principle of Chou and Talalay (T. C. Chou and P. Talalay, Adv. Enzyme Regul., 22: 27-55, 1984). In a s.c. HT-29 STK xenograft tumor model, we demonstrated that the combination of GCV and 5-FU resulted in statistically significant enhanced animal survival over single-agent treatment. Furthermore, we showed that the combination of GCV and ZD1694 in association with the HSV-tk/GCV system was at least as effective as GCV/5-FU in vitro and in vivo. The mechanism for the observed synergy is most likely attributable to the increased GCV phosphorylation in the presence of the tested TS inhibitors. Our data suggest that the HSV-tk/GCV metabolic suicide gene transfer system could serve as an adjuvant of the presently used TS inhibitors, thus potentially improving the efficacy of present cancer gene therapy approaches.


Assuntos
Antivirais/uso terapêutico , Neoplasias do Colo/terapia , Inibidores Enzimáticos/uso terapêutico , Ganciclovir/uso terapêutico , Terapia Genética , Simplexvirus/enzimologia , Timidina Quinase/genética , Timidilato Sintase/antagonistas & inibidores , Bromodesoxiuridina/análogos & derivados , Bromodesoxiuridina/uso terapêutico , Terapia Combinada , Fluoruracila/uso terapêutico , Humanos , Quinazolinas/uso terapêutico , Tiofenos/uso terapêutico , Células Tumorais Cultivadas , Ensaio Tumoral de Célula-Tronco
9.
Hum Gene Ther ; 11(14): 2051-6, 2000 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-11020803

RESUMO

Corrective gene transfer into hematopoietic stem cells (HSCs) is being investigated as therapy for X-linked severe combined immunodeficiency (XSCID) and it is hoped that selective advantage of gene-corrected HSCs will help in achieving full immune reconstitution after treatment. Lines of evidence from the results of allogeneic bone marrow transplantation in patients with XSCID support this hypothesis that, however, has not been rigorously tested in an experimental system. We studied the competition kinetics between normal and XSCID bone marrow (BM) cells using a murine bone marrow transplantation (BMT) model. For easy chimerism determination, we used genetic marking with retrovirus-mediated expression of the enhanced green fluorescent protein (EGFP). We found that XSCID BM cells were able to compete with normal BM cells for engraftment of myeloid lineages in a dose-dependent manner, whereas we observed selective repopulation of T, B, and NK cells deriving from normal BM cells. This was true despite the evidence of competitive engraftment of XSCID lineage marker-negative/c-Kit-positive (Lin-/c-Kit+) cells in the bone marrow of treated animals. From these results we extrapolate that genetic correction of XSCID HSCs will result in selective advantage of gene-corrected lymphoid lineages with consequent restoration of lymphocyte populations and high probability of clinical benefit.


Assuntos
Células da Medula Óssea/metabolismo , Técnicas de Transferência de Genes , Ligação Genética , Terapia Genética/métodos , Imunodeficiência Combinada Severa/terapia , Cromossomo X/genética , Animais , Transplante de Medula Óssea , Separação Celular , Relação Dose-Resposta a Droga , Citometria de Fluxo , Proteínas de Fluorescência Verde , Humanos , Cinética , Leucócitos Mononucleares/metabolismo , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Retroviridae/genética
10.
Hum Gene Ther ; 11(13): 1901-9, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10986562

RESUMO

Dendritic cells (DCs) are potent antigen-presenting cells and are capable of activating naive T cells. Gene transfer of tumor antigen and cytokine genes into DCs could be an important strategy for immunotherapeutic applications. Dendritic cells derived from peripheral blood monocytes do not divide and are therefore poor candidates for gene transfer by Moloney murine leukemia virus (Mo-MuLV)-based retroviral vectors. Lentiviral vectors are emerging as a powerful tool for gene delivery into dividing and nondividing cells. A three-plasmid expression system pseudotyped with the envelope from vesicular stomatitis virus (VSV-G) was used to generate lentiviral vector particles expressing enhanced green fluorescent protein (EGFP). Peripheral blood monocyte-derived DCs were cultured in the presence of GM-CSF and IL-4 and transduced with lentiviral or Mo-MuLV-based vectors expressing EGFP. FACS analysis of lentiviral vector-transduced DCs derived either from normal healthy volunteers or from melanoma patients demonstrated transduction efficiency ranging from 70 to 90% compared with 2-8% using Mo-MuLV-based vectors pseudotyped with VSV-G. Comparison of lentiviral vectors expressing EGFP driven by CMV or human PGK promoters showed similar levels of transgene expression. Lentiviral vector preparations produced in the absence of HIV accessory proteins transduced DCs at efficiencies equal to vectors produced with accessory proteins. Alu-HIV-1 LTR PCR demonstrated the genomic integration of the lentiviral vector in the transduced DCs. Transduced cells showed characteristic dendritic cell phenotype and strong allostimulatory capacity and maintained the ability to respond to activation signals such as CD40 ligand and lipopolysaccharide. These results provide evidence that lentiviral vectors are efficient tools for gene transfer and expression in monocyte-derived DCs that could be useful for immunotherapeutic applications.


Assuntos
Células Dendríticas/fisiologia , Técnicas de Transferência de Genes , Lentivirus/genética , Monócitos/citologia , Elementos Alu/genética , Antígenos CD , Citomegalovirus/genética , Células Dendríticas/imunologia , Células Dendríticas/virologia , Proteínas de Fluorescência Verde , HIV-1/genética , Humanos , Imunoglobulinas/metabolismo , Interleucina-12/metabolismo , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Melanoma/genética , Melanoma/patologia , Glicoproteínas de Membrana/metabolismo , Vírus da Leucemia Murina de Moloney/genética , Monócitos/virologia , Regiões Promotoras Genéticas , Linfócitos T/imunologia , Vírus da Estomatite Vesicular Indiana/genética , Proteínas Virais/genética , Antígeno CD83
11.
Hum Gene Ther ; 8(13): 1555-63, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9322088

RESUMO

Myoblasts have properties that make them suitable vehicles for gene replacement therapy, and lysosomal storage diseases are attractive targets for such therapy. Type II Glycogen Storage Disease, a deficiency of acid alpha-glucosidase (GAA), results in the abnormal accumulation of glycogen in skeletal and cardiac muscle lysosomes. The varied manifestations of the enzyme deficiency in affected patient are ultimately lethal. We used a retroviral vector carrying the cDNA encoding for GAA to replace the enzyme in deficient myoblasts and fibroblasts and analyzed the properties of the transduced cells. The transferred gene was efficiently expressed, and the de novo-synthesized enzyme reached lysosomes where it digested glycogen. In enzyme-deficient myoblasts after transduction, enzyme activity rose to more than 30-fold higher than in normal myoblasts and increased about five-fold more when the cells were allowed to differentiate into myotubes. The transduced cells secreted GAA that was endocytosed via the mannose-6-phosphate receptor into lysosomes of deficient cells and digested glycogen. Moreover, the transduced myoblasts were able to fuse with and provide enzyme for GAA-deficient fusion partners. Thus, the gene-corrected cells, which appear otherwise normal, may ultimately provide phenotypic correction to neighboring GAA-deficient cells by fusion and to distant cells by secretion and uptake mechanisms.


Assuntos
Técnicas de Transferência de Genes , Glucana 1,4-alfa-Glucosidase/genética , Músculos/metabolismo , Retroviridae/genética , Fusão Celular , DNA Complementar , Terapia Genética , Genótipo , Glucana 1,4-alfa-Glucosidase/metabolismo , Doença de Depósito de Glicogênio Tipo II/terapia , Humanos , Microscopia Eletrônica , Músculos/patologia , Fenótipo , Transdução Genética , alfa-Glucosidases
12.
Cancer Gene Ther ; 7(4): 574-80, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10811475

RESUMO

Metabolic suicide gene transfer is widely applied for gene therapy of cancer, and retroviral vectors expressing the herpes simplex virus thymidine kinase (HSV-tk) gene are commonly used in clinical trials. Most of these vectors contain positive selectable markers that undoubtedly facilitate the determination of viral titer and the identification of high-titer producer clones. However, the presence of additional transcriptional units may result in reduced expression of the gene of interest. The use of fusion genes expressing bifunctional proteins may help to overcome this problem. We have constructed a retroviral vector carrying the TNFUS69 chimeric gene, which originates from the fusion of the HSV-tk and neomycin phosphotransferase II genes, and evaluated the functional expression of the encoded fusion protein. In vitro, expression of the fusion gene conferred to target cells both resistance to neomycin and selective sensitivity to the antiherpetic drugs ganciclovir and (E)-5-(2-bromovinyl)-2'-deoxyuridine. Cells transduced with the fusion gene, however, showed reduced ability to phosphorylate ganciclovir compared with cells expressing the native HSV-tk. Therefore, although the fusion gene may be used as a constituent of retroviral cassettes for positive and negative selection in vitro, its usefulness for suicide gene transfer applications in vivo may depend upon the possibility of using (E)-5-(2-bromovinyl)-2'-deoxyuridine in a clinical context.


Assuntos
Antivirais/toxicidade , Bromodesoxiuridina/análogos & derivados , Ganciclovir/toxicidade , Canamicina Quinase/genética , Timidina Quinase/genética , Transfecção , Células 3T3 , Adenocarcinoma , Animais , Bromodesoxiuridina/toxicidade , Neoplasias do Colo , Fibrossarcoma , Ganciclovir/farmacocinética , Vetores Genéticos , Canamicina Quinase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Plasmídeos , Proteínas Recombinantes de Fusão/biossíntese , Retroviridae , Simplexvirus/enzimologia , Simplexvirus/genética , Timidina Quinase/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas
13.
Curr Opin Allergy Clin Immunol ; 1(6): 497-501, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11964732

RESUMO

Primary immunodeficiency diseases have been important targets of corrective gene transfer approaches since the very early days of gene therapy. The potential for selective survival advantage of gene-corrected cells over populations carrying the mutated, causative gene translates into the possibility of obtaining clinical meaningful results in patients with primary immunodeficiency diseases even if levels of gene transfer are low. This critical prospect has fueled the interest of researchers since the mid-1980s and has recently determined the success of a clinical trial of gene therapy for X-linked severe combined immunodeficiency.


Assuntos
Terapia Genética/métodos , Síndromes de Imunodeficiência/terapia , Animais , Humanos , Síndrome da Aderência Leucocítica Deficitária/terapia , Camundongos , Imunodeficiência Combinada Severa/terapia , Cromossomo X
14.
Immunobiology ; 202(2): 106-19, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10993286

RESUMO

Combined immune deficiencies comprise a spectrum of genetic disorders characterized by developmental or functional defects of both T and B lymphocytes. Recent progress in cell biology and molecular genetics has unraveled the pathophysiology of most of these defects. In particular, the most common form of severe combined immune deficiency in humans, with lack of circulating T cells, a normal or increased number of B lymphocytes, and an X-linked pattern of inheritance (SCIDXI) has been shown to be due to defects of the IL2RG gene, encoding for the common gamma chain (gammac), shared by several cytokine receptors. Furthermore, defects of the JAK3 gene, encoding for an intracellular tyrosine kinase required for signal transduction through gammac-containing cytokine receptors, have been identified in patients with autosomal recessive T-B+ SCID. Characterization of the functional properties of cytokines that signal through the gammac-JAK3 signaling pathway has been favored by the detailed analysis of SCID patients. Specifically, the key role of IL-7 in promoting T cell development has been substantiated by the identification of rare patients with T-B+ SCID who have a defect in the alpha subunit of the IL-7 receptor (IL7Ralpha). The heterogeneity of genetic defects along the same signaling pathway that may lead to combined immune deficiency is paralleled by the heterogeneity of immunological phenotypes that may associate with defects in the same gene, thus creating a need for detailed immunological and molecular investigations in order to dissect the spectrum of combined immune deficiencies in humans.


Assuntos
Proteínas Tirosina Quinases/imunologia , Receptores de Interleucina-7/imunologia , Imunodeficiência Combinada Severa/imunologia , Transdução de Sinais , Animais , Citocinas/imunologia , Humanos , Imunofenotipagem , Subunidade gama Comum de Receptores de Interleucina , Janus Quinase 3 , Modelos Imunológicos , Receptores de Citocinas/imunologia
15.
DNA Cell Biol ; 12(7): 645-9, 1993 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8397823

RESUMO

The Wiskott-Aldrich syndrome (WAS) is a severe X-linked, recessive disorder, with a high mortality rate at early age due to hemorrhages, infections, and lymphoid malignancies. The molecular pathogenesis of the disease is unknown. Carrier females of WAS are clinically and immunologically normal, thus precluding carrier detection by simple laboratory tests. Major advances in molecular genetics have allowed mapping of the WAS gene to the pericentromeric short arm of the X chromosome, and have made carrier detection and prenatal diagnosis feasible by segregation analysis with closely linked polymorphic DNA markers. Furthermore, the observation that carriers of WAS exhibit a unilateral inactivation of the X chromosome in hematopoietic cells has provided a new tool for carrier detection. However, critical interpretation of molecular analysis data is essential to provide accurate genetic counseling to WAS families.


Assuntos
Triagem de Portadores Genéticos , Aconselhamento Genético , Síndrome de Wiskott-Aldrich/genética , Cromossomo X , Mecanismo Genético de Compensação de Dose , Feminino , Ligação Genética , Marcadores Genéticos , Testes Genéticos , Humanos , Linhagem , Síndrome de Wiskott-Aldrich/psicologia
16.
Cell Transplant ; 10(1): 81-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11294475

RESUMO

Ex vivo embryonic liver explant culture is a novel and attractive approach to obtain abundant hepatic and hematopoietic stem cells. Gene therapy of autologous hepatic and hematopoietic stem cells represents an alternative therapeutic approach to liver transplantation for genetic and metabolic disorders. In this study we characterize the growth and differentiation of hepatic stem cells utilizing embryonic liver cultures. Day 9.5 liver buds are microdissected and cultured under specific conditions. Modulation of growth conditions by addition of hepatocyte growth factor, Flt-3 ligand, and stem cell factor leads to enrichment of hepatic progenitor cells in embryonic liver explants. Under these conditions, we also demonstrate the role of a novel marker PRAJA-1 to identify hepatic stem cells and transitional hepatocytes. Utilization of dexamethasone enhanced pseudolobule formation with increased hepatocytic and biliary differentiation. Transforming growth factor-beta leads to enrichment of biliary cells in the culture. Gut formation is enhanced in the presence of interleukin-3 and blood formation by increasing the mesodermal tissue in these cultures. We also show increased retroviral-mediated expression of the green fluorescent protein expression in the expanded hepatic and hematopoietic stem cells under different culture conditions. Thus, the embryonic liver explant culture is an attractive source for hepatic progenitors and is a possible step towards generating nontumorigenic immortalized hepatocytes with possible transplantation applications.


Assuntos
Transplante de Tecido Fetal/métodos , Células-Tronco Hematopoéticas/citologia , Hepatócitos/citologia , Transplante de Fígado/métodos , Animais , Sistema Biliar/citologia , Diferenciação Celular/efeitos dos fármacos , Feminino , Transplante de Tecido Fetal/patologia , Terapia Genética , Fator de Crescimento de Hepatócito/farmacologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/transplante , Transplante de Fígado/patologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos ICR , Fenótipo , Gravidez , Fator de Células-Tronco/farmacologia , Transplante de Células-Tronco , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos
17.
Pediatr Clin North Am ; 47(6): 1389-407, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11131002

RESUMO

Several hurdles remain before gene therapy will be a part of mainstream medical therapy; however, the preliminary report of success in HSC correction in patients with XSCID provides hope that gene therapy will become a reality.


Assuntos
Terapia Genética/métodos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/terapia , Animais , Pré-Escolar , Feminino , Humanos , Síndromes de Imunodeficiência/diagnóstico , Lactente , Recém-Nascido , Masculino , Prognóstico , Resultado do Tratamento
18.
Pediatr Med Chir ; 13(2): 135-8, 1991.
Artigo em Italiano | MEDLINE | ID: mdl-1896378

RESUMO

Severe combined immunodeficiencies are usually fatal diseases unless affected children are admitted to protective isolation unit or unless the underlying immunological defect is treated by transplanting bone marrow from an healthy donor. The patients present, with early onset, life-threatening infections from fungal, viral or bacterial agents. Since only a minority of patients has an HLA-identical donor, recently other strategies have been devised including bone marrow transplantation from donors other than HLA-identical within the family or from HLA-non identical family members or from HLA-matched unrelated donors included in the International Bone Marrow Volunteers' Registry. In these cases, in order to realize this approach the "purging" of T-cells of the HLA-non identical donor bone marrow is necessary. Overall survival after HLA-identical BMT is 76%, when all BMT of the European multicenter analysis are considered, while in BMT from non-identical donors is 56%. Recently particular cases of SCID caused by enzyme deficiency, such as adenosine-deaminase (ADA), have been treated by molecular therapy with administration of polyethylene glycol (PEG) conjugated ADA: PEG protects from degradation and inhibits clearance of the enzyme. This approach, already realized in 15 children, allows reconstitution of cellular and humoral immunity, as demonstrated by one case treated by our group.


Assuntos
Adenosina Desaminase/uso terapêutico , Transplante Ósseo , Síndromes de Imunodeficiência/terapia , Criança , Antígenos HLA/análise , Teste de Histocompatibilidade , Humanos , Síndromes de Imunodeficiência/mortalidade , Lactente , Doadores de Tecidos
19.
Pediatr Med Chir ; 13(1): 27-30, 1991.
Artigo em Italiano | MEDLINE | ID: mdl-2052452

RESUMO

BMT can cure several congenital immunological defects: if in these disease the engrafting is easier, the GVH reactions are more frequent and severe. The possibility to deplete from T lymphocyte the marrow before infusion, has overcame this difficulty. From 1968 183 BMT have been performed in Europe on patients with SCID (70 from HLA-identical donor, 113 from HLA-nonidentical donor). The survival after 2 years is 76% in the first group, and 56% in the second group (100 marrows have been T-depleted with different techniques). Strict isolation procedures before the transplant are very important to achieve good results. The possibility to treat different immunodeficiency With BMT are also discussed.


Assuntos
Transplante de Medula Óssea , Síndromes de Imunodeficiência/terapia , Transplante de Medula Óssea/imunologia , Transplante de Medula Óssea/métodos , Criança , Pré-Escolar , Europa (Continente) , Antígenos HLA/imunologia , Teste de Histocompatibilidade , Humanos , Imunidade Celular/imunologia , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/mortalidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA