Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Clin Exp Immunol ; 194(2): 166-179, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30251416

RESUMO

Dipeptidyl peptidase 4 (DPP4, CD26) is a serine protease that is expressed constitutively by many haematopoietic and non-haematopoietic tissues. It exists as a membrane-associated protein, as well as in an active, soluble form (herein called sDPP4), present at high concentrations in bodily fluids. Despite the proposed use of sDPP4 as a biomarker for multiple diseases, its cellular sources are not well defined. Here, we report that individuals with congenital lymphocyte immunodeficiency had markedly lower serum concentrations of sDPP4, which were restored upon successful treatment and restoration of lymphocyte haematopoiesis. Using irradiated lymphopenic mice and wild-type to Dpp4-/- reciprocal bone marrow chimeric animals, we found that haematopoietic cells were a major source of circulating sDPP4. Furthermore, activation of human and mouse T lymphocytes resulted in increased sDPP4, providing a mechanistic link between immune system activation and sDPP4 concentration. Finally, we observed that acute viral infection induced a transient increase in sDPP4, which correlated with the expansion of antigen-specific CD8+ T cell responses. Our study demonstrates that sDPP4 concentrations are determined by the frequency and activation state of lymphocyte populations. Insights from these studies will support the use of sDPP4 concentration as a biomarker for inflammatory and infectious diseases.


Assuntos
Biomarcadores/metabolismo , Dipeptidil Peptidase 4/metabolismo , Vírus da Influenza A/fisiologia , Proteínas de Membrana/metabolismo , Infecções por Orthomyxoviridae/imunologia , Imunodeficiência Combinada Severa/imunologia , Linfócitos T/imunologia , Animais , Secreções Corporais , Dipeptidil Peptidase 4/genética , Modelos Animais de Doenças , Hematopoese/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Solubilidade , Quimeras de Transplante
3.
Gene Ther ; 19(12): 1150-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22189416

RESUMO

Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency caused by the defective expression of the WAS protein (WASP) in hematopoietic cells. It has been shown that dendritic cells (DCs) are functionally impaired in WAS patients and was(-/-) mice. We have previously demonstrated the efficacy and safety of a murine model of WAS gene therapy (GT), using stem cells transduced with a lentiviral vector (LV). The aim of this study was to investigate whether GT can correct DC defects in was(-/-) mice. As DCs expressing WASP were detected in the secondary lymphoid organs of the treated mice, we tested the in vitro and in vivo function of bone marrow-derived DCs (BMDCs). The BMDCs showed efficient in vitro uptake of latex beads and Salmonella typhimurium. When BMDCs from the treated mice (GT BMDCs) and the was(-/-) mice were injected into wild-type hosts, we found a higher number of cells that had migrated to the draining lymph nodes compared with mice injected with was(-/-) BMDCs. Finally, we found that ovalbumin (OVA)-pulsed GT BMDCs or vaccination of GT mice with anti-DEC205 OVA fusion protein can efficiently induce antigen-specific T-cell activation in vivo. These findings show that WAS GT significantly improves DC function, thus adding new evidence of the preclinical efficacy of LV-mediated WAS GT.


Assuntos
Células Dendríticas/imunologia , Terapia Genética , Lentivirus/genética , Proteína da Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Animais , Células da Medula Óssea/imunologia , Movimento Celular , Células Dendríticas/metabolismo , Humanos , Tecido Linfoide/metabolismo , Camundongos , Modelos Genéticos , Fagocitose , Transdução Genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
4.
Curr Gene Ther ; 10(6): 414-22, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21054249

RESUMO

In the past 5 years, European investigators have played a major role in the development of clinical gene therapy. The provision of substantial funds by some individual member states to construct GMP facilities makes it an opportune time to network available gene therapy GMP facilities at an EU level. The integrated coordination of GMP production facilities and human skills for advanced gene and genetically-modified (GM) cell therapy, can dramatically enhance academic-led "First-in-man" gene therapy trials. Once proof of efficacy is gathered, technology can be transferred to the private sector which will take over further development taking advantage of knowledge and know-how. Complex technical challenges require existing production facilities to adapt to emerging technologies in a coordinated manner. These include a mandatory requirement for the highest quality of production translating gene-transfer technologies with pharmaceutical-grade GMP processes to the clinic. A consensus has emerged on the directions and priorities to adopt, applying to advanced technologies with improved efficacy and safety profiles, in particular AAV, lentivirus-based and oncolytic vectors. Translating cutting-edge research into "First-in-man" trials require that pre-normative research is conducted which aims to develop standard assays, processes and candidate reference materials. This research will help harmonise practices and quality in the production of GMP vector lots and GM-cells. In gathering critical expertise in Europe and establish conditions for interoperability, the PEVI infrastructure will contribute to the demands of the advanced therapy medicinal products* regulation and to both health and quality of life of EU-citizens.


Assuntos
Terapia Genética/tendências , Vetores Genéticos , Academias e Institutos , Transplante de Células/tendências , Ensaios Clínicos como Assunto , Desenho de Fármacos , Indústria Farmacêutica/normas , Europa (Continente) , Humanos
5.
Gene Ther ; 14(22): 1555-63, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17984995

RESUMO

This is the second part of a review summarizing progress and prospects in gene therapy clinical research. Twenty key diseases/strategies are succinctly described and commented on by leaders in the field. This part includes clinical trials for skin diseases, neurological disorders, HIV/AIDS, ornithine transcarbamylase deficiency, alpha(1)-antitrypsin deficiency, haemophilia and cancer.


Assuntos
Terapia Genética/tendências , Ensaios Clínicos como Assunto , Técnicas de Transferência de Genes/efeitos adversos , Técnicas de Transferência de Genes/tendências , Terapia Genética/métodos , Vetores Genéticos , Humanos , Neoplasias/terapia , Transplante de Células-Tronco/efeitos adversos , Transplante de Células-Tronco/tendências
6.
Gene Ther ; 14(5): 415-28, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17051251

RESUMO

Gene therapy has been proposed as a potential treatment for Wiskott-Aldrich syndrome (WAS), a severe primary immune deficiency characterized by multiple hematopoietic-specific cellular defects. In order to develop an optimal lentiviral gene transfer cassette for this application, we compared the performance of several internal promoters in a variety of cell lineages from human WAS patients. Vectors using endogenous promoters derived from short (0.5 kb) or long (1.6 kb) 5' flanking sequences of the WAS gene, expressed the transgene in T, B, dendritic cells as well as CD34(+) progenitor cells, but functioned poorly in non-hematopoietic cells. Defects of T-cell proliferation and interleukin-2 production, and the cytoskeletal anomalies in WAS dendritic cells were also corrected. The levels of reconstitution were comparable to those obtained following transduction with similar lentiviral vectors incorporating constitutive PGK-1, EF1-alpha promoters or the spleen focus forming virus gammaretroviral LTR. Thus, native regulatory sequences target the expression of the therapeutic WAS transgene to the hematopoietic system, as is naturally the case for WAS, and are effective for correction of multiple cellular defects. These vectors may have significant advantages for clinical application in terms of natural gene regulation, and reduction in the potential for adverse mutagenic events.


Assuntos
Terapia Genética/métodos , Células-Tronco Hematopoéticas/metabolismo , Lentivirus/genética , Transdução Genética/métodos , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/terapia , Antígenos CD34/imunologia , Linfócitos B/metabolismo , Sequência de Bases , Western Blotting/métodos , Linhagem Celular , Proliferação de Células , Células Cultivadas , Células Dendríticas/metabolismo , Expressão Gênica , Marcação de Genes/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/imunologia , Humanos , Interleucina-2/imunologia , Microscopia de Fluorescência , Dados de Sequência Molecular , Regiões Promotoras Genéticas/genética , Análise de Sequência de DNA , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Síndrome de Wiskott-Aldrich/metabolismo , Proteína da Síndrome de Wiskott-Aldrich/análise , Proteína da Síndrome de Wiskott-Aldrich/genética
9.
Hum Gene Ther ; 12(16): 1979-88, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11686939

