Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Bone Marrow Transplant ; 50(9): 1227-34, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26052909

RESUMO

Allogeneic hematopoietic stem cell transplantation (HSCT) is one of curative treatment options for patients with hematologic malignancies. Although GVHD mediated by the donor's T lymphocytes remains the most challenging toxicity of allo-HSCT, graft-versus-leukemia (GVL) effect targeting leukemic cells, has an important role in affecting the overall outcome of patients with AML. Here we comprehensively characterized the TCR repertoire in patients who underwent matched donor or haplo-cord HSCT using next-generation sequencing approach. Our study defines the functional kinetics of each TCRA and TCRB clone, and changes in T-cell diversity (with identification of CDR3 sequences) and the extent of clonal expansion of certain T-cells. Using this approach, our study demonstrates that higher percentage of cord-blood cells at 30 days after transplant was correlated with higher diversity of TCR repertoire, implicating the role of cord-chimerism in enhancing immune recovery. Importantly, we found that GVHD and relapse, exclusive of each other, were correlated with lower TCR repertoire diversity and expansion of certain T-cell clones. Our results highlight novel insights into the balance between GVHD and GVL effect, suggesting that higher diversity early after transplant possibly implies lower risks of both GVHD and relapse following the HSCT transplantation.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical , Transplante de Células-Tronco Hematopoéticas , Leucemia Mieloide Aguda , Síndromes Mielodisplásicas , Receptores de Antígenos de Linfócitos T alfa-beta , Linfócitos T/imunologia , Adulto , Idoso , Aloenxertos , Regiões Determinantes de Complementaridade/genética , Regiões Determinantes de Complementaridade/imunologia , Feminino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/imunologia , Leucemia Mieloide Aguda/terapia , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Síndromes Mielodisplásicas/imunologia , Síndromes Mielodisplásicas/terapia , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia
2.
Oncogene ; 26(47): 6803-15, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17934487

RESUMO

Red blood cells and megakaryocytes arise from a common precursor, the megakaryocyte-erythroid progenitor and share many regulators including the transcription factors GATA-1 and GFI-1B and signaling molecules such as JAK2 and STAT5. These lineages also share the distinction of being associated with rare, but aggressive malignancies that have very poor prognoses. In this review, we will briefly summarize features of normal development of red blood cells and megakaryocytes and also highlight events that lead to their leukemic transformation. It is clear that much more work needs to be done to improve our understanding of the unique biology of these leukemias and to pave the way for novel targeted therapeutics.


Assuntos
Transformação Celular Neoplásica/patologia , Células Eritroides/citologia , Células Eritroides/patologia , Megacariócitos/citologia , Megacariócitos/patologia , Animais , Transformação Celular Neoplásica/genética , Humanos , Leucemia/genética , Leucemia/patologia
3.
J Clin Oncol ; 23(24): 5728-38, 2005 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-16009946

RESUMO

PURPOSE: This prospective phase II study evaluated toxicity, relapse rate, progression-free survival, and overall survival after allogeneic transplantation and conditioning with fludarabine, melphalan, and alemtuzumab in patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). PATIENTS AND METHODS: Fifty-two consecutive adults with AML and MDS were enrolled onto the study. Median age was 52 years (range, 17 to 71 years) and the majority of patients had high-risk disease, comorbidities, and/or modest reduction in performance status. Fifty-six percent of patients had unrelated or mismatched related donors. RESULTS: After a median follow-up of 18 months (range, 2 to 34 months), 1-year survival was 48% (95% CI, 34% to 61%), progression-free survival was 38% (95% CI, 25% to 52%), relapse rate was 27% (95% CI, 15% to 40%), and treatment-related mortality was 33% (95% CI, 20% to 46%). The cumulative probability of extensive chronic graft-versus-host disease (GVHD) was only 18% (95% CI, 8% to 40%); extensive chronic GVHD was only observed in recipients of unrelated donor transplants. Performance score and disease status were the major predictors of outcome. High-risk disease (ie, active AML or MDS with > 5% blasts) or even modest decreases in performance status were associated with poor outcomes. Patients with standard-risk leukemia (first or second complete remission) or MDS (< 5% blasts) had excellent outcomes despite unfavorable disease characteristics. CONCLUSION: Fludarabine and melphalan combined with in vivo alemtuzumab is a promising transplantation regimen for patients with AML or MDS and low tumor burden. For patients with active disease, this regimen provides at best modest palliation. Despite a low incidence of GVHD, transplantation is still associated with considerable nonrelapse mortality in patients with decreased performance status.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia Mieloide/terapia , Síndromes Mielodisplásicas/terapia , Transplante de Células-Tronco , Condicionamento Pré-Transplante/métodos , Doença Aguda , Adolescente , Adulto , Idoso , Alemtuzumab , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Anticorpos Antineoplásicos/administração & dosagem , Intervalo Livre de Doença , Feminino , Doença Enxerto-Hospedeiro/prevenção & controle , Humanos , Masculino , Melfalan/administração & dosagem , Pessoa de Meia-Idade , Modelos de Riscos Proporcionais , Estudos Prospectivos , Indução de Remissão , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
4.
Bone Marrow Transplant ; 32(5): 471-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12942092

RESUMO

A total of 31 consecutive patients with hematologic malignancies who were considered poor candidates for TBI underwent allogeneic stem cell transplantation after conditioning with fludarabine and melphalan. A total of 25 matched sibling recipients received fludarabine 25 mg/m(2) x 5 days and melphalan 70 mg/m(2) x 2 days. For unrelated and haploidentical donor recipients, fludarabine was increased to 30 mg/m(2) and ATG 30 mg/kg x 4 days was added. Graft-versus-host disease prophylaxis consisted of tacrolimus and mini methotrexate. All patients engrafted. Regimen-related toxicity was considerable and included mainly renal, hepatic and mucosal toxicity. There were seven regimen-related-deaths including two VOD, two pulmonary, one renal, one cardiac and one mucosal toxicity. One case of fatal pulmonary toxicity death could be attributed to pre-existing pulmonary damage. Progression-free survival at 12 months was 44% (90% CI: 30-58%) for recipients of HLA-identical sibling transplants and 33% (90% CI: 21-45%) for all patients. In conclusion, the fludarabine-melphalan regimen leads to consistent engraftment. The regimen-related toxicity is considerable and cannot be explained solely by patient selection. Cardiac toxicity is emerging as a unique toxicity of this regimen. Despite toxicity, fludarabine-melphalan has considerable activity and leads to durable remission in a proportion of patients.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Neoplasias Hematológicas/terapia , Condicionamento Pré-Transplante/efeitos adversos , Vidarabina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Intervalo Livre de Doença , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/prevenção & controle , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/mortalidade , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/mortalidade , Humanos , Melfalan/administração & dosagem , Melfalan/toxicidade , Irmãos , Condicionamento Pré-Transplante/métodos , Condicionamento Pré-Transplante/mortalidade , Transplante Homólogo , Vidarabina/administração & dosagem , Vidarabina/toxicidade
5.
Bone Marrow Transplant ; 32(1): 9-13, 2003 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12815472

RESUMO

Fludarabine, thiotepa and total body irradiation (TBI) has been used as conditioning in haplo-identical transplantation. We studied this conditioning regimen in adults undergoing matched sibling transplantation and alternative donor transplantation. A total of 30 consecutive patients underwent matched related, haplo-identical related or matched unrelated donor transplantation with fludarabine, thiotepa and TBI conditioning. All but four had advanced hematologic malignancies. For haplo-identical transplant, ATG was added to the regimen. All patients received peripheral blood stem cells; these were T-cell depleted for 2-antigen or 3-antigen mismatched related transplantation. Additional graft-versus-host disease prophylaxis consisted of tacrolimus and mini-methotrexate. One recipient of haplo-identical transplant failed to engraft; all other evaluable patients had prompt engraftment. Four patients died of regimen-related toxicity. In all, 14 additional patients died of regimen-related causes including four from failure to thrive with persistent thrombocytopenia and four from delayed pulmonary toxicity. Six patients relapsed. Progression-free survival at 12 months was 47% (90% CI: 25-69%) for recipients of HLA-identical sibling transplants and 30% (90% CI: 14-46%) for all patients. Five of six long-term survivors have extensive chronic GVHD. As a result of the delayed complications and a relatively high recurrence rate, we abandoned this regimen.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco de Sangue Periférico/métodos , Condicionamento Pré-Transplante/métodos , Vidarabina/análogos & derivados , Irradiação Corporal Total/efeitos adversos , Adolescente , Adulto , Antineoplásicos Alquilantes/administração & dosagem , Criança , Terapia Combinada , Sobrevivência de Enxerto , Doença Enxerto-Hospedeiro/tratamento farmacológico , Doença Enxerto-Hospedeiro/imunologia , Haplótipos , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/mortalidade , Histocompatibilidade , Humanos , Pessoa de Meia-Idade , Transplante de Células-Tronco de Sangue Periférico/efeitos adversos , Transplante de Células-Tronco de Sangue Periférico/mortalidade , Análise de Sobrevida , Tiotepa/administração & dosagem , Tiotepa/toxicidade , Condicionamento Pré-Transplante/efeitos adversos , Condicionamento Pré-Transplante/mortalidade , Transplante Homólogo/imunologia , Falha de Tratamento , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/toxicidade , Irradiação Corporal Total/métodos
6.
Bone Marrow Transplant ; 31(9): 813-6, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12732890

RESUMO

To establish the incidence of CMV viremia after allogeneic blood stem cell transplantation, we studied 51 consecutive allogeneic peripheral blood stem cell (PBSC) transplant recipients. A total of 12 recipients were at moderate risk for CMV disease and 39 were at high risk. Conditioning regimens varied, but GvHD prophylaxis consisted of tacrolimus and mini-methotrexate in all patients. All patients received prophylactic ganciclovir from admission until day -2 and prophylactic acyclovir from day -1 until day 180 after transplantation. CMV viremia was treated with ganciclovir. Using a PCR-based technique, the cumulative incidence of CMV viremia was 31+/-14% by day 100 and 35+/-14% by day 150. Donor type, CMV risk group, underlying disorder, conditioning regimen, GvHD, and steroid use were not associated with the risk for CMV viremia. No cases of CMV disease occurred. We hypothesize that the low rate of CMV viremia and the absence of CMV disease in this cohort of PBSCT transplant recipients, which contrasts with other reports, may be related to the prophylactic use of high-dose acyclovir and possibly to pretransplant use of ganciclovir.


Assuntos
Aciclovir/uso terapêutico , Infecções por Citomegalovirus/prevenção & controle , Ganciclovir/uso terapêutico , Transplante de Células-Tronco de Sangue Periférico/efeitos adversos , Pré-Medicação , Aciclovir/administração & dosagem , Adolescente , Adulto , Antivirais/administração & dosagem , Antivirais/uso terapêutico , Infecções por Citomegalovirus/tratamento farmacológico , Feminino , Ganciclovir/administração & dosagem , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , Transplante de Células-Tronco de Sangue Periférico/métodos , Reação em Cadeia da Polimerase , Fatores de Risco , Transplante Homólogo , Viremia/diagnóstico , Viremia/tratamento farmacológico , Viremia/prevenção & controle
7.
Bone Marrow Transplant ; 28(6): 557-62, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11607768

RESUMO

Severe regimen-related toxicity often complicates second transplant procedures performed in patients with hematological malignancies that have relapsed after an initial hematopoietic stem cell (HSC) transplant. Therefore, we studied the safety and efficacy of a reduced-intensity fludarabine and melphalan based conditioning regimen in 11 patients who had relapsed following an autologous (n = 7) or allogeneic (n = 4) HSC transplant. All patients received allogeneic peripheral blood HSC from either an HLA-identical (n = 7) or an HLA-mismatched (n = 4) relative. Diagnoses included AML (n = 9), ALL (n = 1), or Hodgkin's disease (n = 1). Only one patient was in complete remission at the time of second transplant. The median interval between first transplant and relapse was 163 days (range 58-1885). Recipients of HLA-mismatched transplants received antithymocyte globulin in addition to fludarabine and melphalan as part of the conditioning regimen. All 11 patients received acute GVHD prophylaxis consisting of tacrolimus and methotrexate. Ten of 11 patients achieved hematopoietic engraftment with a median time to absolute neutrophil count >0.5 x 10(9)/l and to platelet count of >20 x 10(9)/l of 14 and 19 days, respectively. All engrafting patients achieved 100% donor chimerism on initial analysis, except for one with persistent leukemia at day +19. Two patients experienced grade 3 regimen-related toxicity, manifesting as acute renal failure. Acute GVHD grades 2-4 occurred in two recipients and chronic GVHD in four. The 100-day mortality from all causes was 36%. Ten of 11 patients (91%) died a median of 140 days (range 9-996) after the second transplant. The causes of death included relapse (n = 5), sepsis (n = 4), and idiopathic pneumonia syndrome (n = 1). One patient with AML survives in remission at 880 days post-transplant. We conclude that fludarabine- and melphalan-based conditioning promotes full donor chimerism, even following HLA-mismatched transplants. However, the regimen may be more beneficial when applied to patients undergoing allogeneic HSC transplantation earlier in their disease course.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Condicionamento Pré-Transplante/métodos , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Causas de Morte , Feminino , Sobrevivência de Enxerto/imunologia , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/mortalidade , Transplante de Células-Tronco Hematopoéticas/mortalidade , Humanos , Masculino , Melfalan/administração & dosagem , Pessoa de Meia-Idade , Recidiva , Terapia de Salvação , Taxa de Sobrevida , Quimeras de Transplante , Condicionamento Pré-Transplante/mortalidade , Condicionamento Pré-Transplante/normas , Resultado do Tratamento , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
8.
Blood ; 96(6): 2084-92, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10979952

RESUMO

Erythropoietin (EPO) allows erythroid precursors to proliferate while protecting them from apoptosis. Treatment of the EPO-dependent HCD57 murine cell line with 70 micromol/L orthovanadate, a tyrosine phosphatase inhibitor, resulted in both increased tyrosine protein phosphorylation and prevention of apoptosis in the absence of EPO without promoting proliferation. Orthovanadate also delayed apoptosis in primary human erythroid progenitors. Thus, we investigated what survival signals were activated by orthovanadate treatment. Expression of Bcl-X(L) and BAD phosphorylation are critical for the survival of erythroid cells, and orthovanadate in the absence of EPO both maintained expression levels of antiapoptotic Bcl-X(L) and induced BAD phosphorylation at serine 112. Orthovanadate activated JAK2, STAT1, STAT5, the phosphatidylinositol-3 kinase (PI-3 kinase) pathway, and other signals such as JNK and p38 without activating the EPO receptor, JAK1, Tyk2, Vav, STAT3, and SHC. Neither JNK nor p38 appeared to have a central role in either apoptosis or survival induced by orthovanadate. Treatment with cells with LY294002, an inhibitor of PI-3 kinase activity, triggered apoptosis in orthovanadate-treated cells, suggesting a critical role of PI-3 kinase in orthovanadate-stimulated survival. Mitogen-activated protein kinase (MAPK) was poorly activated by orthovanadate, and inhibition of MAPK with PD98059 blocked proliferation without inducing apoptosis. Thus, orthovanadate likely acts to greatly increase JAK/STAT and PI-3 kinase basal activity in untreated cells by blocking tyrosine protein phosphatase activity. Activated JAK2/STAT5 then likely acts upstream of Bcl-X(L) expression and PI-3 kinase likely promotes BAD phosphorylation to protect from apoptosis. In contrast, MAPK/ERK activity correlates with only EPO-dependent proliferation but is not required for survival of HCD57 cells.


Assuntos
Apoptose/fisiologia , Eritroblastos/patologia , Eritroblastos/fisiologia , Fosfoproteínas Fosfatases/fisiologia , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Eritropoetina , Humanos , Camundongos , Fosfoproteínas Fosfatases/antagonistas & inibidores , Vanadatos/farmacologia
9.
Biochem Biophys Res Commun ; 275(1): 16-9, 2000 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-10944433

RESUMO

Erythropoietin (Epo), stem cell factor (SCF), and insulin-like growth factor-1 (IGF-1) are key regulators of erythroid cell proliferation and differentiation. To understand the mechanisms of generation of signals by each of these growth factors, we determined the activation of the PI3-kinase/Akt pathway during proliferation and differentiation of primary human erythroid progenitors. Our results demonstrate that PKB/Akt is activated by Epo and SCF, but not by IGF-1 in human primary erythroid progenitors. In addition, Epo treatment of erythroid progenitors induces phosphorylation of a member of the Forkhead family (FH) of transcription factors FKHRL1, downstream of activation of the Akt kinase. Such Epo-dependent activation of FKHRL1 apparently regulates the generation of Epo-dependent antiapoptotic signals as evidenced by the induction of apoptosis of erythroid progenitors during treatment of cells with the PI3-kinase (PI3K) inhibitor LY294002. Thus, the PI3K/Akt/FKHRL1 pathway is essential for inhibition of apoptosis in response to Epo and SCF, while the IGF-1 receptor utilizes a different pathway.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Caspase 3 , Caspases/metabolismo , Diferenciação Celular , Células Cultivadas , Cromonas/farmacologia , Ativação Enzimática/efeitos dos fármacos , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/enzimologia , Células Precursoras Eritroides/metabolismo , Proteína Forkhead Box O1 , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Fator de Células-Tronco/farmacologia
10.
Blood ; 95(12): 3742-9, 2000 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-10845905

RESUMO

Based on the hypothesis that interferon gamma (IFN-gamma) may have stimulating effects on survival of hematopoietic progenitor cells, we examined the effect of IFN-gamma on apoptosis of mature erythroid colony-forming cells (ECFCs) derived from human peripheral blood obtained from normal, healthy volunteers. When the cells were cultured in the presence of IFN-gamma, even without erythropoietin (EPO), the viability of the cells was maintained for at least 36 hours. When apoptosis of ECFCs was assessed by flow cytometric analysis', using annexin V, IFN-gamma reduced the extent of apoptosis of the cells, as well as EPO. DNA fragmentation of ECFCs was also reduced by IFN-gamma. In cells cultured with IFN-gamma alone, expression of Bcl-x was detected but the level of expression decreased gradually during incubation for 36 hours, and the expression level was lower than incubation with EPO. Fas expression and activation of downstream caspases were assessed by flow cytometric analysis or fluorometric protease assay. IFN-gamma induced Fas expression of the cells without the activation of caspase8 or caspase3 during 16 hours of incubation, while deprivation of EPO induced expression of Fas and the activation of both caspase8 and caspase3. We propose that IFN-gamma produces a stimulating signal for the survival of mature erythroid progenitor cells by reducing apoptosis through a mechanism other than modulating Fas and one related to the expression of Bcl-x. (Blood. 2000;95:3742-3749)


Assuntos
Apoptose/fisiologia , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Interferon gama/farmacologia , Anexina A5/análise , Apoptose/efeitos dos fármacos , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Meios de Cultura Livres de Soro , Fragmentação do DNA , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Interleucina-3/farmacologia , Cinética , Proteínas Proto-Oncogênicas c-bcl-2/análise , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/farmacologia
11.
J Biol Chem ; 274(35): 24469-74, 1999 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-10455108

RESUMO

Several signaling cascades are activated during engagement of the erythropoietin receptor to mediate the biological effects of erythropoietin. The members of the insulin receptor substrate (IRS) family of proteins play a central role in signaling for various growth factor receptors and cytokines by acting as docking proteins for the SH2 domains of signaling elements, linking cytokine receptors to diverse downstream pathways. In the present study we provide evidence that the recently cloned IRS-related proteins, Gab1 and Gab2, of the Gab family of proteins, are rapidly phosphorylated on tyrosine during erythropoietin treatment of erythropoietin-responsive cells and provide docking sites for the engagement of the SHP2 phosphatase and the p85 subunit of the phosphatidylinositol 3'-kinase. Furthermore, our data show that Gab1 is the primary IRS-related protein activated by erythropoietin in primary erythroid progenitor cells. In studies to identify the erythropoietin receptor domains required for activation of Gab proteins, we found that tyrosines 425 and 367 in the cytoplasmic domain of the erythropoietin receptor are required for the phosphorylation of Gab2. Taken together, our data demonstrate that Gab proteins are engaged in erythropoietin signaling to mediate downstream activation of the SHP2 and phosphatidylinositol 3'-kinase pathways and possibly participate in the generation of the erythropoietin-induced mitogenic responses.


Assuntos
Eritropoetina/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Linhagem Celular , Células-Tronco Hematopoéticas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Fosfotirosina/análise , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Receptores da Eritropoetina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Exp Hematol ; 27(8): 1315-21, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10428508

RESUMO

Interferons are potent regulators of normal and malignant hematopoietic cell proliferation in vitro and in vivo, but the signaling mechanisms by which they exhibit their growth inhibitory effects are unknown. We have recently shown that CrkL is engaged in Type I IFN signaling, as shown by its rapid tyrosine phosphorylation during engagement of the Type I IFN receptor. In the present study, we provide evidence that the related CrkII protein is also rapidly phosphorylated on tyrosine during treatment of U-266 and Daudi cells with IFNalpha or IFNbeta. We also show that both members of the Crk-family, CrkL and CrkII, are phosphorylated in an interferon-dependent manner in primary hematopoietic progenitors. Furthermore, inhibition of CrkL or CrkII protein expression by antisense oligonucleotides, reverses the inhibitory effects of IFNalpha or IFNgamma on the proliferation of normal bone marrow progenitor cells (colony forming units-granulocytic/monocytic [CFU-GM] and burst-forming units-erythroid [BFU-E]). Thus, both CrkL and CrkII are engaged in a signaling pathway (s) that mediates interferon-regulated inhibition of hematopoietic cell proliferation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Inibidores do Crescimento/farmacologia , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Interferon-alfa/farmacologia , Interferon beta/farmacologia , Interferon gama/farmacologia , Proteínas Nucleares/fisiologia , Proteínas Quinases/fisiologia , Proteínas Proto-Oncogênicas , Linfoma de Burkitt/patologia , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Células-Tronco Hematopoéticas/citologia , Humanos , Proteínas Nucleares/genética , Oligonucleotídeos Antissenso/farmacologia , Fosforilação/efeitos dos fármacos , Proteínas Quinases/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-crk , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos , Ensaio Tumoral de Célula-Tronco
13.
Exp Hematol ; 27(7): 1124-32, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10390187

RESUMO

The SHP-1 phosphatase associates with the receptors for erythropoietin, stem cell factor, and interleukin-3, and negatively regulates the mitogenic signals generated during engagement by their respective ligands. The erythroid progenitors of patients with polycythemia vera are hypersensitive to the mitogenic effects of these growth factors despite the fact that the numbers and binding affinities for their receptors are not increased. To determine whether post-receptor signaling defects may account for growth factor-hypersensitivity in polycythemia vera, we determined the expression of SHP-1 in highly purified erythroid progenitors from polycythemia vera patients. Our data demonstrate that in approximately 60% of the patients, expression of SHP-1 in the colony forming unit-erythroid population is diminished. The decreased expression of the protein may result from a transcriptional defect, as suggested by the diminished SHP-1 mRNA expression in the erythroid progenitors of these patients. Studies to determine the level of maturation of polycythemia vera and normal cells indicated that there was no difference between the two at early colony forming unit-erythroid stage of differentiation although polycythemia vera cells showed retarded differentiation kinetics at late colony forming unit-erythroid stage of differentiation. Furthermore, SHP-1 expression in normal colony forming unit-erythroid demonstrated downregulation of mRNA and protein levels during terminal differentiation, suggesting that its function is required for growth control during the early stages of erythropoiesis. These results indicate an important role for SHP-1 in the regulation of normal human erythroid progenitors and suggest that defective expression of the protein may contribute to the pathogenesis of polycythemia vera.


Assuntos
Policitemia Vera/enzimologia , Proteínas Tirosina Fosfatases/deficiência , Diferenciação Celular , Ensaio de Unidades Formadoras de Colônias , Indução Enzimática , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/enzimologia , Células Precursoras Eritroides/patologia , Eritropoese/efeitos dos fármacos , Eritropoese/fisiologia , Eritropoetina/farmacologia , Heme/biossíntese , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Fosforilação , Policitemia Vera/genética , Policitemia Vera/patologia , Processamento de Proteína Pós-Traducional , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/biossíntese , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/biossíntese , Receptores da Eritropoetina/metabolismo , Receptores de Interleucina-3/metabolismo , Transdução de Sinais , Transcrição Gênica
14.
Blood ; 93(11): 3757-73, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10339482

RESUMO

We found that erythropoietin (EPO) and stem cell factor (SCF) activated protein kinase B (PKB/Akt) in EPO-dependent HCD57 erythroid cells. To better understand signals controlling proliferation and viability, erythroid cells that resist apoptosis in the absence of EPO were subcloned and characterized (HCD57-SREI cells). Constitutive activations of PKB/Akt, STAT5a, and STAT5b were noted in these EPO-independent cells. PI3-kinase activity was an upstream activator of PKB/Akt because the PI3-kinase inhibitor LY294002 blocked both constitutive PKB/Akt and factor-dependent PKB/Akt activity. The LY294002 study showed that proliferation and viability of both HCD57-SREI and HCD57 cells correlated with the activity of PKB/Akt; however, PKB/Akt activity alone did not protect these cells from apoptosis. Treatment of HCD57 cells with SCF also activated PKB/Akt, but did not protect from apoptosis. This result suggested that PKB/PI3-kinase activity is necessary but not sufficient to promote viability and/or proliferation. Constitutive STAT5 activity, activated through an unknown pathway not including JAK2 or EPOR, may act in concert with the constitutive PI3-kinase/PKB/Akt pathway to protect the EPO-independent HCD57-SREI cells from apoptosis and promote limited proliferation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Eritrócitos/metabolismo , Eritropoetina/farmacologia , Proteínas do Leite , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular , Eritrócitos/patologia , Humanos , Proteínas Proto-Oncogênicas c-akt , Fator de Transcrição STAT5 , Proteínas Supressoras de Tumor
15.
Mol Cell Biol ; 18(7): 3699-707, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9632752

RESUMO

The transcription factor AP1 has been implicated in the induction of apoptosis in cells in response to stress factors and growth factor withdrawal. We report here that AP1 is necessary for the induction of apoptosis following hormone withdrawal in the erythropoietin (EPO)-dependent erythroid cell line HCD57. AP1 DNA binding activity increased upon withdrawal of HCD57 cells from EPO. A dominant negative AP1 mutant rendered these cells resistant to apoptosis induced by EPO withdrawal and blocked the downregulation of Bcl-XL. JunB is a major binding protein in the AP1 complex observed upon EPO withdrawal; JunB but not c-Jun was present in the AP1 complex 3 h after EPO withdrawal in HCD57 cells, with a concurrent increase in junB message and protein. Furthermore, analysis of AP1 DNA binding activity in an apoptosis-resistant subclone of HCD57 revealed a lack of induction in AP1 DNA binding activity and no change in junB mRNA levels upon EPO withdrawal. In addition, we determined that c-Jun and AP1 activities correlated with EPO-induced proliferation and/or protection from apoptosis. AP1 DNA binding activity increased over the first 3 h following EPO stimulation of HCD57 cells, and suppression of AP1 activity partially inhibited EPO-induced proliferation. c-Jun but not JunB was present in the AP1 complex 3 h after EPO addition. These results implicate AP1 in the regulation of proliferation and survival of erythroid cells and suggest that different AP1 factors may play distinct roles in both triggering apoptosis (JunB) and protecting erythroid cells from apoptosis (c-Jun).


Assuntos
Apoptose/fisiologia , Células Precursoras Eritroides/fisiologia , Fator de Transcrição AP-1/fisiologia , Divisão Celular , Linhagem Celular , DNA/metabolismo , Eritropoese/fisiologia , Eritropoetina/fisiologia , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-jun/genética , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo , Proteína bcl-X
16.
Blood ; 86(1): 381-9, 1995 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-7540887

RESUMO

High-dose therapy with autologous marrow or peripheral blood stem cell (PBSC) rescue has been extensively applied in the treatment of multiple myeloma (MM) patients during the past 10 years resulting in improved event-free and overall survival when compared with standard chemotherapy. However, relapses are common and cure is unlikely in the majority of patients. Because both bone marrow and PBSCs are contaminated with myeloma cells it is conceivable that relapse after autotransplantation originates at least in part from autografted tumor cells. In this study, mobilized PBSCs were examined for the presence of myeloma cells based on immunophenotyping and sensitive polymerase chain reaction (PCR)-based techniques. In addition, CD34+ Lin- Thy+ stem cells were purified from mobilized PBSC harvests of 10 MM patients by sequentially using counterflow elutriation centrifugation, treatment with phenylalanine methylester, and flow sorting, using 5-parameter gating (propidium iodide, forward scatter, side scatter, CD34+ v Lin- and CD34+ v Thy+). Virtually all mobilized unsorted PBSC preparations contained myeloma cells in sufficient quantities (range, < 0.01 to > 10%) potentially causing a disease relapse. Stem cell purification led to an overall enrichment by about 50-fold in all 10 patients; approximately 90% of the final cell population expressed CD34+ Lin- Thy+ with no evidence of myeloma cell contamination based on flow cytometric analysis of CD38bright cells (< 0.1%). Quantitative PCR amplification of patient-specific complementarity determining region III (CDRIII) DNA sequences showed depletion of clonal B cells by 2.7 to 7.3 logs, with the highest log reduction noted in the samples initially containing the most tumor cells. Our results show that purification of CD34+ Lin- Thy+ cells depletes myeloma cells to undetectable levels from up to 10% present in unsorted PBSCs, thus offering a tool to investigate whether MM relapse after autotransplantation can be reduced markedly.


Assuntos
Antígenos CD/análise , Remoção de Componentes Sanguíneos , Separação Celular/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas , Imunofenotipagem , Mieloma Múltiplo/patologia , Células-Tronco Neoplásicas , Antígenos Thy-1/análise , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Antígenos CD34 , Antígenos de Diferenciação/análise , Antígenos de Diferenciação de Linfócitos T/análise , Antígeno CD56 , Centrifugação , Sequência Consenso , Distribuição Contracorrente , Ciclofosfamida/farmacologia , Primers do DNA , Estudos de Viabilidade , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Células-Tronco Hematopoéticas/química , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Cadeias Pesadas de Imunoglobulinas/genética , Glicoproteínas de Membrana , N-Glicosil Hidrolases/análise , Células-Tronco Neoplásicas/química , Fenilalanina/análogos & derivados , Fenilalanina/farmacologia , Plasmócitos/química , Reação em Cadeia da Polimerase
17.
J Cell Physiol ; 160(3): 417-26, 1994 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-8077279

RESUMO

We have used highly purified human early erythroid progenitors to study changes in cytoskeletal proteins during their maturation and terminal differentiation. When erythroid progenitors at the burst-forming unit-erythroid (BFU-E) stage of development are grown in the presence of erythropoietin, the cells mature and terminally differentiate into reticulocytes during a 14-15-day culture period. We have shown by immunofluorescence that spectrin is present in day 3 BFU-E, at which time proteins band 3, ankyrin, and band 4.1 cannot be detected. Ankyrin and band 4.1 were detected in the majority of the cells by day 7 of culture, at the colony-forming unit (CFU)-E stage, whereas only 15% of the cells were positive for band 3 protein on day 7 of culture. The mRNA level for spectrin was already at its maximum on day 8 whereas the mRNAs for band 3, ankyrin, and band 4.1 were just beginning to accumulate. After enucleation, spectrin, band 3, ankyrin, and band 4.1 fluorescence were all associated with the reticulocytes. Actin was localized at the constriction between the extruding nucleus and the incipient reticulocyte in enucleating erythroblasts suggesting a key role for actin in the enucleation of human erythroblasts. Our investigations have also shown that purified human erythroid progenitors cultured in serum-free suspension media are capable of enucleating without the requirement of an extracellular matrix. These results demonstrate that the synthesis and expression of major cytoskeletal proteins in the human erythrocyte membrane occur in an asynchronous manner and that the remodeling of the membrane skeleton begins at a very early stage during erythrocyte development.


Assuntos
Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Células Precursoras Eritroides/metabolismo , Neuropeptídeos , RNA Mensageiro/metabolismo , Actinas/metabolismo , Proteína 1 de Troca de Ânion do Eritrócito/metabolismo , Anquirinas/metabolismo , Senescência Celular , Membrana Eritrocítica/metabolismo , Células Precursoras Eritroides/citologia , Humanos , Proteínas de Membrana/metabolismo , Espectrina/metabolismo , Distribuição Tecidual
18.
Blood ; 84(6): 1992-9, 1994 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-8081001

RESUMO

Erythroid cells express a unique form of beta spectrin I as a result of tissue-specific alternative pre-mRNA processing. Nonerythroid cells that express the beta spectrin I gene include four additional exons at the 3' end of the mature transcript, leading to elongation of the carboxyl terminus of the protein. The nonerythroid beta spectrin I isoform is not present in the red blood cell membrane skeleton; the erythroid isoform is not detected in other cell types. Therefore, developing erythroid cells acquire this tissue-specific pre-mRNA processing activity during differentiation. In the present study, we investigated the developmental timing of erythroid-specific pre-mRNA processing in human erythroid precursors. Partially purified human peripheral blood burst forming uniterythroid (BFU-E) cells were grown in culture for 5 to 12 days. beta Spectrin I mRNA transcripts were analyzed at different time points by S1 nuclease mapping. The processing of beta spectrin I transcripts was found to be exclusively erythroid from day 5 onward, indicating that erythroid-specific processing is not linked temporally to assembly of the mature erythroid membrane skeleton. Human erythroleukemia (HEL) cells had both erythroid and nonerythroid transcripts, indicating that both processing patterns can coexist. Induction of erythroid differentiation in HEL cells using hemin resulted in a partial switch toward the erythroid processing pattern of beta spectrin I transcripts. Using a genomic S1 probe that spans the erythroid polyadenylation signal, we found that a substantial portion of the transcripts detected by the erythroid cDNA S1 probe (in both cultured BFU-E and HEL cells) is incompletely processed pre-mRNA precursors. Poly(A) RNA selection before S1 analysis showed that the unprocessed transcripts are not polyadenylated. We conclude that (1) erythroid-specific pre-mRNA processing activity is present early in erythroid differentiation; (2) beta spectrin I transcripts that are unprocessed at the 3' end accumulate, awaiting either erythroid or nonerythroid processing pathways, from which observation we infer that the regulated alternative pathways are both inefficient; and (3) HEL cells offer a human cell culture model in which to study the balance between the two pre-mRNA processing pathways. We speculate that erythroid cells evolved this tissue-specific pre-mRNA processing machinery for other erythroid genes in addition to beta spectrin I.


Assuntos
Células Precursoras Eritroides/metabolismo , Precursores de RNA/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Espectrina/genética , Sequência de Bases , Diferenciação Celular , Sondas de DNA , Hemina/farmacologia , Humanos , Dados de Sequência Molecular , Endonucleases Específicas para DNA e RNA de Cadeia Simples , Células Tumorais Cultivadas
19.
J Clin Invest ; 94(1): 34-43, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7518834

RESUMO

Erythropoietin (EP), insulin-like growth factor I (IGF-I) and stem cell factor (SCF) each reduce apoptosis of human erythroid progenitor cells. To determine if these growth factors have additional roles in stimulating erythropoiesis, the proliferation, maturation, and survival of highly purified human erythroid colony-forming cells (ECFCs) were studied during the application of different combinations of these growth factors in a serum-free liquid culture. EP maintained cell viability and supported heme synthesis during erythroid maturation, with little increase in viable cell number or stimulation of DNA synthesis. The addition of SCF with EP resulted in a substantial increase in DNA synthesis, which was greater than that seen with the addition of EP and was associated with a large expansion in the number of ECFCs. Thus EP, by itself, produces little increase in cell proliferation, and expansion of the number of erythroid cells depends upon the presence of SCF with EP. The addition of IGF-I with EP led to enhanced heme synthesis and moderate cellular proliferation, but also greatly enhanced nuclear condensation and enucleation in the late erythroblasts. Thus EP, by itself, is not sufficient for complete end-terminal nuclear condensation/enucleation and the presence of IGF-I is necessary for this complete process. While EP greatly reduced apoptosis during 16 h of incubation at 37 degrees C, the addition of SCF and IGF-I with EP had little additional effect, but these additions enhanced DNA synthesis > 3.4-fold. Thus SCF may have an additional role in directly stimulating proliferation through a process that is distinct from apoptosis. Our observations indicate that EP prevents apoptosis and maintains erythroid cell viability and development. IGF-I enhances erythroid maturation and proliferation, but the proliferation of erythroid progenitors is mainly controlled by the addition of SCF with EP, independent of an effect on apoptosis.


Assuntos
Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Fatores de Crescimento de Células Hematopoéticas/farmacologia , Fator de Crescimento Insulin-Like I/farmacologia , Apoptose/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Células Precursoras Eritroides/fisiologia , Heme/biossíntese , Humanos , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco
20.
Blood ; 82(8): 2340-52, 1993 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-8400286

RESUMO

Murine erythroid progenitors infected with the anemia-inducing strain of Friend virus (FVA cells) undergo apoptosis when deprived of erythropoietin (EPO). When cultured with EPO, they survive and complete terminal differentiation. Although cell volume is decreased and nuclear chromatin is condensed during both apoptosis and terminal differentiation, morphologic and biochemical distinctions between these two processes were observed. In apoptosis, homogeneous nuclear condensation with nuclear envelope loss occurred in cells that had not reached the stage of hemoglobin synthesis. In terminal erythroid differentiation, nuclear condensation with heterochromatin, euchromatin, and nuclear envelope preservation occurred simultaneously with hemoglobin synthesis. Cells with apoptotic morphology appeared asynchronously in EPO-deprived cultures, indicating that only a portion of the cells were undergoing apoptosis at any given time. The percentages of apoptotic cells and cleaved DNA increased with time in EPO-deprived cultures. Inhibition of DNA cleavage was directly proportional to EPO concentration over a wide physiologic range, demonstrating a heterogeneity in susceptibility to apoptosis based on variability in the EPO sensitivity of individual cells. A subpopulation of FVA cells with increased EPO sensitivity (decreased EPO requirement) was isolated from EPO-deprived cultures. This increased EPO sensitivity did not result from differences in EPO receptor number, affinity, or structure, suggesting that the differences are in the signal transduction pathway. These results indicate that control of red blood cell production involves both prevention of apoptosis by EPO and heterogeneity in the EPO requirement of individual progenitor cells.


Assuntos
Apoptose , Eritroblastos/efeitos dos fármacos , Eritropoese , Eritropoetina/farmacologia , Animais , Sequência de Bases , Diferenciação Celular , Fracionamento Celular , Células Cultivadas , DNA/metabolismo , Eritroblastos/fisiologia , Envelhecimento Eritrocítico/efeitos dos fármacos , Eritropoetina/biossíntese , Vírus da Leucemia Murina de Friend , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/análise , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...