Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Mol Cell ; 81(2): 239-254.e8, 2021 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-33301730

RESUMO

Metazoan transcription factors typically regulate large numbers of genes. Here we identify via a CRISPR-Cas9 genetic screen ZNF410, a pentadactyl DNA-binding protein that in human erythroid cells directly activates only a single gene, the NuRD component CHD4. Specificity is conveyed by two highly evolutionarily conserved clusters of ZNF410 binding sites near the CHD4 gene with no counterparts elsewhere in the genome. Loss of ZNF410 in adult-type human erythroid cell culture systems and xenotransplantation settings diminishes CHD4 levels and derepresses the fetal hemoglobin genes. While previously known to be silenced by CHD4, the fetal globin genes are exposed here as among the most sensitive to reduced CHD4 levels.. In vitro DNA binding assays and crystallographic studies reveal the ZNF410-DNA binding mode. ZNF410 is a remarkably selective transcriptional activator in erythroid cells, and its perturbation might offer new opportunities for treatment of hemoglobinopathies.


Assuntos
DNA/genética , Células Precursoras Eritroides/metabolismo , Hemoglobina Fetal/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Fatores de Transcrição/genética , Animais , Sítios de Ligação , Células COS , Sistemas CRISPR-Cas , Chlorocebus aethiops , DNA/metabolismo , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Sangue Fetal/citologia , Sangue Fetal/metabolismo , Hemoglobina Fetal/metabolismo , Feto , Edição de Genes , Células HEK293 , Xenoenxertos , Humanos , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/química , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Camundongos , Modelos Moleculares , Células-Tronco Embrionárias Murinas/citologia , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ativação Transcricional
2.
Front Immunol ; 11: 597433, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329589

RESUMO

Newborns are highly susceptible to infectious diseases. The underlying mechanism of neonatal infection susceptibility has generally been related to their under-developed immune system. Nevertheless, this notion has recently been challenged by the discovery of the physiological abundance of immunosuppressive erythroid precursors CD71+erythroid cells (CECs) in newborn mice and human cord blood. Here, as proof of concept, we show that these cells are also abundant in the peripheral blood of human newborns. Although their frequency appears to be more variable compared to their counterparts in mice, they rapidly decline by 4 weeks of age. However, their proportion remains significantly higher in infants up to six months of age compared to older infants. We found CD45 expressing CECs, as erythroid progenitors, were the prominent source of reactive oxygen species (ROS) production in both humans and mice. Interestingly, a higher proportion of CD45+CECs was observed in the spleen versus bone marrow of neonatal mice, which was associated with a higher ROS production by splenic CECs compared to their siblings in the bone marrow. CECs from human newborns suppressed cytokine production by CD14 monocytes and T cells, which was partially abrogated by apocynin in vitro. Moreover, the depletion of CECs in neonatal mice increased the number of activated effector immune cells in their spleen and liver, which rendered them more resistant to Listeria monocytogenes infection. This was evident by a significant reduction in the bacteria load in the spleen, liver and brain of treated-mice compared to the control group, which enhanced their survival rate. Our finding highlights the immunoregulatory processes mediated by CECs in newborns. Thus, such tightly regulated immune system in newborns/infants may explain one potential mechanism for the asymptomatic or mild COVID-19 infection in this population.


Assuntos
Antígenos CD/imunologia , Células Precursoras Eritroides , Terapia de Imunossupressão , Listeria monocytogenes/imunologia , Listeriose , Receptores da Transferrina/imunologia , Animais , Animais Recém-Nascidos , COVID-19/imunologia , COVID-19/patologia , Células Precursoras Eritroides/imunologia , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/transplante , Feminino , Xenoenxertos , Humanos , Recém-Nascido , Listeriose/imunologia , Listeriose/patologia , Listeriose/terapia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , SARS-CoV-2/imunologia
3.
Acta pediatr. esp ; 75(11/12): 122-126, nov.-dic. 2017. tab
Artigo em Espanhol | IBECS | ID: ibc-170222

RESUMO

El enfermo oncológico es un paciente con alto riesgo de desnutrición. En este tipo de pacientes es prioritario el diseño de un soporte nutricional personalizado y precoz para conseguir una mejor tolerancia al tratamiento, una buena evolución en su enfermedad de base y una mejora de su calidad de vida. La nutrición parenteral queda reservada para cortos periodos en los que surgen complicaciones importantes durante la quimioterapia y la radioterapia (mucositis, enteritis...), pero es esencial en el trasplante de progenitores hematopoyéticos, así como en su complicación más importante, la enfermedad de injerto contra huésped, en que puede prolongarse durante largos periodos de tiempo. Es fundamental el conocimiento de las alteraciones metabólicas que tienen lugar, así como las variaciones en el gasto energético en reposo y la composición corporal para ajustar los aportes de forma segura y eficaz, minimizando las complicaciones (AU)


The oncological patient is at high risk of malnutrition. Early and personalized nutritional support is essential to improve tolerance to chemotherapy and achieve a better outcome and quality of life. Parenteral nutrition is usually reserved for short periods with major complications during chemotherapy and radiotherapy (mucositis, enteritis...) but it becomes essential in hematopoietic stem cell transplantation as well as in graftversushost disease. The knowledge of the metabolic alterations is essential, as well as the variations in resting energy expenditure and body composition to adjust the requierements in a safe and effective way, minimizing complications (AU)


Assuntos
Humanos , Criança , Nutrição Parenteral/métodos , Neoplasias/dietoterapia , Transplante de Células-Tronco Hematopoéticas , Neoplasias/complicações , Células Precursoras Eritroides/transplante , Doença Enxerto-Hospedeiro/dietoterapia , Desnutrição/dietoterapia , Apoio Nutricional/métodos
4.
Dev Dyn ; 246(12): 1001-1014, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28975680

RESUMO

BACKGROUND: Accumulating evidence suggests the origin of juvenile myelomonocytic leukemia (JMML) is closely associated with fetal development. Nevertheless, the contribution of embryonic progenitors to JMML pathogenesis remains unexplored. We hypothesized that expression of JMML-initiating PTPN11 mutations in HSC-independent yolk sac erythromyeloid progenitors (YS EMPs) would result in a mouse model of pediatric myeloproliferative neoplasm (MPN). RESULTS: E9.5 YS EMPs from VavCre+;PTPN11D61Y embryos demonstrated growth hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) and hyperactive RAS-ERK signaling. Mutant EMPs engrafted the spleens of neonatal recipients, but did not cause disease. To assess MPN development during unperturbed hematopoiesis we generated CSF1R-MCM+;PTPN11E76K ;ROSAYFP mice in which oncogene expression was restricted to EMPs. Yellow fluorescent protein-positive progeny of mutant EMPs persisted in tissues one year after birth and demonstrated hyperactive RAS-ERK signaling. Nevertheless, these mice had normal survival and did not demonstrate features of MPN. CONCLUSIONS: YS EMPs expressing mutant PTPN11 demonstrate functional and molecular features of JMML but do not cause disease following transplantation nor following unperturbed development. Developmental Dynamics 246:1001-1014, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Células Precursoras Eritroides/enzimologia , Mutação com Ganho de Função , Leucemia Mielomonocítica Juvenil/enzimologia , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Saco Vitelino/metabolismo , Animais , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/transplante , Leucemia Mielomonocítica Juvenil/embriologia , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Saco Vitelino/patologia
5.
Nat Commun ; 8(1): 423, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28871080

RESUMO

A short half-life in the circulation limits the application of therapeutics such as single-domain antibodies (VHHs). We utilize red blood cells to prolong the circulatory half-life of VHHs. Here we present VHHs against botulinum neurotoxin A (BoNT/A) on the surface of red blood cells by expressing chimeric proteins of VHHs with Glycophorin A or Kell. Mice whose red blood cells carry the chimeric proteins exhibit resistance to 10,000 times the lethal dose (LD50) of BoNT/A, and transfusion of these red blood cells into naive mice affords protection for up to 28 days. We further utilize an improved CD34+ culture system to engineer human red blood cells that express these chimeric proteins. Mice transfused with these red blood cells are resistant to highly lethal doses of BoNT/A. We demonstrate that engineered red blood cells expressing VHHs can provide prolonged prophylactic protection against bacterial toxins without inducing inhibitory immune responses and illustrates the potentially broad translatability of our strategy for therapeutic applications.The therapeutic use of single-chain antibodies (VHHs) is limited by their short half-life in the circulation. Here the authors engineer mouse and human red blood cells to express VHHs against botulinum neurotoxin A (BoNT/A) on their surface and show that an infusion of these cells into mice confers long lasting protection against a high dose of BoNT/A.


Assuntos
Toxinas Botulínicas Tipo A/toxicidade , Eritrócitos/metabolismo , Engenharia Genética , Anticorpos de Domínio Único/genética , Animais , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/genética , Anticorpos Neutralizantes/metabolismo , Toxinas Botulínicas Tipo A/metabolismo , Botulismo/etiologia , Botulismo/terapia , Transfusão de Eritrócitos , Eritrócitos/virologia , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/transplante , Células Precursoras Eritroides/virologia , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Glicoforinas/genética , Glicoforinas/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Retroviridae/genética , Retroviridae/metabolismo , Anticorpos de Domínio Único/administração & dosagem , Anticorpos de Domínio Único/metabolismo
6.
J Vis Exp ; (127)2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28930976

RESUMO

The presence of incomplete chimerism is noted in a large proportion of patients following bone marrow transplant for thalassemia major or sickle cell disease. This observation has tremendous implications, as subsequent therapeutic immunomodulation strategies can improve clinical outcome. Conventionally, polymerase chain reaction-based analysis of short tandem repeats is used to identify chimerism in donor-derived blood cells. However, this method is restricted to nucleated cells and cannot distinguish between dissociated single-cell lineages. We applied the analysis of short tandem repeats to flow cytometric-sorted hematopoietic progenitor cells and compared this with the analysis of short tandem repeats obtained from selected burst-forming unit - erythroid colonies, both collected from the bone marrow. With this method we are able to demonstrate the different proliferation and differentiation of donor cells in the erythroid compartment. This technique is eligible to complete current monitoring of chimerism in the stem cell transplant setting and thus may be applied in future clinical studies, stem cell research and design of gene therapy trials.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Transplante de Células-Tronco Hematopoéticas/métodos , Hemoglobinopatias/terapia , Quimeras de Transplante/fisiologia , Hemoglobinopatias/patologia , Humanos , Condicionamento Pré-Transplante/métodos
7.
J BUON ; 19(1): 304-14, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24659680

RESUMO

PURPOSE: The purpose of this study was to examine the gene expression profile of granulocyte colony stimulating factor (G-CSF)-mobilized peripheral blood (mPB)-derived progenitors, used in transplantation. METHODS: We correlated gene expression patterns of highly enriched steady-state peripheral blood (PB)- and mPB-derived CD71+ cells by microarray and ingenuity pathway analyses, to identify the transcriptional program during in vitro erythroid differentiation. RESULTS: The gene expression was more than doubled in mPB-derived (4180 genes) compared to PB-derived erythroid progenitors (1667 genes) while PB-and mPB-derived erythroid progenitors shared 1534 common genes. Comparative analysis of transcript levels showed differential expression of 54 genes between cultured erythroid progenitors of PB and mPB origin, where we identified common 13 downregulated and 30 upregulated genes. The most significant genes in mPB-derived erythroid progenitors were P4HB, DDIA3, ARPC2 and ATP5G3. Regarding G-CSF stimulation the G-CSF receptor CSF2RB (1.1-fold) was linked via STAT3 to erythroid-specific ALAS2 (2.9-fold) and GATA2 (1.3-fold) factors, all upregulated in mPB-derived erythroid progenitors, coupled to common upregulated NUDC gene involved in the proliferation of erythroid cells. CONCLUSION: This report provides an extensive transcriptional profile of cultured erythroid progenitors and leads to a better understanding of diversity among the progenitor sources.


Assuntos
Células Precursoras Eritroides/metabolismo , Fator Estimulador de Colônias de Granulócitos/metabolismo , Receptores de Fator Estimulador de Colônias de Granulócitos/metabolismo , Transcriptoma , Diferenciação Celular/genética , Células Cultivadas , Células Precursoras Eritroides/transplante , Redes Reguladoras de Genes/genética , Fator Estimulador de Colônias de Granulócitos/farmacologia , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Análise Serial de Tecidos
8.
Blood Rev ; 27(1): 55-62, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23261066

RESUMO

Donor lymphocyte infusion (DLI) using unstimulated leukapheresis is one of the most effective treatment strategies for patients with hematological malignancies; its graft-versus-leukemia effects make it especially effective in chronic myeloid leukemia patients who relapsed after allogeneic stem cell transplantation (allo-HSCT). However, DLI application is limited by the development of graft-versus-host disease and aplasia, and thus cannot be routinely applied for prophylaxis. Therefore, important questions remain to be answered, such as when, and whom to DLI? Recent advances enable DLI using allografts of granulocyte colony-stimulating factor-mobilized peripheral blood progenitor cells; allodepleted donor T cells; and infusions of donor-derived, ex vivo-expanded, CD8(+) cytotoxic T lymphocyte, which can decrease relapse and improve transplant outcomes. Preemptive immunotherapy of relapse was also introduced based on the determination of mixed chimerism and minimal residual disease. In this review, we summarize the latest developments in recent strategies that will affect future DLI efficacy - focusing on the disadvantages and advantages of each protocol for the treatment, preemptive therapy, and prophylaxis of relapse.


Assuntos
Células Precursoras Eritroides/transplante , Doença Enxerto-Hospedeiro/terapia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas , Imunização Passiva , Transfusão de Linfócitos , Linfócitos T/transplante , Quimerismo , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/imunologia , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/mortalidade , Doença Enxerto-Hospedeiro/patologia , Fator Estimulador de Colônias de Granulócitos/farmacologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/mortalidade , Neoplasias Hematológicas/patologia , Mobilização de Células-Tronco Hematopoéticas , Humanos , Seleção de Pacientes , Recidiva , Análise de Sobrevida , Linfócitos T/imunologia , Transplante Homólogo
9.
Int J Hematol ; 93(1): 5-9, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21184289

RESUMO

The supply of transfusable red blood cells (RBCs) is not sufficient in many countries. If immortalized erythroid progenitor cell lines able to produce transfusable RBCs in vitro were established, they would be valuable resources. However, such cell lines have not been established. We have developed a robust method to establish immortalized erythroid progenitor cell lines following the induction of hematopoietic differentiation of mouse embryonic stem (ES) cells and have established many immortalized erythroid progenitor cell lines so far. Although their precise characteristics varied among cell lines, each of these lines could differentiate in vitro into more mature erythroid cells, including enucleated RBCs. Following transplantation of these erythroid cells into mice suffering from acute anemia, the cells proliferated transiently, subsequently differentiated into functional RBCs, and significantly ameliorated the acute anemia. Considering the number of human ES cell lines that have been established so far and the number of induced pluripotent stem cell lines that will be established in future, the intensive testing of a number of these lines for establishing immortalized erythroid progenitor cell lines may allow the establishment of such cell lines similar to the mouse erythroid progenitor cell lines.


Assuntos
Células-Tronco Embrionárias/metabolismo , Eritrócitos/citologia , Eritrócitos/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoese/fisiologia , Células-Tronco Pluripotentes/metabolismo , Doença Aguda , Anemia/metabolismo , Anemia/terapia , Animais , Linhagem Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/transplante , Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/transplante , Transplante de Células-Tronco
11.
J Mol Cell Cardiol ; 49(3): 347-53, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20382155

RESUMO

Angiogenesis therapy by bone marrow-mononuclear cell implantation (BMI) has been utilized. We found that erythroid cells played an essential role in angiogenesis by BMI. We then tried to establish a novel cell therapy by implantation of ex vivo expanded immature erythroblasts cultured from hematopoietic stem/precursor cells. Immature to mature erythroblasts were purified from human bone marrow, and mRNA expression were analyzed. Strongly expressed VEGF and PLGF in immature erythroid cells decreased according to erythroid maturation. To expand very immature erythroid cells, we established a two-step culturing system, i.e., bone marrow cells were cultured in the presence of Flt-3L, SCF and TPO for 7 days, and the cells were further cultured in the presence of SCF, IGF-I and EPO for an additional 7 days. The in vivo angiogenic effects of implantation of the ex vivo expanded cells were stronger than that of BMI in mouse limb ischemia model. Three patients with severe chronic lower limb ischemia accompanied by Burger's disease or collagen arteritis were enrolled in a pilot clinical trial of the novel cell therapy by transplantation of ex-vivo expanded immature erythroid cells. In the clinical trial, most clinical symptoms such as rest pain and skin ulcers improved in 4 weeks, and did not recur in the one-year follow-up. No adverse events were observed in any of the patients. Moreover this novel cell therapy required only a small amount of bone marrow collection. Further enrollment of patients with chronic severe lower limb ischemia is necessary to confirm the efficacy and safety of this novel cell therapy, and to estimate the necessary amount of bone marrow aspirate.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Membro Posterior/irrigação sanguínea , Isquemia/terapia , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Medula Óssea/patologia , Técnicas de Cultura de Células , Diferenciação Celular , Células Cultivadas , Doença Crônica , Estudos de Viabilidade , Feminino , Membro Posterior/cirurgia , Humanos , Isquemia/patologia , Lúpus Eritematoso Sistêmico/patologia , Lúpus Eritematoso Sistêmico/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Pessoa de Meia-Idade , Neovascularização Fisiológica , Fator de Crescimento Placentário , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tromboangiite Obliterante/patologia , Tromboangiite Obliterante/terapia , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
12.
Blood ; 115(10): 1976-84, 2010 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-20053758

RESUMO

Leukemia-initiating cells (LICs) in acute myeloid leukemia (AML) are believed to be restricted to the CD34(+) fraction. However, one of the most frequently mutated genes in AML is nucleophosmin (NPM), and this is associated with low CD34 expression. We, therefore, investigated whether NPM-mutated AMLs have LICs restricted to the CD34(+) fraction. We transplanted sorted fractions of primary NPM-mutated AML into immunodeficient mice to establish which fractions initiate leukemia. Approximately one-half of cases had LICs exclusively within the CD34(-) fraction, whereas the CD34(+) fraction contained normal multilineage hematopoietic repopulating cells. Most of the remaining cases had LICs in both CD34(+) and CD34(-) fractions. When samples were sorted based on CD34 and CD38 expression, multiple fractions initiated leukemia in primary and secondary recipients. The data indicate that the phenotype of LICs is more heterogeneous than previously realized and can vary even within a single sample. This feature of LICs may make them particularly difficult to eradicate using therapies targeted against surface antigens.


Assuntos
Antígenos CD34/metabolismo , Leucemia Mieloide Aguda/patologia , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/genética , ADP-Ribosil Ciclase 1/metabolismo , Animais , Separação Celular/métodos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/transplante , Humanos , Imunoterapia Adotiva/métodos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/terapia , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Proteínas Mutantes/metabolismo , Células-Tronco Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , Nucleofosmina , Fenótipo , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Am J Physiol Heart Circ Physiol ; 292(1): H657-65, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16997894

RESUMO

Recent reports have demonstrated that erythroid progenitor cells contain and secrete various angiogenic cytokines. Here, the impact of erythroid colony-forming cell (ECFC) implantation on therapeutic angiogenesis was investigated in murine models of hindlimb ischemia. During the in vitro differentiation, vascular endothelial growth factor (VEGF) secretion by ECFCs was observed from day 3 (burst-forming unit erythroid cells) to day 10 (erythroblasts). ECFCs from day 5 to day 7 (colony-forming unit erythroid cells) showed the highest VEGF productivity, and day 6 ECFCs were used for the experiments. ECFCs contained larger amounts of VEGF and fibroblast growth factor-2 (FGF-2) than peripheral blood mononuclear cells (PBMNCs). In tubule formation assays with human umbilical vein endothelial cells, ECFCs stimulated 1.5-fold more capillary growth than PBMNCs, and this effect was suppressed by antibodies against VEGF and FGF-2. Using an immunodeficient hindlimb ischemia model and laser-Doppler imaging, we evaluated the limb salvage rate and blood perfusion after intramuscular implantation of ECFCs. ECFC implantation increased both the salvage rate (38% vs. 0%, P < 0.05) and the blood perfusion (82.8% vs. 65.6%, P < 0.01). In addition, ECFCs implantation also significantly increased capillaries with recruitment of vascular smooth muscle cells and the capillary density was 1.6-fold higher than in the control group. Continuous production of human VEGF from ECFCs in the skeletal muscle was confirmed at least 7 days after the implantation. Implantation of ECFCs promoted angiogenesis in ischemic limbs by supplying angiogenic cytokines (VEGF and FGF-2), suggesting a possible novel strategy for therapeutic angiogenesis.


Assuntos
Células Precursoras Eritroides/citologia , Células Precursoras Eritroides/transplante , Membro Posterior/irrigação sanguínea , Membro Posterior/cirurgia , Isquemia/patologia , Isquemia/cirurgia , Neovascularização Fisiológica , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Animais , Estudos de Viabilidade , Camundongos , Camundongos Nus , Ratos , Ratos Nus , Resultado do Tratamento
14.
Transfusion ; 46(10): 1795-802, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17002637

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) have been identified among hematopoietic tissue-derived progenitor cells that are mobilized into the peripheral blood (PB) as a result of tissue injury. It therefore seems likely that circulating EPCs have therapeutic potential by aiding in the neovascularization of ischemic tissue. This study provides clinical data on the availability of circulating EPCs at steady state and after recombinant human granulocyte-colony-stimulating factor (rHuG-CSF) mobilization and their collection by leukapheresis. STUDY DESIGN AND METHODS: Eight healthy donors underwent rHuG-CSF treatment over 4 days, followed by leukapheresis. Blood samples taken before rHuG-CSF treatment and before apheresis as well as apheresis-collected samples were analyzed by flow cytometry and by real time reverse transcription-polymerase chain reaction for cells expressing EPC-specific surface markers and tissue markers, respectively, and for EPC colony-forming cells. RESULTS: The median PB concentration of CD34+133+ vascular endothelial growth factor receptor-2 (VEGFR-2)-+ EPCs increased 8-fold from steady state to mobilized, and the concentration of CD34+133-VEGFR-2+ EPCs increased by 10-fold. This mobilization pattern was similar to that of hematopoietic CD34+, CD133+, and CD34+117+ progenitor cells. The increase in the median circulating colony-forming unit EPC concentration was 10-fold over baseline. The median absolute number of CD34+133+VEGFR-2+ cells collected by large-volume leukapheresis was 0.8 x 10(6) per kg of body weight. In addition, a small subset of immature CD133+34- cells coexpressing VEGFR-2 was identified in mobilized PB and in the apheresis collection. EPC-specific cells contained in the apheresis product were also identified as expressing mRNA for the CD31 antigen, Tie-2, and VEGFR-2. CONCLUSION: Circulating EPCs represent a novel blood cell component that can be collected by apheresis in large quantities and can be used clinically, either unmanipulated or EPC-selected, for therapeutic vasculogenesis.


Assuntos
Células Sanguíneas/citologia , Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Mobilização de Células-Tronco Hematopoéticas , Neovascularização Fisiológica , Adulto , Idoso , Antígenos de Diferenciação/análise , Células Endoteliais/transplante , Células Precursoras Eritroides/transplante , Feminino , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Isquemia/terapia , Leucaférese/métodos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes , Transplante de Células-Tronco
15.
Blood ; 108(4): 1230-3, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16609068

RESUMO

The laminins are a group of extracellular matrix proteins with constitutive expression in all tissues, including bone marrow stroma. A functional role for the nonintegrin laminin receptor p67 has been described for cancer metastasis and lymphocyte trafficking. Expression of p67 was also reported for other subsets of mature leukocytes and for malignant hematopoietic or nonhematopoietic cells. We explored p67 expression on normal hematopoietic progenitor cells (HPCs) and its putative role in bone marrow retention of transplanted HPCs. We found p67 expression on a subset of primary human CD34(+) cells coexpressing erythroid markers. Of importance, p67 recognizes early erythroid progenitors, since sorted p67(+) cells were significantly enriched for burst-forming units-erythroid (BFU-Es) and depleted of colony-forming units--granulocyte/macrophage (CFU-GMs). Blockade of p67 binding of donor cells, using antifunctional antibody, reduced bone marrow homing of BFU-Es. These studies identify p67 as a novel phenotypic marker for erythroid HPCs of functional importance for lineage-specific homing/retention among adult transplanted HPCs.


Assuntos
Antígenos CD34/biossíntese , Células Precursoras Eritroides/metabolismo , Receptores de Laminina/biossíntese , Adulto , Animais , Anticorpos/farmacologia , Medula Óssea/metabolismo , Células Cultivadas , Células Precursoras Eritroides/transplante , Citometria de Fluxo , Regulação da Expressão Gênica , Células Precursoras de Granulócitos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Neoplasias/metabolismo , Receptores de Laminina/antagonistas & inibidores , Células Estromais/metabolismo
16.
Bone Marrow Transplant ; 34(12): 1089-93, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15489877

RESUMO

To evaluate the impact of ex vivo expanded megakaryocyte (MK) progenitors on high-dose chemotherapy-induced thrombocytopenia, we conducted a phase II study in 10 patients with relapsed lymphoma. Two fractions of peripheral blood progenitor cells (PBPC) were cryopreserved, one with enough cells for at least 2 x 10(6) CD34+ cells/kg and a second obtained after CD34+ selection. Ten days before autologous stem cell transplantation, the CD34+ fraction was cultured with MGDF+SCF for 10 days. After BEAM (BCNU, cyclophosphamide, cytarabine, and melphalan) chemotherapy, patients were reinfused with standard PBPC and ex vivo expanded cells. No toxicity was observed after reinfusion. The mean fold expansion was 9.27 for nucleated cells, 2 for CD34+ cells, 676 for CD41+ cells, and 627 for CD61+ cells. The median date of platelet transfusion independence was day 8 (range: 7-12). All patients received at least one platelet transfusion. In conclusion, ex vivo expansion of MK progenitors was feasible and safe, but this procedure did not prevent BEAM-induced thrombocytopenia. Future studies will determine if expansion of higher numbers of CD34+ cells towards the MK-differentiation pathway will translate into a functional effect in terms of shortening of BEAM-induced thrombocytopenia.


Assuntos
Células Precursoras Eritroides/citologia , Megacariócitos/citologia , Transplante de Células-Tronco de Sangue Periférico/métodos , Antígenos CD34/análise , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Carmustina/administração & dosagem , Técnicas de Cultura de Células/métodos , Células Cultivadas , Ciclofosfamida/administração & dosagem , Citarabina/administração & dosagem , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/transplante , Humanos , Integrina beta3/análise , Linfoma/complicações , Linfoma/terapia , Megacariócitos/transplante , Melfalan/administração & dosagem , Glicoproteína IIb da Membrana de Plaquetas/análise , Transfusão de Plaquetas , Terapia de Salvação , Trombocitopenia/induzido quimicamente , Trombocitopenia/prevenção & controle , Trombopoetina/farmacologia , Transplante Autólogo , Resultado do Tratamento
17.
Blood ; 104(12): 3445-53, 2004 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-15292064

RESUMO

beta-thalassemias are the most common single gene disorders and are potentially amenable to gene therapy. However, retroviral vectors carrying the human beta-globin cassette have been notoriously unstable. Recently, considerable progress has been made using lentiviral vectors, which stably transmit the beta-globin expression cassette. Thus far, mouse studies have shown correction of the beta-thalassemia intermedia phenotype and a partial, variable correction of beta-thalassemia major phenotype. We tested a lentiviral vector carrying the human beta-globin expression cassette flanked by a chromatin insulator in transfusion-dependent human thalassemia major, where it would be ultimately relevant. We demonstrated that the vector expressed normal amounts of human beta-globin in erythroid cells produced in in vitro cultures for unilineage erythroid differentiation. There was restoration of effective erythropoiesis and reversal of the abnormally elevated apoptosis that characterizes beta-thalassemia. The gene-corrected human beta-thalassemia progenitor cells were transplanted into immune-deficient mice, where they underwent normal erythroid differentiation, expressed normal levels of human beta-globin, and displayed normal effective erythropoiesis 3 to 4 months after xenotransplantation. Variability of beta-globin expression in erythroid colonies derived in vitro or from xenograft bone marrow was similar to that seen in normal controls. Our results show genetic modification of primitive progenitor cells with correction of the human thalassemia major phenotype.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Globinas/administração & dosagem , Talassemia beta/terapia , Animais , Diferenciação Celular , Sobrevivência Celular , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/transplante , Eritropoese , Globinas/genética , Humanos , Lentivirus/genética , Camundongos , Camundongos SCID , Fenótipo , Transplante Heterólogo , Resultado do Tratamento
18.
Development ; 130(18): 4393-403, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12900455

RESUMO

The platelet glycoprotein IIb (alpha(IIb); CD41) constitutes the alpha subunit of a highly expressed platelet surface integrin protein. We demonstrate that CD41 serves as the earliest marker of primitive erythroid progenitor cells in the embryonic day 7 (E7.0) yolk sac and high-level expression identifies essentially all E8.25 yolk sac definitive hematopoietic progenitors. Some definitive hematopoietic progenitor cells in the fetal liver and bone marrow also express CD41. Hematopoietic stem cell competitive repopulating ability is present in CD41(dim) and CD41(lo/-) cells isolated from bone marrow and fetal liver cells, however, activity is enriched in the CD41(lo/-) cells. CD41(bright) yolk sac definitive progenitor cells co-express CD61 and bind fibrinogen, demonstrating receptor function. Thus, CD41 expression marks the onset of primitive and definitive hematopoiesis in the murine embryo and persists as a marker of some stem and progenitor cell populations in the fetal liver and adult marrow, suggesting novel roles for this integrin.


Assuntos
Embrião de Mamíferos/fisiologia , Células Precursoras Eritroides/metabolismo , Hematopoese/fisiologia , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Animais , Linfócitos B/metabolismo , Biomarcadores , Células da Medula Óssea/metabolismo , Linhagem da Célula , Transplante de Células , Embrião de Mamíferos/anatomia & histologia , Células Precursoras Eritroides/transplante , Feminino , Fibrinogênio/metabolismo , Granulócitos/metabolismo , Hemoglobinas/genética , Hemoglobinas/metabolismo , Integrina beta3/metabolismo , Antígenos Comuns de Leucócito/metabolismo , Fígado/citologia , Fígado/embriologia , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Linfócitos T/metabolismo , Quimeras de Transplante , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Saco Vitelino/citologia
19.
Spinal Cord ; 39(11): 609-13, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11641813

RESUMO

The World Health Organization together with the Iceland Ministry of Health and Social Security sponsored a conference entitled 'Human Spinal Cord Injury: New and Emerging Approaches to Treatment' held on May 31-June 2, 2001 in Reykjavik, Iceland. To help catalyze the development of new paradigms to address spinal cord injury, the conference's overall goal was to bring in a diversity of perspectives, ranging from state-of-the-art stem cell biology to the ancient wisdom of Eastern Medicine. The purpose of this paper is to summarize the presentations of the conference's 26 speakers.


Assuntos
Traumatismos da Medula Espinal/reabilitação , Terapias Complementares/métodos , Terapias Complementares/tendências , Terapia por Estimulação Elétrica/métodos , Terapia por Estimulação Elétrica/tendências , Células Precursoras Eritroides/transplante , Humanos , Aparelhos Ortopédicos/tendências , Nervos Periféricos/transplante , Traumatismos da Medula Espinal/cirurgia
20.
Cytotherapy ; 3(1): 5-10, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12028838

RESUMO

BACKGROUND: In recent years there have been clear improvements in the procurement of peripheral blood progenitor cells (PBPC) for autologous transplantation, such as the description of more effective mobilization regimens and the use of CD34 monitoring to determine the appropriate time to start collection. However, currently there is no accurate method of predicting the volume of blood required to be processed by apheresis to yield the target number of CD34(+) progenitor cells. METHODS: This study was performed to determine whether there is a correlation between the harvested number of CD34(+) cells per kilogram body weight and the 'CD34 prediction score' calculated from the concentration of CD34(+) cells in the blood prior to harvest, the blood volume processed, and the patient's weight. RESULTS: A strong correlation between the CD34 prediction score and the quantity of CD34(+) cells harvested was found. This facilitated the construction of an algorithm for calculating the minimum volume of blood required to be processed by apheresis to yield the target number of CD34(+) cells. Subsequent validation of the algorithm facilitated successful tailoring of the apheresis time. DISCUSSION: The ability to accurately calculate the minimum volume of blood to be processed by apheresis to yield a target number of PBPC produces significant benefits in patient management, cost savings and equipment utilization.


Assuntos
Antígenos CD34/análise , Contagem de Células Sanguíneas , Remoção de Componentes Sanguíneos/métodos , Volume Sanguíneo , Células Precursoras Eritroides/transplante , Mobilização de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/métodos , Adolescente , Adulto , Algoritmos , Remoção de Componentes Sanguíneos/economia , Peso Corporal , Redução de Custos , Feminino , Transplante de Células-Tronco Hematopoéticas/economia , Humanos , Modelos Lineares , Masculino , Pessoa de Meia-Idade , Sensibilidade e Especificidade , Fatores de Tempo , Transplante Autólogo/economia , Transplante Autólogo/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...