RESUMO

The hematopoietic stem/progenitor cell (HSPC) represents the ideal target for gene therapy of disorders of the hematopoietic system, but still faces problems related to ex vivo manipulation and gene transfer efficiency. We demonstrate that soluble factors from the human endothelial-like cell line ECV 304/T24 support the growth of human CD34(+) progenitor cells as primary human bone marrow stroma and increase the rate of gene transfer into progenitor cells up to 5-fold. ECV 304/T24 was used to generate split-function amphotropic packaging cell lines (named APEX) with the purpose of combining, in the same cells, hematopoietic support and gene transfer vehicle functions. The APEX cell lines were negative for the presence of replication-competent retroviruses and produced complement-resistant vector particles. When mobilized peripheral blood or umbilical cord blood CD34(+) cells were exposed once to APEX supernatants, the level of gene transfer was equivalent to that observed with GP + Am12, in spite of the lower titer of the APEX producers. More importantly, APEX supernatants gave rise reproducibly to a 2-fold increase in transduction of early progenitors (long-term culture-initiating cells), reaching on average 50% gene transfer. This novel packaging cell represents a significant advance in HSPC genetic modification technology, combining both a beneficial hematopoietic supportive effect and the gene transfer vector function in a human-based system.


Assuntos
Células-Tronco Hematopoéticas/citologia , Retroviridae/genética , Transfecção , Montagem de Vírus , Antígenos CD34/análise , Linhagem Celular , Células-Tronco Hematopoéticas/imunologia , Humanos
10.
J Gene Med ; 3(3): 219-27, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11437327

RESUMO

BACKGROUND: Clinically applicable protocols for ex vivo modification of human CD34+ hematopoietic stem/progenitor cells rely on incubation of the target cell with supernatant containing recombinant retroviral particles. Although components of the supernatant may have a profound impact on both preclinical and clinical outcome, to date supernatant production has not been properly addressed with regard to CD34+ cells. We wanted to investigate and optimise production conditions for this target using simple, reproducible and clinically applicable procedures and reagents. METHODS: Retroviral supernatant was obtained from producer cell GP+Am12 under various production conditions and tested for bulk transduction efficiency and endpoint titre on murine and human cell lines. Gene transfer efficiency into CD34+ cells from mobilised peripheral blood, after a single exposure to retroviral supernatant, was measured by transgene expression, colony forming assay and long-term culture colony forming assay. RESULTS: Bulk gene transfer or endpoint titre values obtained on cell lines for the different production conditions were not predictive of gene transfer efficiency into hematopoietic progenitors. Time of virus production appeared to have the greatest impact on gene transfer, peaking at 6 h and decreasing 2-3-fold at longer time points. Neither the culture vessel used nor the temperature for virus production had any significant effect on gene transfer into CD34+ cells. Supernatant could be produced under defined serum-free conditions as efficiently as serum containing conditions for CD34+ cell gene transfer. CONCLUSIONS: The present data provide important implications for the establishment of quality controls for small- and large-scale clinical grade supernatant production for gene transfer into human hematopoietic stem/progenitor cells.


Assuntos
Vetores Genéticos , Células-Tronco Hematopoéticas/metabolismo , Retroviridae/genética , Transdução Genética , Transfecção/métodos , Células 3T3 , Animais , Antígenos CD34/biossíntese , Células Cultivadas , Técnicas de Transferência de Genes , Genes Virais/fisiologia , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/imunologia , Humanos , Proteínas Luminescentes/genética , Camundongos
11.
AIDS Res Hum Retroviruses ; 16(15): 1471-9, 2000 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-11054260

RESUMO

The mechanisms responsible for the hematopoietic failure in human immunodeficiency virus type 1 (HIV-1)-infected patients are still unknown. Several findings indicate that the in vitro proliferative potential of precursor cells from AIDS patients is reduced. The changes seen in bone marrow (BM) morphology and the defective BM functions associated with cytopenias have both been proposed as potential explanations. In patients treated with highly active antiretroviral therapy (HAART) an immune reconstitution associated with increased whole blood cell counts has been described. We have investigated the effects of HAART on the number of colony-forming cells (CFCs) and long-term culture-initiating cells (LTC-ICs), using long-term BM cell cultures (LTBMC) in a group of subjects with HIV-1 infection enrolled in an open study to evaluate the mechanisms of immune reconstitution during HAART. In each patient, the increase in colony growth was homogeneous, regardless of the type of hematopoietic progenitor cells assayed; in four subjects an increase in the most primitive progenitor cells (LTC-ICs) was observed. These findings were associated with the in vivo data showing increased numbers of BM mononuclear cells (BMMCs) after HAART and with a rise in peripheral CD4(+) T cell counts and decreased levels of plasma HIV-1 RNA. A decreased number of hematopoietic progenitor cells and/or a defective modulation of progenitor cell growth might be the cause of the hematological abnormalities in AIDS patients. Controlling HIV-1 replication by HAART could determine a restoration of stem cell activity, probably because of the suppression of factors that inhibit normal hematopoiesis.


Assuntos
Medula Óssea , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , HIV-1 , Hematopoese , Adulto , Terapia Antirretroviral de Alta Atividade , Medula Óssea/imunologia , Células Cultivadas , DNA Viral/análise , Feminino , Citometria de Fluxo , Infecções por HIV/sangue , HIV-1/genética , HIV-1/imunologia , Hematologia , Hematopoese/imunologia , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Masculino , Metilcelulose , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase/métodos , Fatores de Tempo
13.
Eur J Immunol ; 29(6): 1823-31, 1999 06.
Artigo em Inglês | MEDLINE | ID: mdl-10382744

RESUMO

Stromal cell-derived factor-1 (SDF-1) is a CXC chemokine produced by stromal cells that acts as a chemoattractant for human CD34+ progenitor cells. We investigated the expression of CXCR4, the receptor for SDF-1, on CD34+ cells from different hematopoietic sites and developmental stages. CXCR4 was detected by flow cytometry on 37 % of fetal bone marrow (BM) [gestation weeks (gw) 14-23] and 40% of adult BM CD34+ cells. Interestingly, in fetal liver CD34+ cells, CXCR4 was expressed at lower levels at later stages (9%, gw 20-23) compared to early stages of development (39%, gw 7.5-18), suggesting a development-related change in the migratory capacity of progenitors. CXCR4 was detected at similar levels on both phenotypically primitive and committed progenitors from fetal and adult sites. However, B cell lineage progenitor and precursor cells expressed CXCR4 at the highest density (80% of BM CD34+/CD10+ pro-B cells are CXCR4+). CXCR4 was also expressed in the fetal thymus in early T cell precursors and found to be down-regulated during T cell maturation. Finally, we found that stem cell factor, alone or in combination with other cytokines, can up-modulate CXCR4 expression on CD34+ cells by three- to fourfold. In conclusion, our results suggest that CXCR4 may play an important role in the local and systemic trafficking of human CD34+ cells as well as in human B lymphopoiesis and that its expression can be modulated by cytokines.


Assuntos
Quimiocinas CXC/metabolismo , Receptores CXCR4/metabolismo , Adulto , Antígenos CD34/metabolismo , Linfócitos B/citologia , Linfócitos B/imunologia , Linfócitos B/metabolismo , Quimiocina CXCL12 , Citocinas/farmacologia , Regulação para Baixo , Feto/metabolismo , Hematopoese , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Técnicas In Vitro , Fígado/citologia , Fígado/imunologia , Fígado/metabolismo , Células Estromais/citologia , Células Estromais/imunologia , Células Estromais/metabolismo , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
14.
Blood ; 94(1): 62-73, 1999 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-10381499

RESUMO

Human CD34(+) hematopoietic progenitor cells obtained from bone marrow (BM), umbilical cord blood (UCB), and mobilized peripheral blood (MPB) were purified and investigated for the expression of the chemokine receptor CXCR4 and its ligand, stromal cell-derived factor-1 (SDF-1). CXCR4 was found present on the cell surface of all CD34(+) cells, although it was expressed at lower density on MPB with respect to BM CD34(+) cells. Freshly isolated and in vitro-cultured CD34(+) cells also coexpressed SDF-1 mRNA, as determined by reverse transcriptase-polymerase chain reaction (RT-PCR). Of interest, CD34(+)/CD38(+) committed progenitor cells, unlike primitive CD34(+)/CD38(-) cells, expressed SDF-1 mRNA. Supernatants from in vitro-cultured CD34(+) cells contained substantial (3 to 8 ng/mL) amounts of SDF-1 by enzyme-linked immunosorbent assay and induced migration of CD34(+) cells. Because CD34(+) cells express low levels of CD4, the primary receptor of the human immunodeficiency virus (HIV), and CXCR4 is a coreceptor for T-cell tropic (X4) HIV strains, we investigated the susceptibility of CD34(+) cells to infection by this subset of viruses. Lack of productive infection was almost invariably observed as determined by a conventional RT activity in culture supernatants and by real-time PCR for HIV DNA in CD34(+) cells exposed to both laboratory adapted (LAI) and primary (BON) X4 T-cell tropic HIV-1 strain. Soluble gp120 Env (sgp120) from X4 HIV-1 efficiently blocked binding of the anti-CD4 Leu3a monoclonal antibody (MoAb) to either human CD4(+) T cells or CD34(+) cells. In contrast, sgp120 interfered with an anti-CXCR4 MoAb binding to human T lymphocytes, but not to CD34(+) cells. However, CXCR4 on CD34(+) cells was downregulated by SDF-1. These results suggest that CXCR4 and its ligand SDF-1 expressed in CD34(+) progenitors may play an important role in regulating the local and systemic trafficking of these cells. Moreover, these findings suggest multiple and potentially synergistic mechanisms at the basis of the resistance of CD34(+) cells to X4 HIV infection, including their ability to produce SDF-1, and the lack of CXCR4 internalization following gp120 binding to CD4.


Assuntos
Fármacos Anti-HIV/imunologia , Quimiocinas CXC/imunologia , Infecções por HIV/imunologia , HIV-1/fisiologia , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/virologia , Receptores CXCR4/imunologia , Fármacos Anti-HIV/farmacologia , Antígenos CD34 , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/farmacologia , Humanos , Receptores CXCR4/agonistas , Linfócitos T/imunologia , Linfócitos T/virologia , Replicação Viral/imunologia
15.
Blood ; 93(10): 3276-85, 1999 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-10233879

RESUMO

Targeted expression to specific tissues or cell lineages is a necessary feature of a gene therapy vector for many clinical applications, such as correction of hemoglobinopathies or thalassemias by transplantation of genetically modified hematopoietic stem cells. We developed retroviral vectors in which the constitutive viral enhancer in the U3 region of the 3' LTR is replaced by an autoregulatory enhancer of the erythroid-specific GATA-1 transcription factor gene. The replaced enhancer is propagated to the 5' LTR upon integration into the target cell genome. The modified vectors were used to transduce human hematopoietic cell lines, cord blood-derived CD34(+) stem/progenitor cells, and murine bone marrow repopulating stem cells. The expression of appropriate reporter genes (triangle upLNGFR, EGFP) was analyzed in the differentiated progeny of transduced stem cells in vitro, in liquid culture as well as in clonogenic assay, and in vivo, after bone marrow transplantation in lethally irradiated mice. The GATA-1 autoregulatory enhancer effectively restricts the expression of the LTR-driven proviral transcription unit to the erythroblastic progeny of both human progenitors and mouse-repopulating stem cells. Packaging of viral particles, integration into the target genome, and stability of the integrated provirus are not affected by the LTR modification. Enhancer replacement is therefore an effective strategy to target expression of a retroviral transgene to a specific progeny of transduced hematopoietic stem cells.


Assuntos
Proteínas de Ligação a DNA/genética , Eritroblastos/citologia , Células-Tronco Hematopoéticas/fisiologia , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Linhagem Celular , Proteínas de Ligação a DNA/biossíntese , Elementos Facilitadores Genéticos , Eritroblastos/fisiologia , Fatores de Ligação de DNA Eritroide Específicos , Fator de Transcrição GATA1 , Genes Reporter , Terapia Genética/métodos , Vetores Genéticos , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/citologia , Humanos , Células K562 , Leucemia Eritroblástica Aguda , Proteínas Luminescentes/biossíntese , Proteínas Luminescentes/genética , Camundongos , Proteínas Nucleares/biossíntese , Proteínas Nucleares/genética , Receptor de Fator de Crescimento Neural , Receptores de Fator de Crescimento Neural/biossíntese , Receptores de Fator de Crescimento Neural/genética , Sequências Reguladoras de Ácido Nucleico , Retroviridae , Fatores de Transcrição/biossíntese , Transfecção , Células Tumorais Cultivadas , Células U937
16.
Hepatology ; 28(6): 1645-54, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9828230

RESUMO

It was recently reported that transgenic expression in the liver of truncated human Met renders hepatocytes constitutively resistant to apoptosis and reproducibly permits their immortalization. The derived stable cell lines (MMH from Met murine hepatocyte) are highly differentiated and nontransformed. In this report, the capacity of MMHs to support in vitro hematopoiesis is characterized. By reverse-transcription polymerase chain reaction, the expression by MMHs of cytokines involved in the survival and self-renewal of early progenitor cells (stem cell factor and FLT3 ligand) as well as those acting at different stages of progenitor differentiation (interleukin [IL] 1beta, IL-3, leukemia inhibitory factor, IL-6, granulocyte-macrophage colony-stimulating factor, granulocyte colony-stimulating factor, macrophage colony-stimulating factor, and thrombopoietin) was shown. A ribonuclease protection assay further substantiated the presence of at least six cytokine transcripts in MMH lines. Cocultures between MMH layers and progenitor-enriched fetal liver hematopoietic cells resulted in a 40-fold to 80-fold expansion of total hematopoietic cells and in a 2.5-fold expansion of clonogenic progenitors after 1 to 2 weeks. Hematopoiesis was maintained for up to 6 weeks with formation of typical cobblestone cell areas and continuous differentiation of precursor into cells at various degrees of maturation. At 5 weeks of coculture, clonogenic progenitors were maintained at 20% of the input level in coculture with embryonic-derived hepatocytes, showing the ability of hepatocyte feeder layer to support survival and possibly self-renewal of clonogenic progenitors. Therefore, the data emphasize a direct role of the hepatocyte in sustaining hematopoietic cell proliferation and differentiation.


Assuntos
Citocinas/metabolismo , Hematopoese/fisiologia , Fígado/fisiologia , Animais , Animais Recém-Nascidos/anatomia & histologia , Diferenciação Celular/fisiologia , Linhagem Celular , Senescência Celular/fisiologia , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Feto/citologia , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Fígado/citologia , Fígado/embriologia , Camundongos , Camundongos Transgênicos/genética , Proteínas Proto-Oncogênicas c-met/química , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo
18.
Exp Hematol ; 26(2): 143-57, 1998 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9472804

RESUMO

Using a novel collection of conditionally immortalized mouse stromal cell clones, we evaluated the role of distinct elements of the hematopoietic microenvironment in supporting and regulating the growth, division, and differentiation of a candidate human stem cell population (CD34+/CD38-). We found functional diversity in the capacity of different stromal cell clones to support the growth of primitive (CD34+/CD38-) and committed (CD34+/CD38+) hematopoietic progenitors and their differentiation into mature hematopoietic cells (CD34-/CD45+). Among the stromal cell clones that supported long-term hematopoiesis, we identified two clones that induced expansion of CD34+ progenitor/stem cells during the first 4 weeks of coculture and that supported the maintenance of this CD34+ population for up to 10 weeks in vitro. However, these two clones appeared to represent two different microenvironments with regard to the signals they provide to the different CD34+ progenitor subpopulations: One stromal clone preserved a pool of undifferentiated, relatively quiescent (CD34+/CD38-) progenitor cells, allowing their differentiation at a low rate into more committed (CD34+/CD38+) progenitors; the other fostered a more extensive and rapid differentiation of all CD34+/CD38- progenitors into CD34+/CD38+ cells, preferentially maintaining this committed population at a higher rate of cell division. These stromal cell clones were also able to support the proliferation and differentiation of CD34+/CD38- cells in conditions in which progenitor-stroma contact was prevented. This collection of stromal cell clones may represent a unique tool for the study of stromal regulators of hematopoiesis as well as for the support of gene transfer into hematopoietic progenitor cells.


Assuntos
Células da Medula Óssea/citologia , Células-Tronco Hematopoéticas/citologia , Células Estromais/fisiologia , Animais , Antígenos CD/análise , Antígenos CD34/análise , Diferenciação Celular , Divisão Celular , Células Clonais , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Citometria de Fluxo , Humanos , Camundongos , Células Estromais/citologia , Fatores de Tempo
19.
Hum Gene Ther ; 8(13): 1611-23, 1997 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9322094

RESUMO

Human CD34+ cells lacking detectable levels of HLA-DR antigens (CD34+ DR-) are highly enriched in hematopoietic pluripotent progenitors with long-term marrow repopulating ability. We investigated the feasibility of transducing and marking CD34+ DR- progenitor cells from bone marrow (BM) or mobilized peripheral blood samples (MPB) of 13 patients undergoing BM transplantation with the purpose of developing a protocol for a large-scale clinical application. A new retroviral vector coding for the truncated form (delta) of the low-affinity nerve growth factor receptor (LNGFR) was used to quantitate the level of gene transfer into CD34+ cells and their progeny by multiparameter cytofluorimetry and immunocytochemistry. Light-density mononuclear cells as well as purified CD34+ cells were transduced either by direct incubation with retroviral supernatants or prestimulated in vitro with various combinations of growth factors prior to transduction. Transduction efficiency, assessed as G418-resistant growth of granulocyte-macrophage colony-forming units (CFU-GM) progenitors from MPB, was 1.7-fold higher (14.9% +/- 4.5%) than those from BM (8.5% +/- 3.9%) and it was further improved (26.9% +/- 3.1%) using a purified CD34+ population as target cells. Three-color fluorescence-activated cell sorting (FACS) analysis demonstrated the presence of transduced delta LNGFR+ cells within the CD34+ DR- subpopulation. In the absence of growth factors, gene transfer into BM or MPB CD34+ DR- cells was generally poor, but following a 72-hr prestimulation it peaked at 38% of total CD34+ DR- bone marrow (BM) cells in the presence of the c-kit ligand (KL) and at 31% in the presence of IL-3. Furthermore, KL gave, compared to the other cytokines, the highest absolute yield of BM delta LNGFR+ CD34+ DR- cells recovered after transduction (p = 0.05 compared to 24 hr). Gene transfer into in vitro primitive progenitor cells was further confirmed by expression of the delta LNGFR marker on CD34+ cells and CFU-GM derived from 5-week long-term culture on stroma.


Assuntos
Antígenos CD34/imunologia , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/imunologia , Retroviridae/genética , Adulto , Ciclofosfamida/administração & dosagem , Vetores Genéticos , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Mobilização de Células-Tronco Hematopoéticas , Humanos , Pessoa de Meia-Idade , Fenótipo , Transdução Genética
20.
J Exp Med ; 185(1): 111-20, 1997 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-8996247

RESUMO

Hematopoietic progenitor cells migrate in vitro and in vivo towards a gradient of the chemotactic factor stromal cell-derived factor-1 (SDF-1) produced by stromal cells. This is the first chemoattractant reported for human CD34+ progenitor cells. Concentrations of SDF-1 that elicit chemotaxis also induce a transient elevation of cytoplasmic calcium in CD34+ cells. SDF-1-induced chemotaxis is inhibited by pertussis toxin, suggesting that its signaling in CD34+ cells is mediated by seven transmembrane receptors coupled to Gi proteins. CD34+ cells migrating to SDF-1 include cells with a more primitive (CD34+/CD38- or CD34+/DR-) phenotype as well as CD34+ cells phenotypically committed to the erythroid, lymphoid and myeloid lineages, including functional BFU-E, CFU-GM, and CFU-MIX progenitors. Chemotaxis of CD34+ cells in response to SDF-1 is increased by IL-3 in vitro and is lower in CD34+ progenitors from peripheral blood than in CD34+ progenitors from bone marrow, suggesting that an altered response to SDF-1 may be associated with CD34 progenitor mobilization.


Assuntos
Antígenos CD34 , Antígenos CD , Quimiocinas CXC , Quimiocinas/farmacologia , Quimiotaxia/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Antígenos de Diferenciação , Medula Óssea/patologia , Células da Medula Óssea , Neoplasias da Mama/sangue , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Cálcio/metabolismo , Células Cultivadas , Quimiocina CCL4 , Quimiocina CXCL12 , Quimiocinas/isolamento & purificação , Meios de Cultivo Condicionados , Feminino , Sangue Fetal , Antígenos HLA-DR , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Linfoma não Hodgkin/sangue , Linfoma não Hodgkin/imunologia , Linfoma não Hodgkin/patologia , Proteínas Inflamatórias de Macrófagos/farmacologia , Glicoproteínas de Membrana , N-Glicosil Hidrolases , Gravidez , Células Estromais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA