Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Bone Marrow Transplant ; 50 Suppl 2: S55-62, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26039209

RESUMO

We are entering a very exciting era in umbilical cord blood transplantation (UCBT), where many of the associated formidable challenges may become treatable by ex vivo graft manipulation and/or adoptive immunotherapy utilizing specific cellular products. We envisage the use of double UCBT rather than single UCBT for most patients; this allows for greater ability to treat larger patients as well as to manipulate the graft. Ex vivo expansion and/or fucosylation of one cord will achieve more rapid engraftment, minimize the period of neutropenia and also give certainty that the other cord will provide long-term engraftment/immune reconstitution. The non-expanded (and future dominant) cord could be chosen for characteristics such as better HLA matching to minimize GvHD, or larger cell counts to enable part of the unit to be utilized for the development of specific cellular therapies such as the production of virus-specific T-cells or chimeric-antigen receptor T-cells which are reviewed in this study.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Facilitação Imunológica de Enxerto/métodos , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/prevenção & controle , Engenharia Tecidual/métodos , Humanos
2.
Cytotherapy ; 7(6): 509-19, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16306013

RESUMO

BACKGROUND: Human mesenchymal stem cells (hMSC) have been isolated and characterized extensively for a variety of clinical applications. Yet it is unclear how the phenomenon of hMSC plasticity can be safely and reasonably exploited for therapeutic use. METHODS: We have generated mesenchymal stem cells (MSC) from normal human BM and identified a novel cell population with a transformed phenotype. This cell population was characterized by morphologic, immunophenotypic, cytogenetic analyzes and telomerase expression. Its tumorigenicity in NOD/SCID mice was also studied. RESULTS: A subpopulation of cells in hMSC culture was noted to appear morphologically distinct from typical MSC. The cells were spherical, cuboidal to short spindle in shape, adherent and exhibited contact independent growth. Phenotypically the cells were CD133(+), CD34(-), CD45(-), CD90(low), CD105(-), VEGFR2(+). Cytogenetic analysis showed chromosome aneuploidy and translocations. These cells also showed a high level of telemerase activity compared with typical MSC. Upon transplantation into NOD/SCID mice, multiple macroscopic solid tumors formed in multiple organs or tissues. Histologically, these tumors were very poorly differentiated and showed aggressive growth with large areas of necrosis. DISCUSSION: The possible explanations for the origin of this cell population are: (1) the cells represent a transformed population of MSC that developed in culture; (2) abnormal cells existed in the donor BM at rare frequency and subsequently expanded in culture. In either case, the MSC culture may provide a suitable environment for transformed cells to expand or propagate in vitro. In summary, our data demonstrate the potential of transformed cells in hMSC culture and highlight the need for karyotyping as a release criteria for clinical use of MSC.


Assuntos
Células da Medula Óssea/patologia , Células-Tronco Mesenquimais/patologia , Neoplasias Experimentais/patologia , Células-Tronco Neoplásicas/patologia , Antígeno AC133 , Aneuploidia , Animais , Antígenos CD/análise , Células da Medula Óssea/imunologia , Linhagem Celular Transformada , Glicoproteínas/análise , Humanos , Imunofenotipagem , Cariotipagem , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias Experimentais/enzimologia , Células-Tronco Neoplásicas/imunologia , Peptídeos/análise , Fatores de Tempo
3.
Bone Marrow Transplant ; 36(5): 425-30, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15980880

RESUMO

The purpose of this study was to evaluate pre-transplant T-cell status in autologous hematopoietic progenitor-cell transplantation (HPCT) recipients. Between 1999 and 2002 we prospectively enrolled 85 autologous HPCT recipients with solid tumors (N = 50) or hematological malignancies (n = 35). Patient diagnoses included breast cancer (N = 49), non-Hodgkin's lymphoma (N = 20), myeloma (N = 11), Hodgkin's disease (N = 3), germ-cell tumor (N = 1) and amyloidosis (N = 1). Levels of CD3, CD4, CD8, memory and naïve CD4, and CD8 T-cell subsets were analyzed before autologous HPCT. Autologous HPCT recipients presented with lower pre-transplant counts of CD3, CD4, but not CD8 T cells, as compared to healthy controls. Pre-transplant CD4 T-cell levels correlated with progression-free survival (PFS) (P = 0.002) and overall survival (OS) (P = 0.05), in patients with hematologic malignancies (P = 0.02) and breast cancer (P = 0.04). Specifically, pre-transplant memory CD4 + CD45RA - CD62L - T-cell levels correlated with PFS (P = 0.01). The prognostic effects of pre-transplant CD4 and CD4 + CD45RA - CD62L - T cells were independent of tumor diagnosis, tumor stage, tumor sensitivity, and, for breast cancer patients, Her2 / neu status. Our results suggest that pre-transplant CD4 T-cell status, specifically CD4 + CD45RA - CD62L - memory T cells, correlates with the outcome of autologous HPCT recipients. These observations suggest the feasibility of prospective identification of those patients at higher risk of relapse, based on their immune status.


Assuntos
Linfócitos T CD4-Positivos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Recidiva Local de Neoplasia , Neoplasias/terapia , Adulto , Antígenos CD/sangue , Contagem de Linfócito CD4 , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Neoplasias/sangue , Neoplasias/fisiopatologia , Estudos Retrospectivos , Fatores de Risco , Transplante Autólogo , Resultado do Tratamento
5.
J Biol Chem ; 276(31): 28954-60, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11387320

RESUMO

The hematopoietic cells from patients with Bcr-Abl-positive chronic myelogenous leukemia exhibit multiple abnormalities of cytoskeletal function. The molecular events leading to these abnormalities are not fully understood. Previously we showed that Bcr-Abl elicits ubiquitin-dependent degradation of Abl interactor proteins. Because recent studies have suggested a role of Abl interactor proteins in the pathway that regulates cytoskeletal function, we investigated whether mutations in Bcr-Abl that interfere with the signaling to Abl interactor proteins affect its leukemogenic activity. We report here that the Src homology 3 domain and C-terminal proline-rich sequences of Bcr-Abl are required for its binding to Abl interactor 2 as well as for the induction of Abl interactor 2 degradation. Although the deletion of these regions did not affect the ability of the mutant Bcr-Abl to transform hematopoietic cells to growth factor independence, it abrogated its ability to stimulate spontaneous cell migration on fibronectin-coated surfaces. Furthermore, the mutant Bcr-Abl, defective in binding to Abl interactor 2 and inducing its degradation, failed to induce chronic myelogenous leukemia-like disease in mouse. These results are consistent with a role of Abl interactor proteins in the regulation of cytoskeletal function as well as in the pathogenesis of Bcr-Abl-induced leukemogenesis.


Assuntos
Proteínas de Fusão bcr-abl/genética , Proteínas de Fusão bcr-abl/metabolismo , Proteínas de Homeodomínio/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Ubiquitinas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Sequência de Aminoácidos , Animais , Transplante de Medula Óssea/patologia , Linhagem Celular , Quimiotaxia , Fibronectinas/fisiologia , Proteínas de Fusão bcr-abl/química , Humanos , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Mutagênese , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Prolina , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Deleção de Sequência , Transfecção , Células Tumorais Cultivadas , Domínios de Homologia de src
6.
Stem Cells ; 17(4): 210-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10437984

RESUMO

We used a primate model of autologous peripheral blood progenitor cell (PBPC) transplantation to study the effect of in vitro expansion on committed progenitor cell engraftment and marrow recovery after transplantation. Four groups of baboons were transplanted with enriched autologous CD34+ PBPC collected by apheresis after five days of G-CSF administration (100 microg/kg/day). Groups I and III were transplanted with cryopreserved CD34+ PBPC and Groups II and IV were transplanted with CD34+ PBPC that had been cultured for 10 days in Amgen-defined (serum free) medium and stimulated with G-CSF, megakaryocyte growth and development factor (MGDF), and stem cell factor each at 100 etag/ml. Group III and IV animals were administered G-CSF (100 microg/kg/day) and MGDF (25 microg/kg/day) after transplant, while animals in Groups I and II were not. For the cultured CD34+ PBPC from groups II and IV, the total cell numbers expanded 14.4 +/- 8.3 and 4.0 +/- 0.7-fold, respectively, and CFU-GM expanded 7.2 +/- 0.3 and 8.0 +/- 0.4-fold, respectively. All animals engrafted. If no growth factor support was given after transplant (Groups II and I), the recovery of WBC and platelet production after transplant was prolonged if cells had been cultured prior to transplant (Group II). Administration of post-transplant G-CSF and MGDF shortened the period of neutropenia (ANC < 500/microL) from 13 +/- 4 (Group I) to 10 +/- 4 (Group III) days for animals transplanted with non-expanded CD34+ PBPC. For animals transplanted with ex vivo-expanded CD34+ PBPC, post-transplant administration of G-CSF and MGDF shortened the duration of neutropenia from 14 +/- 2 (Group II) to 3 +/- 4 (Group IV) days. Recovery of platelet production was slower in all animals transplanted with expanded CD34+ PBPC regardless of post-transplant growth factor administration. Progenitor cells generated in vitro can contribute to early engraftment and mitigate neutropenia when growth factor support is administered post-transplant. Thrombocytopenia was not decreased despite evidence of expansion of megakaryocytes in cultured CD34+ populations.


Assuntos
Antígenos CD34 , Fator Estimulador de Colônias de Granulócitos/imunologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Neutropenia/prevenção & controle , Fator de Células-Tronco/imunologia , Trombopoetina/imunologia , Animais , Fator Estimulador de Colônias de Granulócitos/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/imunologia , Neutropenia/imunologia , Papio , Proteínas Recombinantes de Fusão/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Fator de Células-Tronco/farmacologia , Trombopoetina/farmacologia , Fatores de Tempo
8.
Artigo em Inglês | MEDLINE | ID: mdl-11000995

RESUMO

Over the past decade, peripheral blood progenitor cells (PBPCs) have replaced bone marrow as the major source of haematopoietic support. This transition from marrow to PBPCs is due to the fact that, for many clinical studies, a significantly faster rate of engraftment, particularly for platelets, has been demonstrated for patients who receive PBPC support when compared to similarly treated patients who are transplanted with bone marrow. The leukapheresis procedure itself, quality of PBPC graft, methods of PBPC mobilization, purging, and ex vivo expansion will be discussed in detail.


Assuntos
Neoplasias da Mama/terapia , Transplante de Células-Tronco Hematopoéticas/métodos , Técnicas de Cultura de Células/métodos , Feminino , Sobrevivência de Enxerto/imunologia , Mobilização de Células-Tronco Hematopoéticas/métodos , Mobilização de Células-Tronco Hematopoéticas/normas , Transplante de Células-Tronco Hematopoéticas/normas , Humanos , Leucaférese/métodos , Leucaférese/normas
10.
J Hematother ; 7(5): 457-61, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9829320

RESUMO

Ex vivo expansion of cord blood (CB) cells requires CD34+ cell selection before expansion to obtain optimal numbers of progenitor cells. As a preliminary step to preclinical development of CB expansion, we have evaluated two clinical scale selection devices, the Isolex 300i (Baxter Healthcare, Immunotherapy Division) and the CliniMACS (Miltenyi Biotech Inc.), for CD34+ cell selection from frozen CB products. As expansion of CB results in differentiation of cells, there may be a depletion of stem cells. Therefore, only a fraction of the CB should be expanded while a portion of the CB is maintained unmanipulated for infusion. After thawing of 40% fractions of each CB product, we observed >95% viable cells, with a median total WBC count of 1.8 x 10(8) cells. Use of the Isolex 300i resulted in a median purity of 51% CD34+ cells (n=8) and a median recovery of 34% CD34+ cells. Use of the CliniMACS resulted in a median purity of 54% CD34+ cells (n=10) and a median recovery of 80% CD34+ cells. The absolute number of CD34+ cells recovered after selection varied with samples from 6.7 x 10(4) to 3.2 x 10(6) CD34+ cells. Expansion of CD34+ cells from both systems resulted in >20-fold expansion of CFU-GM, with a median of 44-fold expansion. These data demonstrate the feasibility of selecting small fractions of frozen CB products using clinical scale CD34+ cell selection devices.


Assuntos
Sangue Fetal , Mobilização de Células-Tronco Hematopoéticas/instrumentação , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas , Antígenos CD34 , Feminino , Humanos
11.
Bone Marrow Transplant ; 21(10): 975-81, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9632269

RESUMO

We have evaluated the durability of engraftment and the potential of remobilization in mice reconstituted with mobilized peripheral blood progenitor cells (PBPC). Female mice which had been reconstituted with cytokine-mobilized PBPC from male donors were serially transplanted into second, third, fourth and fifth lethally irradiated female recipients at intervals of 6-10 months. Male-derived hematopoiesis was determined in recipient mice at each serial transplantation. Male-positive CFCs were detected after 5 passages for 45 months, but declined from >95% at passage 1 to 74% at passage 2, 33% at passage 4, and 28% at passage 5. Long-term survival also declined from 97% at passage 2 to 53% at passage 4, and 27% at passage 5. The results demonstrated that mobilized PBPC were able to provide engraftment for more than 45 months, but the engraftment provided by mobilized PBPC decreased at each serial passage. In addition, mice reconstituted with mobilized PBPC (at 1 year post transplantation) were treated with the same cytokines as in the primary mobilization (remobilization). The remobilized PBPC were harvested and transplanted into lethally irradiated secondary recipients. Male-derived CFCs were evaluated at 20 months post transplantation. Mice transplanted with PBPC remobilized with rhG-CSF or rhG-CSF plus rrSCF-PEG showed 70% and 89% male-positive CFCs respectively, demonstrating that mice reconstituted with mobilized PBPC could be remobilized and that the remobilized PBPC were also capable of providing long-term hematopoietic reconstitution. Our studies demonstrated that mobilized PBPC have extensive proliferative or self-renewal capacity to provide durable engraftment and that marrow repopulating cells in PBPC reconstituted mice can be remobilized, suggesting that patients who relapse after PBPC transplantation may be remobilized for a second transplantation to support additional chemotherapy.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Animais , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas Recombinantes , Fator de Células-Tronco/farmacologia
12.
Blood ; 90(8): 2939-51, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9376574

RESUMO

The safety and optimal dose and schedule of stem cell factor (SCF) administered in combination with filgrastim for the mobilization of peripheral blood progenitor cells (PBPCs) was determined in 215 patients with high-risk breast cancer. Patients received either filgrastim alone (10 microg/kg/d for 7 days) or the combination of 10 microg/kg/d filgrastim and 5 to 30 microg/kg/d SCF for either 7, 10, or 13 days. SCF patients were premedicated with antiallergy prophylaxis. Leukapheresis was performed on the final 3 days of cytokine therapy and, after high-dose chemotherapy and infusion of PBPCs, patients received 10 microg/kg/d filgrastim until absolute neutrophil count recovery. The median number of CD34+ cells collected was greater for patients receiving the combination of filgrastim and SCF, at doses greater than 10 microg/kg/d, than for those receiving filgrastim alone (7.7 v 3.2 x 10(6)/kg, P < .05). There were significantly (P < .05) more CD34+ cells harvested for the 20 microg/kg/d SCF (median, 7.9 x 10(6)/kg) and 25 microg/kg/d SCF (median, 13.6 x 10(6)/kg) 7-day combination groups than for the filgrastim alone patients (median, 3.2 x 10(6)/kg). The duration of administration of SCF and filgrastim (7, 10, or 13 days) did not significantly affect CD34+ cell yield. Treatment groups mobilized with filgrastim alone or with the cytokine combination had similar hematopoietic engraftment and overall survival after PBPC infusion. In conclusion, the results of this study indicate that SCF therapy enhances CD34+ cell yield and is associated with manageable levels of toxicity when combined with filgrastim for PBPC mobilization. The combination of 20 microg/kg/d SCF and 10 microg/kg/d filgrastim with daily apheresis beginning on day 5 was selected as the optimal dose and schedule for the mobilization of PBPCs.


Assuntos
Neoplasias da Mama Masculina/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator de Células-Tronco/uso terapêutico , Adolescente , Adulto , Idoso , Antígenos CD34/análise , Coleta de Amostras Sanguíneas , Neoplasias da Mama/sangue , Neoplasias da Mama Masculina/sangue , Esquema de Medicação , Quimioterapia Combinada , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Leucaférese , Masculino , Pessoa de Meia-Idade , Transfusão de Plaquetas , Proteínas Recombinantes , Fator de Células-Tronco/administração & dosagem
13.
Blood ; 90(1): 76-84, 1997 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9207441

RESUMO

The effect of chronic expression of flt3 ligand (FL) on in vivo hematopoiesis was studied. Retroviral vector-mediated gene transfer was used in a mouse model of bone marrow transplantation to enforce expression of mouse FL cDNA in hematopoietic tissues. As early as 2 weeks posttransplantation, peripheral blood white blood cell counts in FL-overexpressing recipients were significantly elevated compared with controls. With the exception of eosinophils, all nucleated cell lineages studied were similarly affected in these animals. Experimental animals also exhibited severe anemia and progressive loss of marrow-derived erythropoiesis. All of the FL-overexpressing animals, but none of the controls, died between 10 and 13 weeks posttransplantation. Upon histological examination, severe splenomegaly was noted, with progressive fibrosis and infiltration by abnormal lymphoreticular cells. Abnormal cell infiltration also occurred in other organ systems, including bone marrow and liver. In situ immunocytochemistry on liver sections showed that the cellular infiltrate was CD3+/NLDC145+/CD11c+, but B220- and F4/80-, suggestive of a mixed infiltrate of dendritic cells and activated T lymphocytes. Infiltration of splenic blood vessel perivascular spaces resulted in vascular compression and eventual occlusion, leading to splenic necrosis consistent with infarction. These results show that FL can affect both myeloid and lymphoid cell lineages in vivo and further demonstrate the potential toxicity of in vivo treatment with FL.


Assuntos
Regulação da Expressão Gênica , Leucócitos/patologia , Proteínas de Membrana/genética , Baço/patologia , Animais , Contagem de Células Sanguíneas , Diferenciação Celular/genética , Linhagem da Célula , Movimento Celular/genética , Fibrose/genética , Fibrose/patologia , Expressão Gênica , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Camundongos Endogâmicos
15.
J Hematother ; 6(2): 145-50, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9131444

RESUMO

Allogeneic umbilical cord blood (UCB) cells have recently been used for transplantation following high-dose chemotherapy. However, the numbers of total cells, including progenitor cells, harvested are low compared with bone marrow or peripheral blood progenitor cell harvests. Therefore, we evaluated the potential of UCB cells for their ability to expand granulocyte-macrophage colony-forming cells (GM-CFC) and burst-forming unit-erythroid (BFU-E) cells over 10 days. We used an ammonium chloride lysing buffer to eliminate the majority of contaminating red blood cells. An average recovery of 61% of the starting number of white blood cells was obtained, while retaining 100% of the CD34+ cells. Ex vivo expansion cultures were established in Teflon cell culture bags (American Fluoroseal Corp, Columbia, MD) in 25 ml defined medium (Amgen Inc, Thousand Oaks, CA) containing 100 ng/ml each of stem cell factor (SCF), granulocyte colony-stimulating factor (G-CSF), and megakaryocyte growth and development factor. Either unselected UCB cells or CD34+ UCB cells, selected with Magnetic Activation Cell Sorting technology (Miltenyi Biotech GmbH, Bergisch Gladbach, Germany), were incubated for 10 days at 37 degrees C without refeeding. Unselected UCB cells seeded at 1 X 10(6)/ml produced an average expansion of 1.4-fold in total cells, 0.8-fold in GM-CFC, and 0.3-fold in BFU-E cells. By contrast, CD34+ selected UCB cells seeded at 1.0 X 10(4)/ml produced an average expansion of 113-fold in total cells, 72.6-fold in GM-CFC, and 49-fold in BFU-E cells. These data demonstrate that CD34+ cell selection is necessary for optimal expansion of both GM-CFC and BFU-E cells. The cell numbers thus obtained postexpansion may be sufficient for transplantation in adults.


Assuntos
Antígenos CD34/análise , Sangue Fetal/citologia , Separação Imunomagnética/métodos , Técnicas de Cultura de Células/métodos , Divisão Celular , Ensaio de Unidades Formadoras de Colônias , Células Precursoras Eritroides/citologia , Hemólise , Humanos
16.
Genomics ; 40(1): 86-93, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9070923

RESUMO

Cloning of interleukin-1 beta converting enzyme (ICE) and Caenorhabditis elegans death protein CED-3 revealed the structural and functional homology between these two proteases. It also suggested the involvement of ICE-like cysteine proteases in apoptosis. Several CED-3- and ICE-like cysteine proteases have been described, including Nedd2/Ich-1, CPP32 beta, Tx, ICErel3, and Mch2. We have previously described a mouse ortholog of cysteine protease CPP32 beta that shares strong homology with ICE and CED-3. Here, we describe the cloning of mouse and human Casp7, another member of this family of cysteine proteases. Mouse Casp7 encodes a putative 340-amino-acid polypeptide that contains all the known conserved residues required for protease function, including the QACRG sequence, aspartic acid residues for internal cleavage sites, and the residues required for substrate binding. Three RNA variants of human Casp7 were also cloned. Amino acid sequence analysis indicated that Casp7 shared high homology with CPP32 beta/Casp3 and Mch2/Casp6. Northern blot analysis demonstrated that a 2.6-kb Casp7 mRNA was expressed in various tissues except brain. Mouse interspecific backcross mapping allowed localization of Casp7 to the distal region of mouse chromosome 19, linked to Mxi1, Adra2a, and Aop1.


Assuntos
Caspases , Mapeamento Cromossômico , Cisteína Endopeptidases/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Proteínas de Caenorhabditis elegans , Caspase 1 , Caspase 3 , Caspase 7 , Clonagem Molecular , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , RNA Mensageiro , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
17.
Clin Cancer Res ; 3(9): 1571-8, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9815845

RESUMO

The quantity of hematopoietic progenitors in an apheresis collection is defined by the number of CD34(+) cells or granulocyte macrophage colony-forming units present. These parameters are believed to give roughly equivalent information on graft quality. We here report that the in vitro proliferative potential of r-metHuSCF (stem cell factor) plus filgrastim (granulocyte colony-stimulating factor; r-metHuG-CSF) mobilized peripheral blood (PB) CD34(+) cells obtained from previously heavily treated non-Hodgkin's lymphoma patients inversely correlates with extent of prior therapy. CD34(+) cells were enriched using the CellPro Ceprate system and placed in liquid culture for 4 weeks in the presence of either r-metHuSCF, IL-3, IL-6, filgrastim (S36G), or S36G plus erythropoietin (S36GE) with a weekly exchange of media and cytokines with reestablishment of culture at the starting cell concentration (Delta assay) and enumeration of progenitors. Starting with 4 x 10(4) CD34(+) cells from apheresis samples from patients who had received <10 cycles of prior chemotherapy, progenitors were detectable in culture at 4 weeks 81% of the time as compared to 14% with CD34(+) cells from patients who had received >10 cycles and 5% for >10 cycles plus radiotherapy. The total number of progenitors generated over the duration of culture (area under the curve) was calculated using the trapezoidal rule as a novel measure of the proliferative potential of the enriched PB CD34(+) cell population. The median area under the curve of CD34(+) cells from patients receiving <10 cycles of prior chemotherapy was 7.4 and 5.7 (x10(5)) using S36G or S36GE, respectively, 1.8 and 1.9 if the patients received >10 cycles of prior chemotherapy, and 1.4 and 1.2 if the patients received >10 cycles of prior chemotherapy plus radiotherapy (P < 0.001). These data show that prior therapy impacts on the quality of PB CD34(+) cells as measured by their ability to generate committed progenitors over a number of weeks in liquid culture.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Linfoma não Hodgkin/patologia , Fator de Células-Tronco/análogos & derivados , Antígenos CD34/análise , Área Sob a Curva , Divisão Celular , Ensaio de Unidades Formadoras de Colônias , Filgrastim , Transplante de Células-Tronco Hematopoéticas , Humanos , Interleucina-3/farmacologia , Interleucina-6/farmacologia , Leucaférese , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/terapia , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
18.
Stem Cells ; 14(6): 661-77, 1996 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8948024

RESUMO

Megakaryocyte growth and development factor (MGDF) is a ligand for c-mpl and a member of the hematopoietic growth factor superfamily. Recombinant murine MGDF specifically stimulates thrombopoiesis in mice. Recombinant human (rHu) MGDF stimulates megakaryocytic differentiation of baboon CD34+ marrow cells in vitro. Therefore, we determined the in vivo biological effects of rHuMGDF administered to normal baboons in the absence and presence of myelosuppression with 5-fluorouracil (5-FU). rHuMGDF was administered to normal baboons as a single s.c. injection at doses of 1, 10, 25 and 50 micrograms/kg/day for 10 days and, as a control, heat-inactivated MGDF was administered at a dose of 10 micrograms/kg/day. Platelet counts were markedly increased in all animals administered native rHuMGDF but not in animals given heat-inactivated rHuMGDF. Platelet counts began to increase between three and six days after starting rHuMGDF administration and the maximum average increases were 1.7-, 3.4-, 5.1- and 4.0-fold above baseline in animals administered 1, 10, 25 and 50 micrograms/kg/day, respectively. Maximum platelet counts were reached between 7 and 10 days after starting rHuMGDF and maintained for four days after the last dose. Thereafter, platelet counts decreased, reaching stable pretreatment values between 11 and 14 days after the last dose of rHuMGDF. No changes in red cell mass, peripheral blood white blood cell counts or differentials were observed during rHuMGDF treatment. For animals administered 10, 25 and 50 micrograms/kg/day of rHuMGDF, megakaryocytes increased more than threefold in marrow, were markedly enlarged, and had increased numbers of lobes. Overall marrow cellularity remained unchanged, as did red cell and white cell morphology. No marrow fibrosis was detected. Progenitor cells were not increased in marrow but did increase modestly in the peripheral blood, associated with increased numbers of CD34+ cells in the circulation. Following a single dose of 5-FU (120 mg/kg) animals were given either saline or pegylated (PEG) rHuMGDF (25 micrograms/kg/day) for 14 days. Platelet counts recovered to baseline by 13.8 +/- 1.8 days for PEG-rHuMGDF-treated baboons compared with 16.8 +/- 0.6 days for saline treated controls. Marrow biopsies revealed more rapid recovery of overall marrow cellularity and megakaryocytes in PEG-rHuMGDF-treated animals compared with controls. Thus, rHuMGDF specifically stimulates thrombopoiesis in normal and myelosuppressed baboons. rHuMGDF may be useful for stimulating thrombopoiesis in humans in clinical settings after myelosuppression.


Assuntos
Hematopoese/efeitos dos fármacos , Megacariócitos/citologia , Proteínas Recombinantes/farmacologia , Trombopoetina/farmacologia , Animais , Antígenos CD34/análise , Diferenciação Celular/efeitos dos fármacos , Fluoruracila/farmacologia , Humanos , Megacariócitos/efeitos dos fármacos , Camundongos , Papio , Contagem de Plaquetas/efeitos dos fármacos
19.
Oncogene ; 13(4): 749-55, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8761296

RESUMO

Interleukin-1 beta converting enzyme (ICE) defines a new class of mammalian cysteine protease that shares strong homology with the Caenorhabditis elegans death gene ced-3. Both ICE and CED-3, when introduced into cultured cells, induce apoptosis, indicating that this type of cysteine protease may play an important role in the process of programmed cell death. Here, we report the cloning of a mouse and rat gene encoding a novel cysteine protease. The putative proteins encoded by these cDNAs contain the conserved sequence (QACRG) necessary for covalent linkage to the substrate as well as the three amino acids responsible for substrate binding and catalysis in ICE. Amino acid sequence analysis indicates that this rodent cysteine protease is the homolog of human CPP32 beta. Mouse CPP32 beta mRNA is highly expressed in spleen, and to a lesser degree in brain, lung, liver, and kidney. The mouse CPP32 beta genomic locus spans a region of approximately 20 kb, including seven exons and six introns. Mouse interspecific backcross mapping allowed localization of CPP32 beta to the central region of mouse chromosome 8, linked to Scvr, Lpl, Jund1 and Mlr.


Assuntos
Caspases , Cisteína Endopeptidases/genética , Proteínas de Helminto/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Proteínas de Caenorhabditis elegans , Caspase 1 , Caspase 3 , Mapeamento Cromossômico , DNA Complementar , Éxons , Feminino , Humanos , Íntrons , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Ratos , Homologia de Sequência de Aminoácidos
20.
Blood ; 88(4): 1509-14, 1996 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8695873

RESUMO

Megakaryocyte growth and development factor (MGDF) is a recently characterized ligand for the cell surface receptor mpl. We have evaluated the effects of polyethylene glycollated recombinant human MGDF (PEG-rHuMGDF) on recovery of hematopoietic cells in mice following bone marrow transplantation (BMT) to support lethal irradiation. Mice treated with PEG-rHuMGDF (50 micrograms/kg/d) had accelerated recovery of platelet numbers compared with BMT mice treated with carrier or recombinant human granulocyte colony-stimulating factor (rHuG-CSF, 72 or 200 micrograms/kg/d). In contrast, PEG-rHuMGDF had no effect on white blood cell (WBC) or red blood cell (RBC) recovery. As previously reported, animals treated with rHuG-CSF had an enhanced recovery of WBC but not platelet or RBC levels. Interestingly, BMT receipient mice treated with the combination of PEG-rHuMGDF and rHuG-CSF showed simultaneous enhanced recovery of both leukocytes and platelets. PEGylated rHuMGDF was found to be considerably more potent than non-PEGylated rHuMGDF in this setting. PEG-rHuMGDF is an effective growth factor for enhancing platelet recovery in mice following BMT either alone or in combination with rHuG-CSF. It will be of interest to evaluate in a clinical setting the ratios of PEG-rHuMGDF and rHuG-CSF for simultaneous administration of these factors and accelerated recovery of both leukocytes and platelets.


Assuntos
Plaquetas/citologia , Transplante de Medula Óssea/patologia , Fatores Estimuladores de Colônias/farmacologia , Hematopoese/efeitos dos fármacos , Megacariócitos/citologia , Proteínas de Neoplasias , Polietilenoglicóis , Proteínas/farmacologia , Receptores de Citocinas , Trombopoetina/farmacologia , Animais , Proteínas Ligadas por GPI , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Leucócitos/citologia , Glicoproteínas de Membrana , Mesotelina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Proteínas Proto-Oncogênicas/fisiologia , Quimera por Radiação , Receptores de Trombopoetina , Proteínas Recombinantes , Trombopoetina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA