Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Gene Ther ; 30(3-4): 222-231, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-34997202

RESUMO

Autologous chimeric antigen receptor (CAR) T cells targeting the CD19 antigen have demonstrated a high complete response rate in relapsed/refractory B-cell malignancies. However, autologous CAR T cell therapy is not an option for all patients. Here we optimized conditions for clinical-grade manufacturing of allogeneic CD19-CAR T cells using CD45RA-depleted donor memory T cells (Tm) for a planned clinical trial. Tm were activated using the MACS GMP T Cell TransAct reagent and transduced in the presence of LentiBOOST with a clinical-grade lentiviral vector that encodes a 2nd generation CD19-CAR with a 41BB.zeta endodomain. Transduced T cells were transferred to a G-Rex cell culture device for expansion and harvested on day 7 or 8 for cryopreservation. The resulting CD19-CAR(Mem) T cells expanded on average 34.2-fold, and mean CAR expression was 45.5%. The majority of T cells were CD4+ and had a central memory or effector memory phenotype, and retained viral specificity. CD19-CAR(Mem) T cells recognized and killed CD19-positive target cells in vitro and had potent antitumor activity in an ALL xenograft model. Thus we have successfully developed a current good manufacturing practice-compliant process to manufacture donor-derived CD19-CAR(Mem) T cells. Our manufacturing process could be readily adapted for CAR(Mem) T cells targeting other antigens.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T , Humanos , Antígenos CD19/genética , Imunoterapia Adotiva/métodos , Linfócitos T , GMP Cíclico/metabolismo
2.
Cytotherapy ; 25(11): 1149-1154, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37676217

RESUMO

BACKGROUND AIMS: Intracranial (IC) locoregional delivery of chimeric antigen receptor (CAR) T cells presents an attractive delivery method to central nervous system tumors. Although IC delivery is actively being employed in early-phase clinical studies, no thaw/wash methods have been published to remove the neurotoxic cryoprotectant dimethyl sulfoxide (DMSO) from CAR T-cell products before IC administration. Thus, the aim of this study was to develop and validate a simple thaw/wash procedure. METHODS: We developed a thaw/wash procedure that consist of product thaw at 37°C, equilibration for 5 min in 1 volume of preservative-free normal saline (PFNS), dilution with an additional 8 volumes of PFNS, removal of DMSO through a washing step, resuspension in 2.0 mL of PFNS and storage in a syringe at 20-25°C. Final formulated products (FPs) were assessed for quality and safety attributes and stability over 3 h from the completion of the thaw. Stability parameters included CAR T-cell viability, transgene surface expression and cytolytic activity. RESULTS: The developed procedure reduced the calculated % of DMSO to less than 0.025%. FP cell viability and recovery (versus pre-cryopreservation) were within acceptable specifications (mean viability: 85.3%, range: 83%-88%; total nucleated cell recovery mean: 76.5%, range: 65.4%-82.5%). Other prespecified quality assurance/quality control parameters including appearance/ integrity, sterility and endotoxin level (≤1.0 EU/mL), were also met by all FPs (n = 3). Three hours' post thaw/wash stability was confirmed. All products maintained cell viability greater than 70% (mean, 80.0%; range, 79%-81%), with no significant change in transgene expression or cytolytic activity of B7-H3-CAR T cells compared with thawed not diluted/washed control CAR T cells. CONCLUSIONS: We have developed a simple thaw/wash procedure to prepare B7-H3-CAR T cells for their locoregional delivery to the neural axis. While we focus here on CAR T cells, the methods could be readily adapted to other cryopreserved immune effector cell products.


Assuntos
Receptores de Antígenos Quiméricos , Receptores de Antígenos Quiméricos/genética , Dimetil Sulfóxido , Criopreservação/métodos , Crioprotetores , Linfócitos T
3.
Cytotherapy ; 25(11): 1145-1148, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37598335

RESUMO

BACKGROUND AIMS: The current approach for preventing hemolysis of red blood cells (RBCs) in major ABO-incompatible bone marrow (BM) grafts after infusion is to deplete RBCs from BM products before transplantation. Traditionally, manual density separation (MDS) using Ficoll-Hypaque (Cytiva Sweden AB, Uppsala, Sweden has been used to accomplish RBC depletion. This process yields good CD34+ cell recovery, but it requires open manipulation and is labor-intensive and time-consuming. We hypothesized that an alternative automated method using Haemonetics Cell Saver 5+ (Haemonetics Corporation, Boston, MA, USA) would offer equivalent RBC depletion and CD34+ cell recovery. Small marrow volumes from pediatric donors can be processed using Cell Saver (CS) without adding the third-party RBCs necessary for other automated methods. METHODS: This retrospective analysis comprised data from 58 allogeneic BM grafts. RBC depletion and CD34+ cell recovery from BM using MDS (35 grafts) were compared with CS (14 grafts). Nine products underwent RBC depletion using CS with Ficoll (CS-F) when RBC volume was less than 125 mL. RESULTS: Linear regression analysis of log transformation of CD34+ cell recovery adjusted for log transformation of both baseline CD34+ cell content and baseline total volume showed no significant difference between MDS and CS (estimated coefficient, -0.121, P = 0.096). All products contained an RBC volume of less than 0.25 mL/kg post-processing. CD34+ cell recovery with CS-F was comparable to MDS and CS and suitable for pediatric recipients of allogeneic hematopoietic cell transplantation. CONCLUSIONS: We provide evidence that an automated method using Haemonetics Cell Saver 5+ achieves RBC depletion and CD34+ cell recovery comparable to MDS when adjusting for baseline factors.


Assuntos
Transplante de Medula Óssea , Medula Óssea , Criança , Humanos , Células da Medula Óssea , Transplante de Medula Óssea/métodos , Separação Celular/métodos , Eritrócitos , Ficoll , Estudos Retrospectivos
4.
Br J Haematol ; 198(4): 740-744, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35737751

RESUMO

We adjusted haematopoietic stem and progenitor cell (HSPC) apheresis collection from patients with sickle cell disease (SCD) by targeting deep buffy coat collection using medium or low collection preference (CP), and by increasing anticoagulant-citrate-dextrose-solution A dosage. In 43 HSPC collections from plerixafor-mobilized adult patients with SCD, we increased the collection efficiency to 35.79% using medium CP and 82.23% using low CP. Deep buffy coat collection increased red blood cell contamination of the HSPC product, the product haematocrit was 4.7% with medium CP and 6.4% with low CP. These adjustments were well-tolerated and allowed efficient HSPC collection from SCD patients.


Assuntos
Anemia Falciforme , Remoção de Componentes Sanguíneos , Compostos Heterocíclicos , Adulto , Anemia Falciforme/terapia , Benzilaminas , Ciclamos , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas , Humanos , Leucaférese
5.
Transfusion ; 59(7): 2375-2381, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31066469

RESUMO

BACKGROUND: Bone marrow (BM) grafts are often subject to manipulation to minimize infusion volume, to deplete red blood cells (RBCs), to deplete plasma, and/or to achieve optimal concentration of nucleated cells (NCs) for freezing. Here, we report results of processing and infusion of four major ABO-incompatible BM grafts for allogeneic hematopoietic transplantation in pediatric patients. CASES AND METHODS: Patients ranging in age from 5 months to 18 years received hematopoietic stem cell transplantation for benign or malignant indications. Allogeneic BM grafts were processed using PrepaCyte-CB kits, approved by the Food and Drug Administration for enrichment of mononuclear cells in cord blood units for hematopoietic transplantation. Resulting grafts were dosed and infused using standard institutional protocol. After transplantation patients were evaluated for hematologic recovery. RESULTS: The PrepaCyte-CB processing method achieved on average 97.1% depletion of RBCs and 68.5% volume reduction, with a fairly efficient NC recovery (total NCs, 85.6%; white blood cells, 89.7%; CD34+ cells, 103.3%; and colony-forming unit cells, 87.5%). No microbial contamination was detectable in any of the processed grafts. Infusions were well tolerated, and all four patients achieved durable hematopoietic engraftment within expected time interval. CONCLUSION: Our results suggest that PrepaCyte-CB is safe and effective for minimal processing of BM grafts. Engraftment outcomes were comparable with other published BM processing methods. In addition, NC recovery achieved using this method appears to exceed that of previously described sedimentation reagents such as hetastarch and Ficoll.


Assuntos
Medula Óssea , Separação Celular , Eritrócitos , Transplante de Células-Tronco Hematopoéticas , Adolescente , Sedimentação Sanguínea , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Transplante Homólogo
6.
Transfusion ; 54(11): 2997-3009, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24894338

RESUMO

Methods of handling, thawing, and infusion of cord blood (CB) products vary substantially among thaw/transplant centers (TCs). This review 1) compares currently available CB product types and thaw methods recommended by CB banks (CBBs), 2) discusses causes of inconsistency in thaw method application at TCs, 3) advises elements to consider in thaw method approval or selection at the TC, 4) provides a procedural template for the traditional thaw methods, and 5) suggests acceptable time from product thaw to infusion and other considerations for safe infusion. It also compares postinfusion adverse reaction and engraftment data as functions of thaw methods. Remarks and suggestions made throughout this review are: 1) not intended to supersede manufacturer's instructions but meant to support the standardization of preparative procedures recommended by CBBs and 2) intended to help TCs to investigate relevant quality issues and handle challenges, especially when the TC is unable to follow recommendations due to foreseeable technical, quality, and/or clinical factors.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Criopreservação/métodos , Sangue Fetal , Sobrevivência de Enxerto , Segurança , Humanos , Guias de Prática Clínica como Assunto
7.
J Hematol Oncol ; 17(1): 50, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38937803

RESUMO

BACKGROUND: Relapse remains a challenge after transplantation in pediatric patients with hematological malignancies. Myeloablative regimens used for disease control are associated with acute and long-term adverse effects. We used a CD45RA-depleted haploidentical graft for adoptive transfer of memory T cells combined with NK-cell addback and hypothesized that maximizing the graft-versus-leukemia (GVL) effect might allow for reduction in intensity of conditioning regimen. METHODS: In this phase II clinical trial (NCT01807611), 72 patients with hematological malignancies (complete remission (CR)1: 25, ≥ CR2: 28, refractory disease: 19) received haploidentical CD34 + enriched and CD45RA-depleted hematopoietic progenitor cell grafts followed by NK-cell infusion. Conditioning included fludarabine, thiotepa, melphalan, cyclophosphamide, total lymphoid irradiation, and graft-versus-host disease (GVHD) prophylaxis consisted of a short-course sirolimus or mycophenolate mofetil without serotherapy. RESULTS: The 3-year overall survival (OS) and event-free-survival (EFS) for patients in CR1 were 92% (95% CI:72-98) and 88% (95% CI: 67-96); ≥ CR2 were 81% (95% CI: 61-92) and 68% (95% CI: 47-82) and refractory disease were 32% (95% CI: 11-54) and 20% (95% CI: 6-40). The 3-year EFS for all patients in morphological CR was 77% (95% CI: 64-87) with no difference amongst recipients with or without minimal residual disease (P = 0.2992). Immune reconstitution was rapid, with mean CD3 and CD4 T-cell counts of 410/µL and 140/µL at day + 30. Cumulative incidence of acute GVHD and chronic GVHD was 36% and 26% but most patients with acute GVHD recovered rapidly with therapy. Lower rates of grade III-IV acute GVHD were observed with NK-cell alloreactive donors (P = 0.004), and higher rates of moderate/severe chronic GVHD occurred with maternal donors (P = 0.035). CONCLUSION: The combination of a CD45RA-depleted graft and NK-cell addback led to robust immune reconstitution maximizing the GVL effect and allowed for use of a submyeloablative, TBI-free conditioning regimen that was associated with excellent EFS resulting in promising long-term outcomes in this high-risk population. The trial is registered at ClinicalTrials.gov (NCT01807611).


Assuntos
Neoplasias Hematológicas , Transplante de Células-Tronco Hematopoéticas , Células Matadoras Naturais , Células T de Memória , Condicionamento Pré-Transplante , Transplante Haploidêntico , Humanos , Feminino , Masculino , Células Matadoras Naturais/transplante , Células Matadoras Naturais/imunologia , Criança , Adolescente , Transplante Haploidêntico/métodos , Pré-Escolar , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Condicionamento Pré-Transplante/métodos , Neoplasias Hematológicas/terapia , Doença Enxerto-Hospedeiro/prevenção & controle , Doença Enxerto-Hospedeiro/etiologia , Lactente , Adulto Jovem , Adulto , Resultado do Tratamento , Efeito Enxerto vs Leucemia
8.
Transfusion ; 53(12): 3251-61, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23621871

RESUMO

BACKGROUND: The St Louis Cord Blood Bank submitted a biologics license application for cord blood (CB) products processed by PrepaCyte-CB (BioE), supported with a validation study of a microbial detection system for product sterility testing (BACTEC-FX, Becton Dickinson). This article provides the validation approach followed to fulfill Food and Drug Administration requirements pertinent to sterility testing method. STUDY DESIGN AND METHODS: System qualification, culture media quality verification, and validation of CB processing by-product (CB-BP) sample as surrogate to final product for sterility testing were followed by studies evaluating method sensitivity, specificity, reproducibility, ruggedness or robustness, and bacteriostatic or fungistatic effect of CB-BP sample. CB-BP cultures and control samples were formulated using BACTEC Plus Aerobic/F, Plus Anaerobic/F, and Myco F/Lytic media. Samples were seeded with selected test organisms (n = 13 at 10-100 colony-forming units [CFUs] per vial) and cultured for 14 days (bacterial) and 30 days (fungal). RESULTS: Under testing conditions, no stasis effect of test sample on microbial growth and no false-positive or false-negative results were reported. Although a 7-day culture was sufficient to detect all validation test organisms seeded at ≤ 26 CFUs/vial, growth in actual product sterility testing practice may require a 10- to 14-day culture. Assay reproducibility was uncertain at very low bioburden (< 10 CFUs/vial). Growth time to detection neither varied between different media lots nor prolonged in culture vials with loading delay (6-8 hr at room temperature). CONCLUSION: BACTEC-FX culture and detection system and BACTEC media formulae have high detection capability and can be effectively validated for sterility testing of CB products.


Assuntos
Sangue Fetal , Meios de Cultura , Humanos , Controle de Infecções
9.
Blood Adv ; 6(21): 5737-5749, 2022 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-35446934

RESUMO

T cells expressing CD19-specific chimeric antigen receptors (CD19-CARs) have potent antileukemia activity in pediatric and adult patients with relapsed and/or refractory B-cell acute lymphoblastic leukemia (B-ALL). However, not all patients achieve a complete response (CR), and a significant percentage relapse after CD19-CAR T-cell therapy due to T-cell intrinsic and/or extrinsic mechanisms. Thus, there is a need to evaluate new CD19-CAR T-cell products in patients to improve efficacy. We developed a phase 1/2 clinical study to evaluate an institutional autologous CD19-CAR T-cell product in pediatric patients with relapsed/refractory B-ALL. Here we report the outcome of the phase 1 study participants (n = 12). Treatment was well tolerated, with a low incidence of both cytokine release syndrome (any grade, n = 6) and neurotoxicity (any grade, n = 3). Nine out of 12 patients (75%) achieved a minimal residual disease-negative CR in the bone marrow (BM). High disease burden (≥40% morphologic blasts) before CAR T-cell infusion correlated with increased side effects and lower response rate, but not with CD19-CAR T-cell expansion. After infusion, CD8+ CAR T cells had a proliferative advantage over CD4+ CAR T cells and at peak expansion, had an effector memory phenotype with evidence of antigen-driven differentiation. Patients that proceeded to allogeneic hematopoietic cell transplantation (AlloHCT) had sustained, durable responses. In summary, the initial evaluation of our institutional CD19-CAR T-cell product demonstrates safety and efficacy while highlighting the impact of pre-infusion disease burden on outcomes. This trial was registered at www.clinicaltrials.gov as #NCT03573700.


Assuntos
Linfoma de Burkitt , Linfoma de Células B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Receptores de Antígenos Quiméricos , Humanos , Antígenos CD19 , Linfócitos T CD8-Positivos , Efeitos Psicossociais da Doença , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Linfócitos T
10.
Stem Cell Res Ther ; 12(1): 276, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33957983

RESUMO

BACKGROUND: Mucopolysaccharidosis IVA (Morquio A syndrome) is a lysosomal storage disease caused by the deficiency of enzyme N-acetylgalactosamine-6-sulfate sulfatase (GALNS), which results in the accumulation of the glycosaminoglycans (GAGs), keratan sulfate, and chondroitin-6-sulfate in the lysosomes of all tissues causing systemic dysfunction. Current treatments include enzyme replacement therapy (ERT) which can treat only certain aspects of the disease such as endurance-related biological endpoints. A key challenge in ERT is ineffective enzyme uptake in avascular tissues, which makes the treatment of the corneal, cartilage, and heart valvular tissue difficult. The aim of this study was to culture human umbilical mesenchymal stem cells (UMSC), demonstrate presence of GALNS enzyme activity within the extracellular vesicles (EVs) derived from these UMSC, and study how these secreted EVs are taken up by GALNS-deficient cells and used by the deficient cell's lysosomes. METHODS: We obtained and cultured UMSC from the umbilical cord tissue from anonymous donors from the Saint Louis Cord Blood Bank. We characterized UMSC cell surface markers to confirm phenotype by cell sorting analyses. In addition, we confirmed that UMSC secrete GALNS enzyme creating conditioned media for co-culture experiments with GALNS deficient cells. Lastly, we isolated EVs derived from UMSC by ultracentrifugation to confirm source of GALNS enzyme. RESULTS: Co-culture and confocal microscopy experiments indicated that the lysosomal content from UMSC migrated to deficient cells as evidenced by the peak signal intensity occurring at 15 min. EVs released by UMSC were characterized indicating that the EVs contained the active GALNS enzyme. Uptake of GALNS within EVs by deficient fibroblasts was not affected by mannose-6-phosphate (M6P) inhibition, suggesting that EV uptake by these fibroblasts is gradual and might be mediated by a different means than the M6P receptor. CONCLUSIONS: UMSC can deliver EVs containing functional GALNS enzyme to deficient cells. This enzyme delivery method, which was unaffected by M6P inhibition, can function as a novel technique for reducing GAG accumulation in cells in avascular tissues, thereby providing a potential treatment option for Morquio A syndrome.


Assuntos
Condroitina Sulfatases , Vesículas Extracelulares , Células-Tronco Mesenquimais , Mucopolissacaridose IV , Fibroblastos , Humanos , Mucopolissacaridose IV/genética , Mucopolissacaridose IV/terapia
12.
Oncogene ; 24(37): 5751-63, 2005 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-16123808

RESUMO

Transforming growth factor-beta (TGF-beta) is a pleiotropic regulator of all stages of hematopoieis. The three mammalian isoforms (TGF-beta1, 2 and 3) have distinct but overlapping effects on hematopoiesis. Depending on the differentiation stage of the target cell, the local environment and the concentration and isoform of TGF-beta, in vivo or in vitro, TGF-beta can be pro- or antiproliferative, pro- or antiapoptotic, pro- or antidifferentiative and can inhibit or increase terminally differentiated cell function. TGF-beta is a major regulator of stem cell quiescence, at least in vitro. TGF-beta can act directly or indirectly through effects on the bone marrow microenvironment. In addition, paracrine and autocrine actions of TGF-beta have overlapping but distinct regulatory effects on hematopoietic stem/progenitor cells. Since TGF-beta can act in numerous steps in the hematopoietic cascade, loss of function mutations in hematopoeitic stem cells (HSC) have different effects on hematopoiesis than transient blockade of autocrine TGF-beta1. Transient neutralization of autocrine TGF-beta in HSC has therapeutic potential. In myeloid and erythroid leukemic cells, autocrine TGF-beta1 and/or its Smad signals controls the ability of these cells to respond to various differentiation inducers, suggesting that this pathway plays a role in determining the cell fate of leukemic cells.


Assuntos
Hematopoese , Fator de Crescimento Transformador beta/fisiologia , Animais , Proliferação de Células , Sobrevivência Celular , Proteínas de Ligação a DNA/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/fisiologia , Humanos , Leucemia Eritroblástica Aguda/etiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Transdução de Sinais , Proteínas Smad , Transativadores/fisiologia
13.
Stem Cells Int ; 2016: 3274054, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27034683

RESUMO

Recent studies have demonstrated that the umbilical cord (UC) is an excellent source of mesenchymal stromal cells (MSCs). However, current protocols for extracting and culturing UC-MSCs do not meet current good manufacturing practice (cGMP) standards, in part due to the use of xenogeneic reagents. To support the development of a cGMP-compliant method, we have examined an enzyme-free isolation method utilizing tissue homogenization (t-H) followed by culture in human platelet lysate (PL) supplemented media. The yield and viability of cells after t-H were comparable to those obtained after collagenase digestion (Col-D). Importantly, kinetic analysis of cultured cells showed logarithmic growth over 10 tested passages, although the rate of cell division was lower for t-H as compared to Col-D. This slower growth of t-H-derived cells was also reflected in their longer population doubling time. Interestingly, there was no difference in the expression of mesenchymal markers and trilineage differentiation potential of cells generated using either method. Finally, t-H-derived cells had greater clonogenic potential compared to Col-D/FBS but not Col-D/PL and were able to maintain CFU-F capacity through P7. This bench scale study demonstrates the possibility of generating therapeutic doses of good quality UC-MSCs within a reasonable length of time using t-H and PL.

14.
Saudi Med J ; 26(10): 1523-8, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16228049

RESUMO

OBJECTIVE: To assess, in vitro, the effect of Amifostine (AMF, WR-2721) on angiogenesis and levels of vascular endothelial growth factor (VEGF) secreted from hemopoietic stem/progenitor cell populations. METHODS: We conducted the study in the research laboratories of the Hashemite University, Jordan between September 2003 and January 2005 where we took samples were from Myelodysplastic syndrome (MDS) patients and healthy donors attending Al-Hussein Cancer Center and We determined the proliferation of human umbilical vein endothelial cells (HUVECs) in cultures supplemented with media conditioned with AMF-treated and AMF-untreated pure hemopoietic cells [CD34+ cells, and erythroid, myeloid and megakaryocytic progenitors]. Furthermore, in the same conditioned media, we evaluated levels of elaborated VEGF by a sensitive enzyme linked immunosorbent assay. RESULTS: Biologically, media conditioned with AMF-treated cells reduced proliferation of HUVECs compared to media conditioned with untreated control cells (p<0.05). In cultures of AMF-untreated cells, elaboration of VEGF was higher (p<0.05) in media conditioned with cells from MDS patients compared to healthy donors. A 30 minutes pre-exposure of cells to AMF (500 mM) suppressed levels of VEGF secreted within 24 hours in 63 of 89 evaluated cultures. The percentage of reduction of VEGF in AMF-sensitive cultures was comparable in cultures of MDS cells (18%, 2-37%; median, range) and normal cells (12%, 2-45%). CONCLUSION: The results showed that AMF exerts an anti-angiogenic activity and suppresses the secretion of VEGF in hemopoietic stem/progenitor cells obtained from both healthy individuals and patients with MDS.


Assuntos
Amifostina/farmacologia , Proliferação de Células/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/efeitos dos fármacos , Estudos de Casos e Controles , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Feminino , Células-Tronco Hematopoéticas/citologia , Humanos , Masculino , Síndromes Mielodisplásicas/sangue , Síndromes Mielodisplásicas/patologia , Probabilidade , Valores de Referência , Estudos de Amostragem , Sensibilidade e Especificidade , Células-Tronco/citologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
15.
Int J Hematol ; 77(5): 476-81, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12841386

RESUMO

Coexistence of Philadelphia chromosome-negative (Ph-) progenitors with the Ph+ clone in the early chronic phase of chronic myeloid leukemia (CML) has been documented in previous reports. A different evaluation of methods is needed to justify the clonality of the residual Ph- progenitors. Therefore, the X chromosome inactivation patterns in individual granulocyte-monocyte colony-forming unit (CFU-GM) colonies were studied with the clonality assay for the human androgen receptor gene. A prerequisite for this evaluation was the validation of T-lymphocytes and buccal cells as control cells representing the constitutional lyonization. The percentages of polyclonal CFU-GM cells were determined in 9 Ph+ women with CML and in 5 healthy women. Results of the clonal analysis of CFU-GM colonies were compared with those from reverse transcriptase-polymerase chain reaction analysis of single colonies for BCR/ABL transcripts. Both methods of CFU-GM cell analysis were in agreement regarding the presence of variable proportions (0%-94%) of normal cells in CML. Our results suggest that (a) T-cells and buccal cells have potential for use as controls for the clonal analysis of CML cases and (b) this method can evaluate the frequency of polyclonal/clonal CFU-GM cells in CML cases and is applicable to the analysis of myeloid clonal disorders that lack specific molecular markers.


Assuntos
Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Células Progenitoras Mieloides/patologia , Receptores Androgênicos/genética , Adulto , Estudos de Casos e Controles , Células Clonais/patologia , Mecanismo Genético de Compensação de Dose , Feminino , Proteínas de Fusão bcr-abl/genética , Humanos , Leucemia Mieloide Crônica Atípica BCR-ABL Negativa/patologia , Métodos , Pessoa de Meia-Idade , RNA Mensageiro/análise
16.
J Leuk (Los Angel) ; 1(2)2013 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-24533426

RESUMO

The role of crosstalk between the Smad and the MAPK signaling pathways in activin-, transforming growth factor-ß (TGF-ß)-, hydroxyurea (HU) - and butyrate-dependent erythroid differentiation of K562 leukemic cells was studied. Treatment with all four inducers caused transient phosphorylation of Smad2/3 and MAPK proteins including ERK, p38 and JNK. Use of specific inhibitors of p38, ERK and JNK MAPK proteins, and TGF-ß type I receptor indicated that differentiation induced by each of these agents involves activation of Smad2/3 and p38 MAPK, and inhibition of ERK MAPK. Also, treatment of cells with an inhibitor of protein serine/threonine phosphatase, okadaic acid (OA), induced phosphorylation of Smad2/3, and p38 MAPK, coincident with its induction of erythroid differentiation. Specific inhibition of TGF-ß type I receptor kinase activity not only abolished TGF-ß/activin effects but also prevented Smad2/3 activation and erythroid differentiation induced by OA, HU and butyrate. The TGF-ß type I receptor kinase inhibitor blocked OA-induced differentiation but not p38 MAPK phosphorylation demonstrating that signals from both pathways are needed. As previously observed, addition of ERK1/2 MAPK inhibitors upregulated Smad2/3 phosphorylation and enhanced differentiation, but these effects were dependent on signals from the TGF-ß type I receptor. These data indicate that activation of both Smad2/3 and p38 MAPK signaling pathways is a prerequisite to induce erythroid differentiation of erythroleukemia cells by activin, TGF-ß, HU, OA and butyrate.

17.
Platelets ; 18(8): 566-78, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18041647

RESUMO

Smad and MAPK signaling cascades are involved in erythroid and megakaryocytic differentiation. The inhibitory Smad for TGF-beta/activin signaling, Smad7, may directly or indirectly affect these signaling pathways. By modulating Smad7 expression, we attempted to delineate the relevance of Smad7 during erythro-megakaryocytic (E/M) differentiation of human erythroleukemia cells. Smad7 transcripts were detected at low levels in different erythroleukemia cell lines (TF-1, HEL and K562). Reduction of expression of endogenous Smad7 by RNA interference enhanced erythroid differentiation of K562 cells in response to physiological doses of activin-A/TGF-beta1. Stable over-expression of Smad7 in K562 cells (K562/7) prevented activation of Smad2/3 and MAPK (ERK1/2, p38 and JNK1/2) proteins by activin-A/TGF-beta1 and subsequent induction of erythroid differentiation. High levels of Smad7 also interfered with hydroxyurea- and butyrate-, but not hemin-induced erythroid differentiation. Interestingly, K562/7 cells were found to harbor a significant proportion (about 35%) of large ploy nucleated cells compared to fewer than 12% in control cells. K562/7 cells treated with phorbol 12-myristate 13-acetate (PMA), showed a great shift in ploidy towards high ploidy classes (> or =8N) accompanied with an increase in the expression of the maturation marker CD42b. We showed here that: (a) low levels of endogenous Smad7 in erythroleukemia cells are physiologically relevant, and (b) high levels of Smad7 interferes with TGF-beta/activin-induced Smad/MAPK signaling and erythro-differentiation and promotes megakaryocytic differentiation, possibly by blocking autocrine TGF-beta.


Assuntos
Diferenciação Celular , Células Eritroides/citologia , Leucemia Eritroblástica Aguda/patologia , Megacariócitos/citologia , Transdução de Sinais , Proteína Smad7/análise , Ativinas/metabolismo , Comunicação Autócrina , Regulação da Expressão Gênica , Humanos , Células K562 , Sistema de Sinalização das MAP Quinases , Proteínas Smad/metabolismo , Proteína Smad7/genética , Fator de Crescimento Transformador beta/metabolismo
18.
J Immunol ; 174(7): 4262-70, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15778389

RESUMO

The retrovirus human T cell leukemia virus (HTLV) type I (HTLV-I) is primarily transmitted by breast-feeding or sexual contact, by cell-to-cell contact between T cells. TGF-beta, which has been shown to enhance transmission of HTLV-I in vitro, is found at high levels in breast milk and semen. In this study, the ability of TGF-beta to regulate expression of molecules involved in HTLV-I binding and entry was examined. Previous studies using a soluble form of the HTLV-I envelope protein SU have shown that quiescent human T cells do not express cell surface molecules that specifically bind SU. After T cell activation, HTLV SU binding proteins are rapidly induced. In this study, we report that TGF-beta induces expression of proteins that bind soluble HTLV SU and HTLV virions on naive CD4(+) T lymphocytes. The induction of these proteins occurred without cell cycle entry or expression of activation markers, involved TGF-beta-induced intracellular signaling, and required de novo transcription and translation. Treatment of naive CD4(+) T lymphocytes with TGF-beta induced expression of GLUT-1, which has recently been reported to function as a receptor for HTLV. Treatment of a TGF-beta-sensitive human myeloid cell line increased the titer of both HTLV-I- and HTLV-II-pseudotyped viruses. Although earlier studies suggested that HTLV SU binding proteins might be an early marker of T cell activation and/or cell proliferation, we report in this study that TGF-beta induces binding of HTLV virions and expression of glucose transporter type 1 in primary CD4(+) T lymphocytes that remain quiescent.


Assuntos
Linfócitos T CD4-Positivos/virologia , Regulação da Expressão Gênica/efeitos dos fármacos , Vírus Linfotrópico T Tipo 1 Humano , Proteínas de Transporte de Monossacarídeos/genética , Receptores Virais/genética , Fator de Crescimento Transformador beta/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Sangue Fetal , Produtos do Gene env/metabolismo , Transportador de Glucose Tipo 1 , Humanos , Ativação Linfocitária , Ligação Proteica , Regulação para Cima
19.
Stem Cells ; 21(5): 557-67, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12968110

RESUMO

Transforming growth factor (TGF)-beta1 exerts autocrine and paracrine effects on hematopoiesis. Here, we have attempted to evaluate the effect of endogenous TGF-beta1 on early erythroid development from primitive human hematopoietic stem cells (HSCs) and to assess the effects of TGF-beta1 on different phases of erythropoiesis. Cord blood CD34(+)CD38(-) lineage-marker-negative (Lin(-)) cells were cultured in serum-free conditions using various combinations of stem cell factor (SCF), erythropoietin (Epo), and TGF-beta-neutralizing antibody. Generation of erythroid progenitors was assessed using colony assay and flow cytometry. Terminal erythroid differentiation was examined when SCF/Epo-stimulated cells were recultured in the presence of Epo with and without TGF-beta1. Anti-TGF-beta augmented the proliferation of CD34(+)CD38(-)Lin(-) cells (day 21) in SCF-stimulated (6.4-fold +/- 1.5-fold) and SCF/Epo-stimulated (2.9-fold +/- 1.2-fold) cultures. Cells stimulated by SCF/Epo underwent similar levels of erythroid differentiation with and without anti-TGF-beta. While SCF alone stimulated the production of tryptase-positive mast cells, cells stimulated by SCF/anti-TGF-beta were predominantly erythroid (CD36(+)CD14(-) and glycophorin A positive). A distinct expansion of erythroid progenitors (CD34(+)CD36(+)CD14(-)) with the potential to form erythroid colonies was seen, revealing early Epo-independent erythroid development. In contrast, the kinetics of erythroid progenitor generation from primitive HSCs indicate that TGF-beta1 is not inhibitory in late erythropoiesis, but it accelerated the conversion of large BFU-E into colony-forming units-erythroid. Finally, TGF-beta1 accelerated Epo-induced terminal erythroid differentiation and resulted in a greater level of enucleation (22% +/- 6% versus 7% +/- 3%) in serum-free conditions. Serum addition stimulated enucleation (54% +/- 18%), which was lower (26% +/- 14%) with anti-TGF-beta, suggesting that optimal erythroid enucleation is Epo dependent, requiring serum factors including TGF-beta1.


Assuntos
ADP-Ribosil Ciclase/análise , Antígenos CD34/análise , Antígenos CD/análise , Células Precursoras Eritroides/citologia , Eritropoese , Sangue Fetal/citologia , Fator de Células-Tronco/farmacologia , Fator de Crescimento Transformador beta/fisiologia , ADP-Ribosil Ciclase/imunologia , ADP-Ribosil Ciclase 1 , Antígenos CD/imunologia , Antígenos CD34/imunologia , Antígenos de Diferenciação/análise , Comunicação Autócrina , Diferenciação Celular , Linhagem da Célula , Células Precursoras Eritroides/química , Células Precursoras Eritroides/efeitos dos fármacos , Eritropoetina/farmacologia , Sangue Fetal/metabolismo , Hematopoese , Fatores de Crescimento de Células Hematopoéticas/efeitos dos fármacos , Humanos , Cinética , Glicoproteínas de Membrana , Testes de Neutralização , Fator de Células-Tronco/antagonistas & inibidores , Fator de Células-Tronco/metabolismo , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA