Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Rev Rep ; 20(1): 247-257, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37861968

RESUMO

Ischemic heart attack is the leading cause of death worldwide. Ten percent of cases will die within an hour. Of the survivors, around 30% will have suffered a severe infarction which will lead to the irreparable destruction of 1 to 2 billion myocardial cells, causing an irreversible secondary heart failure with a poor prognosis in the short. The heart is a totally differentiated organ with a very low capacity for self-regeneration. No current treatment can prevent this fatal outcome, but only slow it down. For these reasons, cell therapy has generated enormous hope, but achieved somewhat disappointing results, depending on the type/source of cells which were used. From the end of 2002, our group conducted a Pilot study using immuno-selected autologous peripheral-blood (PB) CD34+ cells in a small cohort of patients who had experienced a heart attack with poor prognosis. Three of these patients were immediately considered for heart transplant but lacked a readily available donor. CD34+ cells were trans-epicardially delivered at the end of a coronary artery by-pass graft (CABG) operation without reperfusing the ischemic area, which was performed on a compassionate basis. All but one patient showed a marked and sustained improvement in their cardiac function parameters from the baseline values, associated with both cardiac tissue repair and revascularization, as demonstrated by PetScan examination. The patients' outcomes have been recently updated. Six out of seven patients have survived in good enough conditions for at least 12 years after cell therapy, including those three initially recommended for heart transplant and who have avoided it. Presently, five out of seven patients are still alive with an average follow-up of 17 years (range 16-20 years), which is very unusual after CABG for patients with such a poor initially prognosis.


Assuntos
Coração , Infarto do Miocárdio , Humanos , Projetos Piloto , Resultado do Tratamento , Infarto do Miocárdio/terapia , Células Sanguíneas
2.
Sci Rep ; 13(1): 19665, 2023 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-37952030

RESUMO

We have previously shown that intracardiac delivery of autologous CD34+ cells after acute myocardial infarction (AMI) is safe and leads to long term improvement. We are now conducting a multicenter, randomized, controlled Phase I/IIb study in post-AMI to investigate the safety and efficacy of intramyocardial injection of expanded autologous CD34+ cells (ProtheraCytes) (NCT02669810). Here, we conducted a series of in vitro studies characterizing the growth factor secretion, exosome secretion, gene expression, cell surface markers, differentiation potential, and angiogenic potential of ProtheraCytes clinical batches to develop a potency assay. We show that ProtheraCytes secrete vascular endothelial growth factor (VEGF) and its concentration is significantly correlated with the number of CD34+ cells obtained after expansion. ProtheraCytes also secrete exosomes containing proangiogenic miRNAs (126, 130a, 378, 26a), antiapoptotic miRNAs (21 and 146a), antifibrotic miRNAs (133a, 24, 29b, 132), and miRNAs promoting myocardial regeneration (199a and 590). We also show that ProtheraCytes have in vitro angiogenic activity, express surface markers of endothelial progenitor cells, and can differentiate in vitro into endothelial cells. After the in vitro characterization of multiple ProtheraCytes clinical batches, we established that measuring the concentration of VEGF provided the most practical, reliable, and consistent potency assay.


Assuntos
Células Progenitoras Endoteliais , MicroRNAs , Infarto do Miocárdio , Humanos , Antígenos CD34/metabolismo , Células Progenitoras Endoteliais/metabolismo , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Neovascularização Fisiológica , Fator A de Crescimento do Endotélio Vascular/metabolismo
3.
Front Bioeng Biotechnol ; 11: 1150522, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37288358

RESUMO

Knee osteoarthritis (OA) is a degenerative joint disease of the knee that results from the progressive loss of articular cartilage. It is most common in the elderly and affects millions of people worldwide, leading to a continuous increase in the number of total knee replacement surgeries. These surgeries improve the patient's physical mobility, but can lead to late infection, loosening of the prosthesis, and persistent pain. We would like to investigate if cell-based therapies can avoid or delay such surgeries in patients with moderate OA by injecting expanded autologous peripheral blood derived CD34+ cells (ProtheraCytes®) into the articular joint. In this study we evaluated the survival of ProtheraCytes® when exposed to synovial fluid and their performance in vitro with a model consisting of their co-culture with human OA chondrocytes in separate layers of Transwells and in vivo with a murine model of OA. Here we show that ProtheraCytes® maintain high viability (>95%) when exposed for up to 96 hours to synovial fluid from OA patients. Additionally, when co-cultured with OA chondrocytes, ProtheraCytes® can modulate the expression of some chondrogenic (collagen II and Sox9) and inflammatory/degrading (IL1ß, TNF, and MMP-13) markers at gene or protein levels. Finally, ProtheraCytes® survive after injection into the knee of a collagenase-induced osteoarthritis mouse model, engrafting mainly in the synovial membrane, probably due to the fact that ProtheraCytes® express CD44, a receptor of hyaluronic acid, which is abundantly present in the synovial membrane. This report provides preliminary evidence of the therapeutic potential of CD34+ cells on OA chondrocytes in vitro and their survival after in vivo implantation in the knee of mice and merits further investigation in future preclinical studies in OA models.

4.
Int J Mol Sci ; 24(11)2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-37298503

RESUMO

Ex vivo monitored human CD34+ stem cells (SCs) injected into myocardium scar tissue have shown real benefits for the recovery of patients with myocardial infarctions. They have been used previously in clinical trials with hopeful results and are expected to be promising for cardiac regenerative medicine following severe acute myocardial infarctions. However, some debates on their potential efficacy in cardiac regenerative therapies remain to be clarified. To elucidate the levels of CD34+ SC implication and contribution in cardiac regeneration, better identification of the main regulators, pathways, and genes involved in their potential cardiovascular differentiation and paracrine secretion needs to be determined. We first developed a protocol thought to commit human CD34+ SCs purified from cord blood toward an early cardiovascular lineage. Then, by using a microarray-based approach, we followed their gene expression during differentiation. We compared the transcriptome of undifferentiated CD34+ cells to those induced at two stages of differentiation (i.e., day three and day fourteen), with human cardiomyocyte progenitor cells (CMPCs), as well as cardiomyocytes as controls. Interestingly, in the treated cells, we observed an increase in the expressions of the main regulators usually present in cardiovascular cells. We identified cell surface markers of the cardiac mesoderm, such as kinase insert domain receptor (KDR) and the cardiogenic surface receptor Frizzled 4 (FZD4), induced in the differentiated cells in comparison to undifferentiated CD34+ cells. The Wnt and TGF-ß pathways appeared to be involved in this activation. This study underlined the real capacity of effectively stimulated CD34+ SCs to express cardiac markers and, once induced, allowed the identification of markers that are known to be involved in vascular and early cardiogenesis, demonstrating their potential priming towards cardiovascular cells. These findings could complement their paracrine positive effects known in cell therapy for heart disease and may help improve the efficacy and safety of using ex vivo expanded CD34+ SCs.


Assuntos
Infarto do Miocárdio , Células-Tronco , Humanos , Células-Tronco/metabolismo , Miocárdio/metabolismo , Antígenos CD34/metabolismo , Miócitos Cardíacos/metabolismo , Infarto do Miocárdio/metabolismo , Diferenciação Celular/fisiologia , Moléculas de Adesão Celular/metabolismo , Receptores Frizzled/metabolismo
5.
Stem Cell Rev Rep ; 18(5): 1614-1626, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35420389

RESUMO

Regenerative medicine now needs to pass a crucial turning point, from academic research to the market. Several sources/types of cells have been experimented with, more or less successfully. CD34+ cells have demonstrated multipotent or even pluripotent capacities, making them good candidates for regenerative medicine, particularly for treating heart diseases. Strongly encouraged by the results we achieved in a pilot study using CD34+ stem cells in patients with poor-prognosis acute myocardial infarcts (AMIs), we soon began the development of an industrialized platform making use of a closed automated device (StemXpand®) and a disposable kit (StemPack®) for the large-scale expansion of CD34+ cells with reproducible good manufacturing practice (GMP). This scalable platform can produce expanded CD34+ cells (ProtheraCytes®) of sufficient quality that, interestingly, express early markers of the cardiac and endothelial pathways and early cardiac-mesoderm markers. They also contain CD34+ pluripotent cells characterized as very small embryonic-like stem cells (VSELs), capable of differentiating under appropriate stimuli into different tissue lineages, including endothelial and cardiomyocytic ones.


Assuntos
Infarto do Miocárdio , Isquemia Miocárdica , Antígenos CD34/metabolismo , Células-Tronco Embrionárias/metabolismo , Humanos , Infarto do Miocárdio/terapia , Projetos Piloto , Medicina Regenerativa/métodos
6.
Biology (Basel) ; 10(4)2021 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-33918035

RESUMO

CD9 plays a crucial role in cellular growth, mobility, and signal transduction, as well as in hematological malignancy. In myeloid neoplasms, CD9 is involved in the altered interactions between leukemic and stromal cells. However, apart from its role in CD34+ progenitors and myeloid and megakaryocytic differentiation, its function in normal and leukemic pluripotent cells has not yet been determined. Very small embryonic-like stem cells (VSELs) are promising pluripotent stem cells found in adult tissues that can be developed for safe and efficient regenerative medicine. VSELs express different surface receptors of the highest importance in cell functioning, including CD9, and can be effectively mobilized after organ injury or in leukemic patients. In the present study, we observed that CD9 is among the most expressed receptors in VSELs under steady-state conditions; however, once the VSELs are expanded, CD9+ VSELs decrease and are more apoptotic. CD9- VSELs had no proliferative improvement in vitro compared to those that were CD9+. Interestingly, the addition of SDF-1 induced CD9 expression on the surface of VSELs, as observed by flow cytometry, and improved their migration. In addition, we observed, in the phenotypically identical VSELs present in the peripheral blood of patients with myeloproliferative neoplasms, compared to healthy subjects, a significantly higher number of CD9+ cells. However, in their hematopoietic stem cell (HSC) counterparts, the expression remained comparable. These results indicate that, likewise, in progenitors and mature cells, CD9 may play an important function in normal and malignant VSELs. This could explain the refractoriness observed by some groups of expanded stem cells to repairing efficiently damaged tissue when used as a source in cell therapies. Understanding the function of the CD9 receptor in normal and malignant CD34+ and VSELs, along with its relationship with the CXCR4/SDF-1 pathway, will enable advances in the field of adult pluripotent cell usage in regenerative medicine and in their role in leukemia.

7.
Stem Cell Rev Rep ; 16(3): 441-458, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32297205

RESUMO

Stem cell therapy offers a breakthrough opportunity for the improvement of ischemic heart diseases. Numerous clinical trials and meta-analyses appear to confirm its positive but variable effects on heart function. Whereas these trials widely differed in design, cell type, source, and doses reinjected, cell injection route and timing, and type of cardiac disease, crucial key factors that may favour the success of cell therapy emerge from the review of their data. Various types of cell have been delivered. Injection of myoblasts does not improve heart function and is often responsible for severe ventricular arrythmia occurrence. Using bone marrow mononuclear cells is a misconception, as they are not stem cells but mainly a mix of various cells of hematopoietic lineages and stromal cells, only containing very low numbers of cells that have stem cell-like features; this likely explain the neutral results or at best the modest improvement in heart function reported after their injection. The true existence of cardiac stem cells now appears to be highly discredited, at least in adults. Mesenchymal stem cells do not repair the damaged myocardial tissue but attenuate post-infarction remodelling and contribute to revascularization of the hibernated zone surrounding the scar. CD34+ stem cells - likely issued from pluripotent very small embryonic-like (VSEL) stem cells - emerge as the most convincing cell type, inducing structural and functional repair of the ischemic myocardial area, providing they can be delivered in large amounts via intra-myocardial rather than intra-coronary injection, and preferentially after myocardial infarct rather than chronic heart failure.


Assuntos
Cardiopatias/terapia , Medicina Regenerativa , Animais , Terapia Baseada em Transplante de Células e Tecidos , Ensaios Clínicos como Assunto , Vias de Administração de Medicamentos , Humanos , Índice de Gravidade de Doença
8.
Stem Cells Transl Med ; 8(8): 822-832, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31037857

RESUMO

We previously demonstrated that intracardiac delivery of autologous peripheral blood-derived CD34+ stem cells (SCs), mobilized by granulocyte-colony stimulating factor (G-CSF) and collected by leukapheresis after myocardial infarction, structurally and functionally repaired the damaged myocardial area. When used for cardiac indication, CD34+ cells are now considered as Advanced Therapy Medicinal Products (ATMPs). We have industrialized their production by developing an automated device for ex vivo CD34+ -SC expansion, starting from a whole blood (WB) sample. Blood samples were collected from healthy donors after G-CSF mobilization. Manufacturing procedures included: (a) isolation of total nuclear cells, (b) CD34+ immunoselection, (c) expansion and cell culture recovery in the device, and (d) expanded CD34+ cell immunoselection and formulation. The assessment of CD34+ cell counts, viability, and immunophenotype and sterility tests were performed as quality tests. We established graft acceptance criteria and performed validation processes in three cell therapy centers. 59.4 × 106 ± 36.8 × 106 viable CD34+ cells were reproducibly generated as the final product from 220 ml WB containing 17.1 × 106 ± 8.1 × 106 viable CD34+ cells. CD34+ identity, genetic stability, and telomere length were consistent with those of basal CD34+ cells. Gram staining and mycoplasma and endotoxin analyses were negative in all cases. We confirmed the therapeutic efficacy of both CD34+ -cell categories in experimental acute myocardial infarct (AMI) in immunodeficient rats during preclinical studies. This reproducible, automated, and standardized expansion process produces high numbers of CD34+ cells corresponding to the approved ATMP and paves the way for a phase I/IIb study in AMI, which is currently recruiting patients. Stem Cells Translational Medicine 2019;8:822&832.


Assuntos
Antígenos CD34/genética , Automação Laboratorial/métodos , Citometria de Fluxo/métodos , Infarto do Miocárdio/terapia , Transplante de Células-Tronco de Sangue Periférico/métodos , Células-Tronco de Sangue Periférico/citologia , Adulto , Animais , Antígenos CD34/metabolismo , Células Cultivadas , Ensaios Clínicos como Assunto , Humanos , Imunofenotipagem/métodos , Masculino , Pessoa de Meia-Idade , Células-Tronco de Sangue Periférico/metabolismo , Cultura Primária de Células/métodos , Ratos
9.
Stem Cell Rev Rep ; 14(4): 510-524, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29736843

RESUMO

The very small embryonic-like stem cells (VSELs) are known as a subset of adult pluripotent stem cells able to differentiate to all three germ layers. However, their small number and quiescence restrict the possibility of their use in cell therapy. In the present study, we first delineate different subpopulation of VSELs from human cord blood CD34+ cells to define their purity. We next determine genes expression levels in the whole transcriptome of VSELs expressing the pluripotent marker NANOG and control cells under the steady state condition. We found that more than a thousand of genes are downregulated in VSELs, as well as many membrane receptors, cells signaling molecules and CDKs mRNAs. In addition, we observed discordance in some pluripotent genes expression levels with embryonic stem cells (ESCs), which could explain VSELs quiescence. We then evaluate VSELs capacity to expand and differentiate in vitro in specific and appropriate media. After 12 days culture in specific medium containing a pyrimidoindole derivative (UM171), VSELs were significantly expanded for the first time without feeder cells and importantly preserve their capacities to differentiate into hematopoietic and endothelial cells. Interestingly, this stimulation of VSELs self-renewal restores the expression of some downregulated genes known as key regulators of cell proliferation and differentiation. The properties of such pluripotent expanded cells make them a potential candidate in regenerative medicine.


Assuntos
Diferenciação Celular/genética , Proliferação de Células/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Adulto , Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Células Cultivadas , Células-Tronco Embrionárias/citologia , Sangue Fetal/citologia , Perfilação da Expressão Gênica , Camadas Germinativas/citologia , Camadas Germinativas/metabolismo , Humanos , Proteína Homeobox Nanog/genética , Células-Tronco Pluripotentes/citologia , Medicina Regenerativa/métodos , Fatores de Transcrição SOXF/genética
10.
Stem Cells Int ; 2016: 7651645, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26633977

RESUMO

The purpose of our study was to determine whether the number of human very small embryonic-like stem cells (huVSELs) would vary depending on the age of humans. HuVSELs frequency was evaluated into the steady-state (SS) peripheral blood (PB) of healthy volunteers using flow cytometry analysis. Their numbers were compared with volunteers' age. Blood samples were withdrawn from 28 volunteers (age ranging from 20 to 70 years), who were distributed among three groups of age: "young" (mean age, 27.8 years), "middle" (mean age, 49 years), and "older" (mean age, 64.2 years). Comparing the three groups, we did not observe any statistically significant difference in huVSELs numbers between them. The difference in mRNA expression for PSC markers as SSEA-4, Oct-4, Nanog, and Sox2 between the three groups of age was not statistically significant. A similar frequency of huVSELs into the SS-PB of young, middle-aged, and aged subjects may indicate that the VSELs pool persists all along the life as a reserve for tissue repair in case of minor injury and that there is a continuous efflux of these cells from the BM into the PB.

11.
Bioorg Med Chem ; 21(7): 2135-44, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23428964

RESUMO

Aminopeptidase-N (APN/CD13) is highly expressed on the surface of numerous types of cancer cells and particularly on the endothelial cells of neoangiogenic vessels during tumourigenesis. This metallo-aminopeptidase has been identified as a potential target for cancer chemotherapy. In this work, we evaluated the efficacy of a novel series of benzosuberone analogues, which were previously reported to be highly potent, selective APN inhibitors with Ki values in the micromolar to sub-nanomolar range. Endothelial cell morphogenesis as well as cell motility were inhibited in vitro in a dose-dependent manner at concentrations that correlated with the potency of the compounds, thus confirming the key role of APN in these established models of angiogenesis. We report toxicity studies in mice showing that these compounds are well tolerated. We report the effects of the compounds, used alone or in combination with rapamycin, on the growth of a select panel of tumours that were subcutaneously xenografted onto Swiss nude mice. Our data indicate that the in vivo efficacy of these new APN inhibitors during the initial phase of tumour growth can be ascribed to their anti-angiogenic activities. However, we also provide evidence that these compounds are effective against established solid tumours. For colonic tumours, the anti-tumour effect depends on the level of APN expression in epithelial cells, and APN expression is associated with down-regulation of the transcription factor HIF-1α. These effects seem to be distinct from those of rapamycin. Our finding that the anti-tumour effect of the inhibitors in the colon requires APN expression strongly suggests that APN plays a crucial function in tumour cells that is distinct from its known role in neovascularisation.


Assuntos
Anisóis/química , Anisóis/uso terapêutico , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Antígenos CD13/antagonistas & inibidores , Cicloeptanos/química , Cicloeptanos/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Inibidores da Angiogênese/uso terapêutico , Animais , Anisóis/farmacologia , Antineoplásicos/farmacologia , Antígenos CD13/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Cicloeptanos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Neoplasias/genética , Neoplasias/patologia
12.
Exp Hematol ; 39(4): 495-505, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21238532

RESUMO

OBJECTIVE: Recently, we demonstrated that normal human bone marrow (hBM)-derived CD34(+) cells, released into the peripheral blood after granulocyte colony-stimulating factor mobilization, contain cell subpopulations committed along endothelial and cardiac differentiation pathways. These subpopulations could play a key role in the regeneration of post-ischemic myocardial lesion after their direct intracardiac delivery. We hypothesized that these relevant cells might be issued from very small embryonic-like stem cells deposited in the BM during ontogenesis and reside lifelong in the adult BM, and that they could be mobilized into peripheral blood by granulocyte colony-stimulating factor. MATERIALS AND METHODS: Samples of normal hBM and leukapheresis products harvested from cancer patients after granulocyte colony-stimulating factor mobilization were analyzed and sorted by multiparameter flow cytometry strategy. Immunofluorescence and reverse transcription quantitative polymerase chain reaction assays were performed to analyze the expression of typical pluripotent stem cells markers. RESULTS: A population of CD34(+)/CD133(+)/CXCR4(+)/Lin(-) CD45(-) immature cells was first isolated from the hBM or from leukapheresis products. Among this population, very small (2-5 µm) cells expressing Oct-4, Nanog, and stage-specific embryonic antigen-4 at protein and messenger RNA levels were identified. CONCLUSIONS: Our study supports the hypothesis that very small embryonic-like stem cells constitute a "mobile" pool of primitive/pluripotent stem cells that could be released from the BM into the peripheral blood under the influence of various physiological or pathological stimuli. In order to fully support that hBM- and leukapheresis product-derived very small embryonic-like stem cells are actually pluripotent, we are currently testing their ability to differentiate in vitro into cells from all three germ layers.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Hematopoéticas/citologia , Antígeno AC133 , Adulto , Idoso , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Células da Medula Óssea/metabolismo , Separação Celular , Células-Tronco Embrionárias/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Glicoproteínas/metabolismo , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Imunofenotipagem , Leucaférese , Masculino , Pessoa de Meia-Idade , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Peptídeos/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Receptores CXCR4/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
13.
Cytotherapy ; 11(8): 1002-15, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19929464

RESUMO

BACKGROUND AIMS: Starting from experimental data proposing hematopoietic stem cells as candidates for cardiac repair, we postulated that human peripheral blood (PB) CD34+ cells mobilized by hematopoietic growth-factor (G-CSF) would contain cell subpopulations capable of regenerating post-ischemic myocardial damages. METHODS: In a phase I clinical assay enrolling seven patients with acute myocardial infarct, we directly delivered to the injured myocardium autologous PB CD34+ cells previously mobilized by G-CSF, collected by leukapheresis and purified by immunoselection. In parallel, we looked for the eventual presence of cardiomyocytic and endothelial progenitor cells in leukapheresis products of these patients and controls, using flow cytometry, reverse transcription-quantitative (RTQ)-polymerase chain reaction (PCR), cell cultures and immunofluorescence analyzes. RESULTS: The whole clinical process was feasible and safe. All patients were alive at an average follow-up of 49 months (range 24-76 months). Improvement of heart function parameters became obvious from the third month following cell reinjection. Left ventricular ejection fraction values progressively and dramatically increased with time, associated with PetScan demonstration of myocardial structure regeneration and revascularization and New York Heart Association (NYHA) grade improvement. Furthermore, we identified PB CD34+ cell subpopulations expressing characteristics of both immature and mature endothelial and cardiomyocyte progenitor cells. In vitro CD34+ cell cultures on a specific medium induced development of adherent cells featuring morphologies, gene expression and immunocytochemistry characteristics of endothelial and cardiac muscle cells. CONCLUSIONS: Mobilized CD34+ cells contain stem cells committed along endothelial and cardiac differentiation pathways, which could play a key role in a proposed two-phase mechanism of myocardial regeneration after direct intracardiac delivery, probably being responsible for the long-term clinical benefit observed.


Assuntos
Antígenos CD34/metabolismo , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Células-Tronco/citologia , Adulto , Idoso , Adesão Celular/efeitos dos fármacos , Desdiferenciação Celular/efeitos dos fármacos , Células Cultivadas , Vias de Administração de Medicamentos , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Feminino , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/patologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Volume Sistólico/efeitos dos fármacos , Volume Sistólico/fisiologia , Fatores de Tempo , Transplante Autólogo , Resultado do Tratamento
14.
Cytometry A ; 73(1): 16-21, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18061957

RESUMO

Circulating immature blood cells are inhomogeneous in most cytometric parameters, including CD34 expression. This surface antigen forms patches, and we studied their spatial distribution pattern by processing staggered and digitalized microscopical images; the number, fluorescence intensity, and volume were determined in randomly identified CD34-positive cord blood cells in suspension. Quantitative fluorescence analysis of individual CD34-labeled cells was performed after acquisition of microscopical images in high resolution by using a CCD camera and adjacent deconvolution. Two major (n = 42 and n = 41) and one minor (n = 5) group of haematopoietic progenitors with different CD34+ distribution patterns were detected. All CD34 antigen patches are localized on the spherical cell membrane, and differ most significantly in fluorescence intensity (P < 10(-4)), antigen volume (P < 0.004), and patch number per cell (P < 0.02). If all parameters are employed, 96.6% of CD34+ cells are correctly classified into one of the three groups as shown by discriminant analysis. If fluorescence intensity is eliminated, recognition efficiency decreases to 68.2%, indicating that this is the most powerful single parameter. While total fluorescence intensity is most characteristic for each group, total patch volume and number per cell, and heterogeneity of patch volumes and their spatial distribution also contribute significantly to reliable classification into their groups.


Assuntos
Antígenos CD34/biossíntese , Técnicas Citológicas , Sangue Fetal/citologia , Medula Óssea/metabolismo , Membrana Celular/metabolismo , Separação Celular , Análise por Conglomerados , Células-Tronco Hematopoéticas/citologia , Humanos , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Imunofenotipagem/métodos
15.
Adv Exp Med Biol ; 534: 27-45, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12903709

RESUMO

Over the past few years, research on animal and human stem cells has experienced tremendous advances which are almost daily loudly revealed to the public on the front-page of newspapers. The reason for such an enthusiasm over stem cells is that they could be used to cure patients suffering from spontaneous or injuries-related diseases that are due to particular types of cells functioning incorrectly, such as cardiomyopathy, diabetes mellitus, osteoporosis, cancers, Parkinson's disease, spinal cord injuries or genetic abnormalities. Currently, these diseases have slightly or non-efficient treatment options, and millions of people around the world are desperately waiting to be cured. Even if not any person with one of these diseases could potentially benefit from stem cell therapy, the new concept of "regenerative medicine" is unprecedented since it involves the regeneration of normal cells, tissues and organs which could allow to treat a patient whereby both, the immediate problem would be corrected and the normal physiological processes restored, without any need for subsequent drugs. However, conflicting ethical controversies surround this new medicine approach, inside and outside the medical community, especially when human embryonic stem cells (h-ESCs) are concerned. This ethical debate on clinical use of h-ESCs has recently encouraged the research on "adult" stem cells (ASCs) regarded as a less conflicting alternative for the future of regenerative medicine.


Assuntos
Bioética , Embrião de Mamíferos/citologia , Células-Tronco/citologia , Engenharia Tecidual , Adulto , Técnicas de Cultura de Células , Humanos , Engenharia Tecidual/ética
16.
Br J Haematol ; 120(3): 496-9, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12580968

RESUMO

Panels of immunological markers are useful in refining diagnosis in view of certain variability between B-cell leukaemias. A statistical multivariate approach was used on 100 B leukaemias (preliminary sample) to explore the potential value of the combination of CD43, and the classical markers CD5, CD23, CD79b, FMC7, CD22 and surface immunoglobulin to differentiate chronic lymphoid leukaemia (CLL) from lymphoma (non-CLL). CD43 was highly effective (P < 0.00001) and its inclusion in the panels improved the accuracy of discrimination in a 'control' sample of 74 B leukaemias to 98.6%. Inclusion of CD43 facilitates the diagnosis of B-lymphoproliferative disorders and improves their classification.


Assuntos
Antígenos CD , Linfócitos B/imunologia , Biomarcadores Tumorais/análise , Leucemia de Células B/diagnóstico , Linfoma de Células B/diagnóstico , Sialoglicoproteínas/análise , Antígenos de Neoplasias/análise , Diagnóstico Diferencial , Humanos , Imunofenotipagem , Leucossialina , Análise Multivariada , Proteínas de Neoplasias/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...