Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
1.
J Eur Acad Dermatol Venereol ; 36(1): 133-143, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34543471

RESUMO

BACKGROUND: Dystrophic epidermolysis bullosa pruriginosa (DEB-Pr) is a rare subtype of hereditary epidermolysis bullosa, with a poorly understood pathogenesis and no satisfactory treatment. OBJECTIVES: To assess the clinical and biological features, genetic basis and therapeutic management, to better characterize this rare genodermatosis. METHODS: We have conducted a retrospective study, reviewing the clinical presentation, genetic diagnosis, immunohistopathological findings and biological characteristics and management of patients with dystrophic epidermolysis bullosa pruriginosa. This study was conducted in the Department of Dermatology at Saint-Louis Hospital and the Department of Genetics at Necker Hospital (Paris, France). All patients with a diagnosis of DEB-Pr seen between 2010 and 2020 were included. RESULTS: Seven patients were included, the average age of 50.1 years [range 36-76]. Pruriginous-lichenified papules, plaques or nodules appeared at 27.6 years on average [range 7-66] on pretibial areas and forearms, associated with milia and toenails dystrophy. All patients received multiple treatments, but none could sustainably reduce pruritus. Immunohistopathological analysis of lesion skin revealed subepidermal blister with fibrosis, milia and mast cell infiltration. Serum TNFα, IL1ß and IL6 levels were elevated in 2/6 patients. Total serum IgE levels were increased in 7/7 patients, with no history of atopy. Immunophenotyping of circulating T-cells revealed an increased Th2 subset in 4/4 patients, with reduced Th1 and Th17 subpopulations. Genetic analysis of COL7A1 identified 7 distinct causative mutations, six of which were new. Intra-familial clinical variability was documented in 5/7 patients and was associated with the co-inheritance of a recessive COL7A1 mutation or an FLG2 mutation in 2 families. CONCLUSION: Our study confirms the stereotyped presentation of DEB-Pr with large intra-familial variability in disease expression. Mast cell infiltration, elevated IgE and increased Th2 subset without atopy strongly support a role of Th2-mediated immunity in DEB-Pr, and further argue for new targeted therapeutic options such as dupilumab.


Assuntos
Colágeno Tipo VII , Epidermólise Bolhosa Distrófica , Proteínas Filagrinas/genética , Adulto , Idoso , Colágeno Tipo VII/genética , Epidermólise Bolhosa Distrófica/genética , Humanos , Pessoa de Meia-Idade , Mutação , Fenótipo , Estudos Retrospectivos
4.
Gene Ther ; 17(7): 930-7, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20376098

RESUMO

Recessive dystrophic epidermolysis bullosa (RDEB) is a severe genodermatosis caused by loss-of-function mutations in COL7A1 encoding type VII collagen, the component of anchoring fibrils. As exogenous type VII collagen may elicit a deleterious immune response in RDEB patients during upcoming clinical trials of gene therapies or protein replacement therapies, we developed enzyme-linked immunosorbent assay (ELISA) and enzyme-linked immunosorbent spot (ELISPOT) assays to analyze B- and T-cell responses, to the full-length type VII collagen. The ELISA was highly sensitive and specific when tested against sera from 41 patients with epidermolysis bullosa acquisita (EBA), and the IFN-gamma ELISPOT detected a cellular response that correlated with ongoing EBA manifestations. Both tests were next applied to assess the risk of an immune response to type VII collagen in seven RDEB patients with a range of type VII collagen expression profiles. Immune responses against type VII collagen were dependent on the expression of type VII collagen protein, and consequently on the nature and position of the respective COL7A1 mutations. These immunologic tests will be helpful for the selection of RDEB patients for future clinical trials aiming at restoring type VII collagen expression, and in monitoring their immune response to type VII collagen after treatment.


Assuntos
Colágeno Tipo VII/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Epidermólise Bolhosa Distrófica/imunologia , Imunidade Celular , Colágeno Tipo VII/genética , Epidermólise Bolhosa Distrófica/genética , Humanos , Mutação , Sensibilidade e Especificidade , Estudos de Validação como Assunto
5.
Clin Genet ; 74(4): 392-5, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18783408

RESUMO

Microphthalmia and anophthalmia are at the severe end of the spectrum of abnormalities in ocular development. Mutations in several genes have been involved in syndromic and non-syndromic anophthalmia. Previously, RAX recessive mutations were implicated in a single patient with right anophthalmia, left microphthalmia and sclerocornea. In this study, we report the findings of novel compound heterozygous RAX mutations in a child with bilateral anophthalmia. Both mutations are located in exon 3. c.664delT is a frameshifting deletion predicted to introduce a premature stop codon (p.Ser222ArgfsX62), and c.909C>G is a nonsense mutation with similar consequences (p.Tyr303X). This is the second report of a patient with anophthalmia caused by RAX mutations. These findings confirm that RAX plays a major role in the early stages of eye development and is involved in human anophthalmia.


Assuntos
Anoftalmia/genética , Proteínas do Olho/genética , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Anoftalmia/patologia , Pré-Escolar , Córnea/anormalidades , Feminino , Humanos , Dados de Sequência Molecular , Órbita/anormalidades , Análise de Sequência de DNA
6.
Gene Ther ; 13(19): 1422-9, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16724094

RESUMO

Posterior capsule opacification is the main complication of cataract surgery. Using adenovirus-mediated gene transfer, we recently reported that it was feasible to prevent PCO by overexpressing pro-apoptotic molecules such as pro-caspase 3 or Bax in the residual lens epithelial cells post-cataract surgery. However, this approach is feasible only if gene transfer can be restricted to the residual cells responsible for PCO. Initially, we tested an adenovirus (human serotype 5, HAd5), a lentivirus (HIV) and an oncoretrovirus (MLV) vector for the their in vivo transduction efficiency of rabbit lens cells. We found that HAd5 vectors were the most efficient (>90% of the cells could be transduced). Six potential lens-specific promoters were then cloned into HAd5 vectors and assayed for their ability to target expression to a specific population of cells, using in vitro, ex vivo and in vivo rabbit tissues and human lens capsular bags. We found that the LEP503, MIP and Filensin promoters induced strong lens-specific expression of a reporter gene, in human lens cells. Following this ex vivo assay, we showed in a rabbit PCO model that gene transfer could be spatially restricted to the capsular bag by confining the vector with Matrigel. Our combined approach using a lens-specific promoter and a biocompatible gel should render feasible a novel therapeutic strategy for PCO that targets the remaining lens cells.


Assuntos
Catarata/prevenção & controle , Cristalinas/genética , Terapia Genética/métodos , Cápsula do Cristalino/metabolismo , Regiões Promotoras Genéticas , Transdução Genética/métodos , Adenoviridae/genética , Idoso , Idoso de 80 Anos ou mais , Animais , Câmara Anterior , Aquaporinas/genética , Catarata/metabolismo , Catarata/patologia , Colágeno , Proteínas de Ligação a DNA/genética , Combinação de Medicamentos , Proteínas do Olho/genética , Expressão Gênica , Marcação de Genes , Engenharia Genética , Vetores Genéticos/administração & dosagem , Humanos , Imuno-Histoquímica/métodos , Injeções , Proteínas de Filamentos Intermediários/genética , Laminina , Cápsula do Cristalino/patologia , Glicoproteínas de Membrana/genética , Microscopia de Contraste de Fase , Modelos Animais , Proteoglicanas , Coelhos , Recidiva , beta-Galactosidase/genética
7.
Exp Cell Res ; 298(2): 431-44, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15265691

RESUMO

We have previously cloned and characterized the human synemin gene, which encodes two intermediate filament proteins (IFPs). We now show that the mouse synemin gene encodes three different synemin isoforms through an alternative splicing mechanism. Two of them, synemin H and M are similar to human alpha and beta synemin, and the third isoform, L synemin, constitutes a new form of IFP. It has a typical rod domain and a short tail (49 residues) with a novel sequence that is produced by a different open reading frame. The synthesis of H/M synemins starts in the embryo, whereas the synemin L isoform is present in adult muscles. The H/M isoforms are bound to desmin or vimentin in the muscle cells of wild-type mice. Using desmin- and vimentin-deficient mice, we have obtained direct evidence that synemin is associated with muscle intermediate filaments in vivo. The organization of the synemin fibril is disrupted in skeletal and cardiac muscle when desmin is absent and in smooth muscle when vimentin is absent. The fact that the three synemin isoforms differ in the sequences of their tail domains as well as in their developmental patterns suggests that they fulfill different functions.


Assuntos
Processamento Alternativo/genética , Proteínas de Filamentos Intermediários/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Sequência de Aminoácidos/genética , Animais , Animais Recém-Nascidos , Sequência de Bases/genética , DNA Complementar/análise , DNA Complementar/genética , Desmina/metabolismo , Éxons/genética , Feto , Proteínas de Filamentos Intermediários/genética , Proteínas de Filamentos Intermediários/isolamento & purificação , Camundongos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/citologia , Proteínas Musculares/genética , Proteínas Musculares/isolamento & purificação , Músculo Esquelético/embriologia , Músculo Esquelético/ultraestrutura , Fases de Leitura Aberta/genética , Especificidade de Órgãos , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Isoformas de Proteínas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vimentina/metabolismo
8.
Leukemia ; 18(4): 788-97, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-14990974

RESUMO

To examine the role of the Notch ligand Delta-4 on hematopoietic stem cells, human CD34+CD38low cord blood cells were cocultured on S17 cells transduced with transmembrane Delta-4 (mbD4/S17) or an empty vector (C/S17). By the end of a 3-week culture, mbD4/S17 induced a 25-fold reduction in nucleated cell production, as compared to C/S17, by maintaining a higher proportion of cells in G0/G1 phase. A specific retention of a high proportion of CD34+ cells throughout the culture was observed with mbD4/S17, contrary to C/S17. Although mbD4/S17 promoted expansion of cells with the phenotype of committed lymphoid precursors (CD34+CD7+CD45RA+), these cells still retained their myeloid differentiation potential. mbD4/S17 maintained a higher LTC-IC frequency in output CD34+ cells, compared to C/S17, as in the subsets of cells having completed the same number of divisions on mbD4/S17. A Delta4-Fc protein (extracellular part of human Delta4 fused to Fc human IgG1 portion), immobilized on plastic, also reduced cell production and retained the LTC-IC potential. Transplantation of cells grown on mbD4/S17 into NOD/SCID mice showed no significant enhancement of the long-term repopulating ability. Thus, Delta4 appears to inhibit hematopoietic stem cell proliferation, in association with the maintenance of short-term lymphoid and myeloid repopulation capacity.


Assuntos
Proteínas Sanguíneas/genética , Proteínas Sanguíneas/fisiologia , Células-Tronco Hematopoéticas/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , ADP-Ribosil Ciclase/análise , ADP-Ribosil Ciclase 1 , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos CD/análise , Antígenos CD34/análise , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Divisão Celular , Técnicas de Cocultura , Sangue Fetal/citologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Glicoproteínas de Membrana , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos NOD , Fase de Repouso do Ciclo Celular , Transdução Genética , Transplante Heterólogo
9.
J Hematother Stem Cell Res ; 11(2): 327-36, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11983104

RESUMO

Hematopoietic stem cells (HSC) are subject to great interest because of their medical importance and their biological properties. Therefore, the possibility of genetically modifying human HSC is a major concern in several inherited pathologies. In this study, we aimed to demonstrate that a murine oncoretroviral vector can transduce multipotential cord blood (CB) stem cells. Sorted CB CD34(+)CD38(low) cells were transduced with a Moloney-based MFG retroviral vector containing the coding sequence of the murine CD2 (mCD2). CD34(+)mCD2(+) cells were sorted by flow cytometry and cultured either in bulk or at one cell per well in culture conditions that allow differentiation along lymphoid (T, B, and NK) and myeloid (M) lineages. Phenotypic analysis of cells generated in culture showed that CD34(+)mCD2(+) cells could give rise to all lymphoid and myeloid progeny, indicating that the MFG/mCD2 vector had transduced progenitors of all tested lineages. Moreover, clonal cultures of 660 CD34(+)mCD2(+) cells showed that approximately 5% of these cells were able to generate both myeloid and lymphoid (B + NK) progenies; for 25% of them, this included the production of lymphoid T cells. We also demonstrate that transduced CD34(+)CD38(low) CB cells with lymphoid and myeloid potentials were capable of engraftment into the bone marrow (BM) of nonobese diabetic-severe combined immunodeficiency (NOD-SCID) mice during several months. These results show that MFG retroviral vectors can transduce multipotent (T, B, NK, M) human hematopoietic progenitors with in vivo repopulating activity.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Células-Tronco Multipotentes/metabolismo , Transdução Genética/métodos , Animais , Antígenos CD34 , Células da Medula Óssea , Antígenos CD2/genética , Movimento Celular , Genes Reporter , Vetores Genéticos , Sobrevivência de Enxerto , Células-Tronco Hematopoéticas/metabolismo , Humanos , Camundongos , Camundongos SCID , Transdução Genética/normas
10.
Eur J Biochem ; 268(24): 6435-49, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11737198

RESUMO

Intermediate filament (IF) proteins are constituents of the cytoskeleton, conferring resistance to mechanical stress, and are encoded by a dispersed multigene family. In man we have identified two isoforms (180 and 150 kDa) of the IF protein synemin. Synemin alpha and beta have a very short N-terminal domain of 10 amino acids and a long C-terminal domain consisting of 1243 amino acids for the alpha isoform and 931 amino acids for the beta isoform. An intronic sequence of the synemin beta isoform is used as a coding sequence for synemin alpha. Both mRNA isoforms (6.5 and 7.5 kb) result from alternative splicing of the same gene, which has been assigned to human chromosome 15q26.3. Analyses by Northern and Western blot revealed that isoform beta is the predominant isoform in striated muscles, whereas both isoforms (alpha and beta) are present in almost equal quantities in smooth muscles. Co-transfection and immunolabeling experiments indicate that both synemin isoforms are incorporated with desmin to form heteropolymeric IFs. Furthermore synemin and desmin are found aggregated together in certain pathological situations.


Assuntos
Processamento Alternativo , Proteínas de Filamentos Intermediários/genética , Proteínas Musculares/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Mapeamento Cromossômico , Cromossomos Humanos Par 15 , DNA Complementar , Humanos , Proteínas de Filamentos Intermediários/química , Dados de Sequência Molecular , Proteínas Musculares/química , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Homologia de Sequência de Aminoácidos
11.
Blood ; 96(13): 4103-10, 2000 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-11110680

RESUMO

Gene transfer in human hematopoietic stem cells (HSCs) has great potential for both gene therapy and the understanding of hematopoiesis. As HSCs have extensive proliferative capacities, stable gene transfer should include genomic integration of the transgene. Lentiviral vectors are now preferred to oncoretroviral vectors especially because they integrate in nondividing cells such as HSCs, thereby avoiding the use of prolonged cytokine stimulation. Human immunodeficiency virus type-1 (HIV-1) has evolved a complex reverse transcription strategy including a central strand displacement event controlled in cis by the central polypurine tract (cPPT) and the central termination sequence (CTS). This creates, at the center of HIV-1 linear DNA molecules, a 99-nucleotide-long plus-strand overlap, the DNA flap, which acts as a cis-determinant of HIV-1 genome nuclear import. The reinsertion of the DNA flap sequence in an HIV-derived lentiviral vector promotes a striking increase of gene transduction efficiency in human CD34(+) hematopoietic cells, and the complementation of the nuclear import defect present in the parental vector accounts for this result. In a short ex vivo protocol, the flap-containing vector allows efficient transduction of the whole hierarchy of human HSCs including both slow-dividing or nondividing HSCs that have multiple lymphoid and myeloid potentials and primitive cells with long-term engraftment ability in nonobese diabetic/severe combined immunodeficiency mice (NOD/SCID).


Assuntos
Núcleo Celular/metabolismo , DNA Complementar/metabolismo , DNA Viral/metabolismo , Vetores Genéticos/genética , HIV-1/genética , Células-Tronco Hematopoéticas/metabolismo , Transfecção/métodos , Adulto , Animais , Transporte Biológico , Células da Medula Óssea/metabolismo , Células da Medula Óssea/virologia , Divisão Celular , DNA Viral/química , Sangue Fetal/citologia , Vetores Genéticos/metabolismo , Sobrevivência de Enxerto , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/virologia , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Reação em Cadeia da Polimerase , Especificidade da Espécie , Relação Estrutura-Atividade , Transplante Heterólogo
12.
Exp Hematol ; 28(12): 1470-80, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11146169

RESUMO

The aim of the present report is to describe clinically relevant culture conditions that support the expansion of primitive hematopoietic progenitors/stem cells, with maintenance of their hematopoietic potential as assessed by in vitro assays and the NOD-SCID in vivo repopulating capacity.CD34(+) cord blood (CB) cells were cultured in serum-free medium containing stem cell factor, Flt3 ligand, megakaryocyte growth and development factor, and granulocyte colony-stimulating factor. After 14 days, the primitive functions of expanded and nonexpanded cells were determined in vitro using clonogenic cell (colony-forming cells, long-term culture initiating cell [LTC-IC], and extended [E]-LTC-IC) and lymphopoiesis assays (NK, B, and T) and in vivo by evaluating long-term engraftment of the bone marrow of NOD-SCID mice. The proliferative potential of these cells also was assessed by determining their telomere length and telomerase activity. Levels of expansion were up to 1,613-fold for total cells, 278-fold for colony-forming unit granulocyte-macrophage, 47-fold for LTC-IC, and 21-fold for E-LTC-IC. Lymphoid B-, NK, and T-progenitors could be detected. When the expanded populations were transplanted into NOD-SCID mice, they were able to generate myeloid progenitors and lymphoid cells for 5 months. These primitive progenitors engrafted the NOD-SCID bone marrow, which contained LTC-IC at the same frequency as that of control transplanted mice, with conservation of their clonogenic capacity. Moreover, human CD34(+)CDl9(-) cells sorted from the engrafted marrow were able to generate CD19(+) B-cells, CD56(+)CD3(-) NK cells, and CD4(+)CD8(+)alphabetaTCR(+) T-cells in specific cultures. Our expansion protocol also maintained the telomere length in CD34(+) cells, due to an 8.8-fold increase in telomerase activity over 2 weeks of culture. These experiments provide strong evidence that expanded CD34(+) CB cells retain their ability to support long-term hematopoiesis, as shown by their engraftment in the NOD-SCID model, and to undergo multilineage differentiation along all myeloid and the B-, NK, and T-lymphoid pathways. The expansion protocol described here appears to maintain the hematopoietic potential of CD34(+) CB cells, which suggests its relevance for clinical applications.


Assuntos
Antígenos CD34/análise , Diferenciação Celular , Sangue Fetal/citologia , Granulócitos/citologia , Células-Tronco Hematopoéticas/citologia , Linfócitos/citologia , Animais , Linfócitos B/citologia , Células Cultivadas , Sobrevivência de Enxerto , Fator Estimulador de Colônias de Granulócitos/farmacologia , Hematopoese , Transplante de Células-Tronco Hematopoéticas , Humanos , Células Matadoras Naturais/citologia , Proteínas de Membrana/farmacologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Fator de Células-Tronco/farmacologia , Linfócitos T/citologia , Trombopoetina/farmacologia
13.
Blood ; 93(9): 2859-66, 1999 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-10216080

RESUMO

Thrombopoietin (TPO) is a hematopoietic growth factor that regulates megakaryocytopoiesis and platelet production through binding to its receptor, Mpl, encoded by the c-mpl proto-oncogene. Circulating levels of TPO are regulated by receptor-mediated uptake and degradation. To better understand this mode of TPO regulation, we examined whether expression of Mpl was regulated by its ligand. Using RNase protection analysis, we found no differences in the levels of c-mpl transcripts in megakaryocytes (MKs) produced in vitro either in the presence or absence of TPO and in platelets (PLTs) obtained from mice hyperstimulated in vivo by ectopic secretion of TPO. Similarly, Western blot analysis of MKs produced in the presence or absence of TPO showed no difference in Mpl levels. Levels of Mpl, GpIIb, or P-selectin were virtually identical in platelet lysates obtained from normal, TPO knockout and mildly TPO-stimulated mice. In contrast, the expression of Mpl was significantly reduced in PLTs from severely thrombocythemic mice. These results show that TPO does not have a major effect on the transcription or translation of Mpl. However, they do suggest that an excess of circulating TPO can lead to the disappearance of Mpl from PLTs via catabolism.


Assuntos
Plaquetas/fisiologia , Megacariócitos/fisiologia , Proteínas de Neoplasias , Proteínas Proto-Oncogênicas/genética , Receptores de Citocinas , Trombocitopenia/genética , Trombopoetina/genética , Transcrição Gênica , Animais , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Selectina-P/sangue , Selectina-P/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/sangue , Receptores de Trombopoetina , Trombocitopenia/sangue , Trombopoetina/farmacologia , Trombopoetina/fisiologia , Transcrição Gênica/efeitos dos fármacos
14.
Oncogene ; 16(23): 2989-95, 1998 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-9662331

RESUMO

Spi-1 transcriptional activation and wild-type p53 extinction are two oncogenic alterations involved in the malignant transformation of erythroblasts during the Friend acute erythroleukemia. To dissect the contribution of these alterations in the deregulation of the differentiation and proliferation of erythroblasts, we generated spi-1 transgenic mice. Analysis of these animals revealed that Spi-1 overexpression was directly involved in the block of proerythroblast differentiation. However, the erythroleukemia that develops in these animals evolved in two steps. During the early step (HS1 step), non tumorigenic proerythroblasts remained strictly dependent upon erythropoietin (Epo) for their survival and proliferation. Later on, Epo-independent and tumorigenic proerythroblasts emerged (HS2 step) suggesting that other oncogenes cooperate with Spi-1 to lead to a fully malignant phenotype. By provirus tagging, we demonstrate that the HS1 step was clonal indicating that a cell selection must occur in vivo. Analysis of the nature of p53 in both the in vivo HS1 and HS2 proerythroblasts and in cultured erythroblastic cell lines showed that--p53 was normal in the HS1 primary tissues but was mutated in the HS1 cultured cell lines--p53 was frequently altered in HS2 primary tissues but was found normal in some mice. These data indicate that (i) the blockage of the erythroblast differentiation by Spi-1 occurs independently of p53 alteration (ii) p53 alteration is not necessary to confer Epo independence and tumorigenicity to spi-1 transgenic proerythroblasts.


Assuntos
Leucemia Eritroblástica Aguda/genética , Mutação , Proteínas Proto-Oncogênicas/fisiologia , Transativadores/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Células Clonais , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
15.
Blood ; 88(7): 2578-84, 1996 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-8839850

RESUMO

Mpl ligand (thrombopoietin [TPO]) is the physiological regulator of platelet production. In mice, mRNA encoding the Mpl ligand (Mpl-L) is predominantly found by Northern blot analysis in the liver and kidney. To investigate the mode of regulation of the Mpl-L gene, we have developed several experimental models of severe thrombocytopenia differing in their kinetics and an opposite model of chronic thrombocytosis. Northern analysis performed at various times after induction of a thrombocytopenic state demonstrates that, whatever the number of circulating platelets, no change in Mpl-L mRNA level occurs in liver and kidney. By ribonuclease protection assays, we analyzed the ratios between mRNAs coding for the wild-type Mpl-L form and various splice variants encoding inactive or nonsecreted Mpl-L proteins. No modification in levels of these various isoforms was detected confirming the data of a previous report. Because the highest level of Mpl-L bioactivity in sera was observed only in mice with drastically reduced numbers of both platelets and megakaryocytes, these results further suggest that not only platelets, but also megakaryocytes, must be involved in the regulation of the level of circulating Mpl-L. In addition, we show that no downregulation of wild-type Mpl-L mRNA and no change in the ratio of Mpl-L mRNA isoforms were detected in mice in which a chronic thrombocytosis was induced. Together, these different models extend and further confirm that the regulation of Mpl-L does not occur at a transcriptional level or by a modulation in the ratios of Mpl-L mRNA isoforms.


Assuntos
Regulação da Expressão Gênica , Trombocitopenia/genética , Trombocitose/genética , Trombopoetina/biossíntese , Animais , Plaquetas/imunologia , Plaquetas/metabolismo , Feminino , Fluoruracila/toxicidade , Soros Imunes/toxicidade , Rim/metabolismo , Fígado/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , RNA Mensageiro/biossíntese , Lesões Experimentais por Radiação/genética , Trombocitopenia/etiologia , Trombopoetina/genética
16.
Mol Cell Biol ; 16(5): 2453-63, 1996 May.
Artigo em Inglês | MEDLINE | ID: mdl-8628313

RESUMO

Insertional mutagenesis of the spi-1 gene is associated with the emergence of malignant proerythroblasts during Friend virus-induced acute erythroleukemia. To determine the role of spi-1/PU.1 in the genesis of leukemia, we generated spi-1 transgenic mice. In one founder line the transgene was overexpressed as an unexpected-size transcript in various mouse tissues. Homozygous transgenic animals gave rise to live-born offspring, but 50% of the animals developed a multistep erythroleukemia within 1.5 to 6 months of birth whereas the remainder survived without evidence of disease. At the onset of the disease, mice became severely anemic. Their hematopoietic tissues were massively invaded with nontumorigenic proerythroblasts that express a high level of Spi-1 protein. These transgenic proerythroblasts are partially blocked in differentiation and strictly dependent on erythropoietin for their proliferation both in vivo and in vitro. A complete but transient regression of the disease was observed after erythrocyte transfusion, suggesting that the constitutive expression of spi-1 is related to the block of the differentiation of erythroid precursors. At relapse, erythropoietin-independent malignant proerythroblasts arose. Growth factor autonomy could be partially explained by the autocrine secretion of erythropoietin; however, other genetic events appear to be necessary to confer the full malignant phenotype. These results reveal that overexpression of spi-1 is essential for malignant erythropoiesis and does not alter other hematopoietic lineages.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Leucemia Eritroblástica Aguda/genética , Animais , Linhagem Celular , Chlorocebus aethiops , Proteínas de Ligação a DNA/genética , Éxons , Vírus da Leucemia Murina de Friend/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/patologia , Homozigoto , Leucemia Eritroblástica Aguda/patologia , Leucemia Eritroblástica Aguda/fisiopatologia , Fígado/citologia , Fígado/patologia , Camundongos , Camundongos Transgênicos , Mutagênese Insercional , Especificidade de Órgãos , Proteínas Oncogênicas de Retroviridae/biossíntese , Proteínas Oncogênicas de Retroviridae/genética , Baço/citologia , Baço/patologia , Transcrição Gênica , Transfecção
17.
Blood ; 87(5): 1746-53, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8634420

RESUMO

The human pluripotent UT-7 cell line is growth factor-dependent for proliferation and differentiation. We have previously shown that (1) granulocyte-macrophage colony-stimulating factor (GM-CSF) and erythropoietin (Epo) induce a myeloid and erythroid pattern of differentiation, respectively; (2) GM-CSF acts predominantly over Epo for cell differentiation; (3) GM-CSF induces a rapid downmodulation (4 hours) of Epo receptors (Epo-R) at the mRNA and binding site levels; and (4) in contrast, Epo has no effect on GM-CSF receptor (GM-CSF-R) expression. These results suggested that UT-7 cell commitment or differentiation may be directed by a hierarchical action of growth factors through an early and rapid transmodulation of growth factor receptors. To test this hypothesis, we introduced and expressed the murine Epo-R (muEpo-R) in UT-7 cells using a retroviral strategy. Two retroviral vectors were constructed: one carrying the neomycin resistance gene, and another carrying a mouse Epo-R cDNA devoid of its regulatory untranslated 3' sequence placed under the transcriptional control of the viral long terminal repeat element (LTR) and the neomycin resistance gene. Three UT-7/Epo-R infected clones (12, 6, 10) and one UT-7/neomycin clone (Neo) were selected in medium containing G418. After growth factor deprivation (18 hours), Epo-Rs were expressed at the same level (approximately 6,000 receptors per cell) in all four clones 12, 6, 10, Neo, and in parental UT-7 cells, and exhibited similar affinity (0.1 to 0.2 nmol/L). Cross-linking experiments showed that Epo is associated with three proteins of about 66, 85, and 100 kD in cells of parental UT-7, as well as in cells of clones 10 and 12. An inhibitory antibody directed specifically against the human Epo-R (huEpo-R Ab) abolished almost completely the cross-linking on parental UT-7 cells, but not on cells of clone 12, demonstrating that more than 90% cell surface Epo-Rs were of murine origin. The presence of GM-CSF significantly reduced the number of Epo-Rs expressed on parental UT-7 cells, but not on cells of clones 12, 10, and 6. HuEpo-R Ab inhibited Epo-induced parental UT-7 cell growth, but not that of cells of clone 12, suggesting that the muEpo-R is able to induce human UT-7 cell proliferation. When cells of clone 12 were switched from a medium containing GM-CSF to one with Epo, cell surface glycophorin A (GPA) was induced, as in parental UT-7 cells without inhibition by the huEpo-R Ab, demonstrating that the muEpo-R is also able to transduce a differentiation signal in human cells. However, in cells of clones 12, 6, 10 and Neo, as well as in parental UT-7 cells, the induction of GPA by Epo was inhibited by GM-CSF. This finding demonstrates that, although GM-CSF does not downregulate muEpo-R binding sites on UT-7/muEpo-R infected clones, it still inhibits the effects of Epo on cell differentation. Therefore, hierarchical regulation induced by growth factors for cell commitment or differntiation more likely acts downstream of cell surface receptors at either the signal transduction or transcriptional levels.


Assuntos
Eritropoese/efeitos dos fármacos , Eritropoetina/antagonistas & inibidores , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Receptores da Eritropoetina/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular , DNA Complementar/genética , Regulação para Baixo/efeitos dos fármacos , Vetores Genéticos/genética , Glicoforinas/biossíntese , Glicoforinas/genética , Humanos , Camundongos , Modelos Biológicos , Receptores da Eritropoetina/biossíntese , Receptores da Eritropoetina/genética , Proteínas Recombinantes de Fusão/biossíntese , Sequências Repetitivas de Ácido Nucleico , Retroviridae/genética , Transfecção
18.
Ann Pharm Fr ; 54(4): 177-82, 1996.
Artigo em Francês | MEDLINE | ID: mdl-8881104

RESUMO

After 35 years of research, the physiological regulator of platelet production has been isolated and its gene cloned. This discovery originates from studies performed with the myeloproliferative leukemia virus (MPLV), a murine retrovirus which induces an acute myeloproliferative syndrome in adult mice. MPLV carries in its genome the v-mpl oncogene which corresponds to a truncated form the c-mpl proto-oncogene. c-mpl encodes a cytokine receptor (Mpl-R) belonging to the hematopoietin receptor superfamily. Among the hematopoietic cell lineages, Mpl-R is preferentially expressed on late megakaryocyte progenitors, megakaryocytes and platelets. The ligand for Mpl-R, called Mpl-L or TPO or MGDF or megapoietin, is a glycosylated hormone of 352 amino acids in human which comprises two domains: the N-terminus domain shares 50% similarity with erythropoietin and is responsible for the biological activity; the C-terminus part is required for secretion. Notwithstanding its major action on megakaryocytopoiesis and thrombocytopoiesis, Mpl-L also potentiates the action of other cytokines on several hematopoietic lineages. Mpl-L/TPO/MGDF, the homeostatic regulator of platelet production, might be a useful therapeutical cytokine to treat thrombocytopenia induced in patients by chemotherapy.


Assuntos
Trombopoetina , Animais , Cães , Camundongos , Contagem de Plaquetas , Proto-Oncogene Mas , Coelhos , Trombocitopenia/tratamento farmacológico , Trombopoetina/genética , Trombopoetina/isolamento & purificação , Trombopoetina/metabolismo , Trombopoetina/uso terapêutico
19.
Thromb Haemost ; 74(1): 526-8, 1995 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8578519

RESUMO

v-mpl is a mouse oncogene that is part of the genome of the myeloproliferative leukemia virus (MPLV). The cellular homolog gene c-mpl encodes an hematopoietic cytokine receptor mainly restricted to the megakaryocytic lineage of differentiation. Using the Mpl receptor protein, cDNAs corresponding to the receptor ligand were isolated in humans, pigs, dogs and mice. In vivo and in vitro, the recombinant molecule has both the thrombopoietin (TPO) and megakaryocyte-colony stimulating factor (MK-CSF) activities. This molecule (Mpl ligand/TPO/MK-CSF) seems to be the humoral physiological regulator of platelet production.


Assuntos
Plaquetas/citologia , Hematopoese/fisiologia , Megacariócitos/citologia , Proteínas de Neoplasias , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia , Receptores de Citocinas , Trombopoetina/metabolismo , Animais , DNA Complementar/genética , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Cães , Genes Virais , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Camundongos , Camundongos Transgênicos , Oncogenes , Proto-Oncogenes , Receptores de Trombopoetina , Suínos
20.
C R Acad Sci III ; 318(4): 479-82, 1995 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7648360

RESUMO

v-mpl is a mouse oncogene that is part of the genome of the myeloproliferative leukemia virus (MPLV). The cellular homolog gene c-mpl encodes an hematopoietic cytokine receptor restricted to the megakaryocytic lineage of differentiation. Using the Mpl receptor protein, cDNAs corresponding to the receptor ligand were isolated in humans, pigs, dogs and mice. In vivo and in vitro, the recombinant molecule has both the thrombopoietin (TPO) and megakaryocyte-colony stimulating factor (MK-CSF) activities. This molecule (Mpl ligand/TPO/MK-CSF) is the humoral physiological regulator of platelet production.


Assuntos
Vírus da Leucemia Murina/genética , Oncogenes , Trombopoetina/metabolismo , Animais , Fatores Estimuladores de Colônias/genética , Fatores Estimuladores de Colônias/metabolismo , Cães , Humanos , Técnicas In Vitro , Ligantes , Megacariócitos/metabolismo , Camundongos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proto-Oncogenes/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA