Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 221
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mediators Inflamm ; 2018: 2691934, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30116144

RESUMEN

BACKGROUND: Acute myocardial infarction (AMI) and coronary artery bypass graft (CABG) surgery are associated with a pathogen-free inflammatory response (sterile inflammation). Complement cascade (CC) and bioactive sphingolipids (BS) are postulated to be involved in this process. AIM: The aim of this study was to evaluate plasma levels of CC cleavage fragments (C3a, C5a, and C5b9), sphingosine (SP), sphingosine-1-phosphate (S1P), and free hemoglobin (fHb) in AMI patients treated with primary percutaneous coronary intervention (pPCI) and stable coronary artery disease (SCAD) undergoing CABG. PATIENTS AND METHODS: The study enrolled 37 subjects (27 male) including 22 AMI patients, 7 CABG patients, and 8 healthy individuals as the control group (CTRL). In the AMI group, blood samples were collected at 5 time points (admission to hospital, 6, 12, 24, and 48 hours post pPCI) and 4 time points in the CABG group (6, 12, 24, and 48 hours post operation). SP and S1P concentrations were measured by high-performance liquid chromatography (HPLC). Analysis of C3a, C5a, and C5b9 levels was carried out using high-sensitivity ELISA and free hemoglobin by spectrophotometry. RESULTS: The plasma levels of CC cleavage fragments (C3a and C5b9) were significantly higher, while those of SP and S1P were lower in patients undergoing CABG surgery in comparison to the AMI group. In both groups, levels of CC factors showed no significant changes within 48 hours of follow-up. Conversely, SP and S1P levels gradually decreased throughout 48 hours in the AMI group but remained stable after CABG. Moreover, the fHb concentration was significantly higher after 24 and 48 hours post pPCI compared to the corresponding postoperative time points. Additionally, the fHb concentrations increased between 12 and 48 hours after PCI in patients with AMI. CONCLUSIONS: Inflammatory response after AMI and CABG differed regarding the release of sphingolipids, free hemoglobin, and complement cascade cleavage fragments.


Asunto(s)
Proteínas del Sistema Complemento/análisis , Enfermedad de la Arteria Coronaria/sangre , Hemoglobinas/análisis , Infarto del Miocardio/sangre , Esfingolípidos/metabolismo , Anciano , Estudios de Casos y Controles , Puente de Arteria Coronaria , Femenino , Humanos , Inflamación , Lisofosfolípidos/metabolismo , Masculino , Persona de Mediana Edad , Intervención Coronaria Percutánea , Esfingolípidos/sangre , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Resultado del Tratamiento
2.
Am J Transplant ; 16(2): 440-53, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26550777

RESUMEN

CD8(+)/TCR(-) facilitating cells (FCs) in mouse bone marrow (BM) significantly enhance engraftment of hematopoietic stem/progenitor cells (HSPCs). Human FC phenotype and mechanism of action remain to be defined. We report, for the first time, the phenotypic characterization of human FCs and correlation of phenotype with function. Approximately half of human FCs are CD8(+)/TCR(-)/CD56 negative (CD56(neg)); the remainder are CD8(+)/TCR(-)/CD56 bright (CD56(bright)). The CD56(neg) FC subpopulation significantly promotes homing of HSPCs to BM in nonobese diabetic/severe combined immunodeficiency/IL-2 receptor γ-chain knockout mouse recipients and enhances hematopoietic colony formation in vitro. The CD56(neg) FC subpopulation promotes rapid reconstitution of donor HSPCs without graft-versus-host disease (GVHD); recipients of CD56(bright) FCs plus HSPCs exhibit low donor chimerism early after transplantation, but the level of chimerism significantly increases with time. Recipients of HSPCs plus CD56(neg) or CD56(bright) FCs showed durable donor chimerism at significantly higher levels in BM. The majority of both FC subpopulations express CXCR4. Coculture of CD56(bright) FCs with HSPCs upregulates cathelicidin and ß-defensin 2, factors that prime responsiveness of HSPCs to stromal cell-derived factor 1. Both FC subpopulations significantly upregulated mRNA expression of the HSPC growth factors and Flt3 ligand. These results indicate that human FCs exert a direct effect on HSPCs to enhance engraftment. Human FCs offer a potential regulatory cell-based therapy for enhancement of engraftment and prevention of GVHD.


Asunto(s)
Antígenos CD8/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Células Madre Hematopoyéticas/inmunología , Subunidad gamma Común de Receptores de Interleucina/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Animales , Apoptosis , Western Blotting , Células Cultivadas , Enfermedad Injerto contra Huésped/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Animales , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Donantes de Tejidos , Quimera por Trasplante
3.
Saudi Pharm J ; 23(3): 303-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26106278

RESUMEN

In microbiological terms, pharmaceutical products can be divided into two groups: sterile and non-sterile. Non-sterile drugs must satisfy the appropriate microbiological purity criteria which are included in pharmacopoeial monographs. Pharmacopoeial studies are prepared specifically with a view to ensuring that the medicinal product is therapeutically effective and safe for the patient. The analysis comprised the results of microbiological purity tests performed before the products are marketed. Total of 1285 samples of non-sterile drugs manufactured by different pharmaceutical plants in Polish were taken into study. The microbiological quality of drugs was assessed in accordance with the criteria included in the European Pharmacopoeia (EP). An analysis of test results demonstrated that the percentage of non-compliant samples was 1.87%. The groups of drugs, which the most often did not satisfy EPs' requirements, were drugs containing raw materials of natural origin (5.7%). The samples of studied drugs that did not meet the criteria contained in EP, exceed the maximum allowable microbiological count limits and contained microbes whose presence is prohibited. The most common non-compliance was the excessive levels of the maximum acceptable fungal count (n = 12) and the excessive the maximum acceptable aerobic microbial count (n = 10).

4.
Appl Environ Microbiol ; 79(15): 4684-93, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23728810

RESUMEN

Escherichia coli, a commensal bacterium from the intestinal tracts of humans and vertebrate animals, has been used as one of two bacterial indicators of fecal contamination, along with intestinal enterococci, to monitor the microbiological quality of water. However, water environments are now recognized as a secondary habitat where some strains can survive. We investigated the survival of E. coli isolates collected from bodies of water in France exhibiting distinct profiles of contamination, defined according to the following criteria: vicinity of the point sources of contamination, land use, hydrology, and physicochemical characteristics of the receiving water. We selected 88 E. coli strains among a collection of 352 strains to carry out a microcosm experiment in filtered estuarine water for 14 days at 10°C. The relationship between the survival of E. coli strains and genotypic and phenotypic characteristics was analyzed. This work showed that distinct E. coli survival types, able to survive from between 7 and 14 days to less than 2 days, coexisted in the water. E. coli isolates that rapidly lost their culturability were more frequently isolated in water recently contaminated by fecal bacteria of human origin, and most were multiresistant to antibiotics and harbored several virulence factors. In contrast, persistent strains able to survive from 4 to 14 days were more often found in water with low levels of fecal bacteria, belonged mainly to the B1 phylogroup, often harbored only one virulence factor, kspE or ompT, and were able to grow at 7°C.


Asunto(s)
Biopelículas , Escherichia coli/fisiología , Estuarios , Ríos/microbiología , Antibacterianos/farmacología , Dictyostelium/fisiología , Farmacorresistencia Microbiana , Escherichia coli/clasificación , Escherichia coli/genética , Escherichia coli/patogenicidad , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Heces/microbiología , Cadena Alimentaria , Francia , Genotipo , Reacción en Cadena de la Polimerasa , Factores de Virulencia/genética , Contaminación del Agua/análisis
5.
J Exp Med ; 175(3): 743-50, 1992 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-1371302

RESUMEN

To determine whether N-ras expression is required at specific stages of the process of in vitro normal human hematopoiesis, adherent- and T lymphocyte-depleted mononuclear marrow cells (A-T-MNC) or highly purified progenitors (CD34+ cells) were cultured in semisolid medium, under conditions that favor the growth of specific progenitor cell types, after exposure to N-ras sense and antisense oligodeoxynucleotides. N-ras antisense, but not sense, oligodeoxynucleotide treatment of A-T-MNC and CD34+ cells resulted in a significantly decreased number of granulocyte/macrophage colony-forming units (CFU-GM) induced by interleukin 3 (IL-3) or granulocyte/macrophage colony-stimulating factor (GM-CSF) and of macrophage colonies (CFU-M) induced by M-CSF, but not of granulocytic colonies induced with G-CSF or IL-5. However, the same treatment significantly inhibited colony formation induced by each of the above factors in combination with IL-3. Megakaryocytic colony (CFU-Meg) formation from A-T-MNC or CD34+ cells in the presence of IL-6 + IL-3 + erythropoietin (Epo) was also markedly decreased after antisense oligodeoxynucleotide treatment. Erythroid colonies derived from A-T-MNC in the presence of Epo (CFU-E) were not inhibited upon antisense treatment, whereas those arising from A-T-MNC or CD34+ cells in the presence of IL-3 + Epo (BFU-E) were markedly affected. These results are consistent with the hypothesis that distinct signal transduction pathways, involving N-ras or not, are activated by different growth factors in different hematopoietic progenitor cells.


Asunto(s)
Sustancias de Crecimiento/farmacología , Hematopoyesis/efectos de los fármacos , Oligonucleótidos Antisentido/genética , Antígenos CD/análisis , Antígenos CD34 , Secuencia de Bases , Médula Ósea/inmunología , Células de la Médula Ósea , Depresión Química , Células Precursoras Eritroides/efectos de los fármacos , Genes ras/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/efectos de los fármacos , Humanos , Megacariocitos/citología , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/farmacología , ARN Mensajero/análisis , Células Madre/efectos de los fármacos , Transcripción Genética
6.
J Exp Med ; 178(6): 1923-33, 1993 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-8245773

RESUMEN

There is evidence to suggest that the p120 GAP (GAP), originally described as an inhibitor of p21ras, may also serve as a downstream effector of ras-regulated signal transduction. To determine whether GAP expression is required for the growth of human normal and leukemic hematopoietic cells, we used GAP antisense oligodeoxynucleotides to inhibit it and analyzed the effects of this inhibition on the colony-forming ability of nonadherent, T lymphocyte-depleted mononuclear cells and of highly purified progenitors (CD34+ MNC) obtained from the bone marrow and peripheral blood of healthy volunteers or chronic myeloid leukemia (CML, bcr-abl-positive) patients. The acute myelogenous leukemia cell line MO7, the Philadelphia BV173 cell line, and the acute promyelocytic leukemia NB4 and HL-60 cell lines were similarly examined. GAP antisense treatment inhibited colony formation from normal myelo-, erythro-, and megakaryopoietic progenitor cells as well as from CML progenitor cells. Proliferation of MO7 (growth factor-dependent) and BV173 (bcr-abl-dependent) cells, but not that of NB4 and HL-60 (growth factor-independent) cells, was also inhibited, even though a specific downregulation of GAP was observed in each cell line, as analyzed by either or both mRNA and protein expression. Stimulation of MO7 cells with hematopoietic growth factors increased the expression of GAP as well as the levels of active GTP-bound p21ras. Stimulation of GAP expression was inhibited upon GAP antisense treatment. These data indicate that p120 GAP is involved in human normal and leukemic hemopoiesis and strongly suggest that GAP is not only a p21ras inhibitor (signal terminator), but also a positive signal transducer.


Asunto(s)
Hematopoyesis , Proteínas/fisiología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Células de la Médula Ósea , División Celular/efectos de los fármacos , Línea Celular , Proteínas de Fusión bcr-abl/fisiología , Proteínas Activadoras de GTPasa , Expresión Génica , Sustancias de Crecimiento/farmacología , Humanos , Oligonucleótidos Antisentido , ARN Mensajero/genética , Transducción de Señal , Proteínas Activadoras de ras GTPasa
7.
Herz ; 35(7): 467-72, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20981396

RESUMEN

The bone marrow (BM) niche contains small heterogenous populations of cells which may contribute to cardiac and endothelial repair, including committed lineages [endothelial progenitor cells (EPCs), multipotent mesenchymal stromal cells (MSCs) and more primitive very small embryonic-like cells (VSELs) expressing pluripotent stem cell (PSC) markers (Oct-4, Nanog, SSEA-1)]. VSELs are present in BM, peripheral blood and some solid organs in mice and were recently identified in peripheral blood in patients with acute coronary syndromes and stroke. VSELs can be expanded in vitro and differentiated into cells from all three germ layers. This population of cells displays the morphology of primitive PSC (small size, open type chromatin, large nucleus, narrow rim of cytoplasm) and express PSC markers. The isolation of human VSELs is based on their size and presence of several surface markers (CXCR4, CD133, CD34) and lack of markers of hematopoietic lineage (lin, CD45). In acute myocardial infarction and ischemic stroke VSELs are rapidly mobilized into peripheral blood, and express increased levels of PSC markers as well as early cardiac (GATA-4, Nkx2.5/Csx), neural (GFAP, nestin, beta-III-tubulin, Olig1, Olig2, Sox2, Musashi) and endothelial lineage markers (VE-cadherin, von Willebrand factor). The number of VSELs mobilized in acute myocardial infarction is inversely correlated with left ventricular ejection fraction and the release of cardiac necrosis markers. Mobilization of these cells is also reduced in patients with diabetes and in the elderly. BM-derived VSELs were expanded and after cardiogenic pre-differentiation injected intramyocardially in mice models of myocardial infarction leading to improved left ventricular contractility. VSELs are probably progeny of epiblast cells which migrated to the BM and developing organs during embryonic development. The cells are present in a quiescent state in the adult BM and solid organs and might serve as a reserve pool of resident stem cells. VSELs are promising candidates for further pre-clinical and clinical studies on cellular cardiovascular therapy.


Asunto(s)
Síndrome Coronario Agudo/cirugía , Células Madre Embrionarias/trasplante , Miocitos Cardíacos/trasplante , Accidente Cerebrovascular/cirugía , Ingeniería de Tejidos/métodos , Síndrome Coronario Agudo/complicaciones , Síndrome Coronario Agudo/patología , Animales , Células Madre Embrionarias/patología , Humanos , Ratones , Miocitos Cardíacos/patología , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/patología
8.
Minerva Med ; 101(3): 179-91, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20562805

RESUMEN

The leading cause of death from cancer is tumor expansion, which usually leads to dissemination and metastasis of malignant cells. Accumulating evidence suggests growing tumors contain some very rare primitive cells that are mobile and thus endowed with metastatic potential. If these cells survive radio/chemotherapy, they are responsible for tumor re-growth after treatment. In this review, we discuss the origin of these cells, which: 1) are true cancer stem cells (CSCs) that initiate tumor growth and are subsequently responsible for metastatic dissemination; or 2) are derived from transformed tumor cells by the epithelial mesenchymal transition phenomenon. We also address major molecular mechanisms involved in trafficking of these cells during metastasis, paying special attention to the underappreciated side effects of radio/chemotherapy that may induce pro-metastatic environments in various organs. Overall, we envision that the process of pathological metastasis of cancer cells reflects a physiological property of normal SCs for their ability to migrate, as seen during embryogenesis. Finally, we discovered highly migratory, very small embryonic-like SCs that are deposited during development in adult tissues. As we hypothesize, these cells could: 1) give rise to some primitive types of tumors; and 2) may have a direct role in cancer expansion by being involved in tumor angiogenesis and formation of tumor stroma.


Asunto(s)
Metástasis de la Neoplasia/patología , Células Madre Neoplásicas/patología , Adulto , Antineoplásicos/uso terapéutico , Movimiento Celular/fisiología , Quimiocinas/fisiología , Humanos , MicroARNs/fisiología , Metástasis de la Neoplasia/tratamiento farmacológico , Neoplasias/embriología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/fisiología , Células Madre Neoplásicas/efectos de la radiación , Radioterapia/efectos adversos , Receptores CXCR/fisiología
9.
Stem Cell Rev Rep ; 16(1): 198-211, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31828580

RESUMEN

BACKGROUND: Bone marrow-derived mononuclear cells (BM-MNC) consist of a heterogeneous mix of mesenchymal stem cells (MSC), hematopoietic progenitor cells (HPC), endothelial progenitor cells (EPC), monocytes, lymphocytes and pluripotent stem cells. Whereas the importance of MSC and EPC has been well documented in bone healing and regeneration studies, the role of pluripotent stem cells is still poorly understood. In the present study we evaluated if and how Very Small Embryonic Like cells (VSEL), isolated from rat BM-MNC, contribute to bone healing. METHODS: Large bone defects were made in the femurs of 38 Sprague Dawley female rats and treated with ß-TCP scaffold granules seeded with male VSEL; BM-MNC, VSEL-depleted BM-MNC or scaffold alone, and bone healing was evaluated at 8 weeks post-surgery. RESULTS: Bone healing was significantly increased in defects treated with VSEL and BM-MNC, compared to defects treated with VSEL-depleted BM-MNC. Donor cells were detected in new bone tissue, in all the defects treated with cells, and in fibrous tissue only in defects treated with VSEL-depleted BM-MNC. The number of CD68+ cells was the highest in the VSEL-depleted group, whereas the number of TRAP positive cells was the lowest in this group. CONCLUSIONS: Based on the results, we can conclude that VSEL play a role in BM-MNC induced bone formation. In our rat femur defect model, in defects treated with VSEL-depleted BM-MNC, osteoclastogenesis and bone formation were decreased, and foreign body reaction was increased.


Asunto(s)
Células Madre Adultas/trasplante , Regeneración Ósea/genética , Trasplante de Células Madre Mesenquimatosas , Células Madre Pluripotentes/trasplante , Adulto , Animales , Células Progenitoras Endoteliales/trasplante , Humanos , Monocitos/trasplante , Osteogénesis/genética , Ratas
10.
Mech Ageing Dev ; 130(1-2): 58-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-18377952

RESUMEN

Recently our group identified in murine bone marrow (BM) and human cord blood (CB), a rare population of very small embryonic-like (VSEL) stem cells. We hypothesize that these cells are deposited during embryonic development in BM as a mobile pool of circulating pluripotent stem cells (PSC) that play a pivotal role in postnatal tissue turnover both of non-hematopoietic and hematopoietic tissues. During in vitro co-cultures with murine myoblastic C2C12 cells, VSELs form spheres that contain primitive stem cells. Cells isolated from these spheres may give rise to cells from all three germ layers when plated in tissue specific media. The number of murine VSELs and their ability to form spheres decreases with the age and is reduced in short-living murine strains. Thus, developmental deposition of VSELs in adult tissues may potentially play an underappreciated role in regulating the rejuvenation of senescent organs. We envision that the regenerative potential of these cells could be harnessed to decelerate aging processes.


Asunto(s)
Envejecimiento/fisiología , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Adulto , Anciano , Animales , Tamaño de la Célula , Senescencia Celular/fisiología , Células Madre Embrionarias/fisiología , Células Madre Hematopoyéticas/fisiología , Humanos
11.
Eur Ann Otorhinolaryngol Head Neck Dis ; 136(1): 33-36, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30072288

RESUMEN

Osteoma is a benign, usually asymptomatic bone tumour, frequently arising in the nose and paranasal sinuses. Surgical treatment is required when the patient becomes symptomatic or presents ophthalmological or neurological complications. Although an endoscopic approach is increasingly used, depending on the size and site of the osteoma, open surgery may be preferable and remains the standard treatment. This technical note describes a case of giant osteoma of the frontal sinus that required a bicoronal approach with reconstruction by a custom-made titanium prosthesis.


Asunto(s)
Prótesis Anclada al Hueso , Seno Frontal/cirugía , Osteoma/cirugía , Neoplasias de los Senos Paranasales/cirugía , Seno Frontal/diagnóstico por imagen , Humanos , Imagenología Tridimensional , Masculino , Persona de Mediana Edad , Osteoma/diagnóstico por imagen , Neoplasias de los Senos Paranasales/diagnóstico por imagen , Procedimientos de Cirugía Plástica , Cirugía Asistida por Computador/instrumentación , Titanio , Tomografía Computarizada por Rayos X
12.
Clin Biomech (Bristol, Avon) ; 63: 104-111, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30851565

RESUMEN

BACKGROUND: One of the most severe traumatic brain injuries, the subdural haematoma, is related to damage and rupture of the bridging veins, generating an abnormal collection of blood between the dura mater and arachnoid mater. Current numerical models of these vessels rely on very simple geometries and material laws, limiting its accuracy and bio-fidelity. METHODS: In this work, departing from an existing human head numerical model, a realistic geometry for the bridging veins was developed, devoting special attention to the finite elements type employed. A novel and adequate constitutive model including damage behavior was also successfully implemented. FINDINGS: Results attest that vessel tearing onset was correctly captured, after comparison against experiments on cadavers. INTERPRETATION: Doing so, the model allow to precisely predict the individual influence of kinematic parameters such as the pulse duration, linear and rotational accelerations in promoting vessel tearing.


Asunto(s)
Hematoma Subdural/diagnóstico , Rotura/diagnóstico , Aceleración , Fenómenos Biomecánicos , Cadáver , Simulación por Computador , Elasticidad , Femenino , Análisis de Elementos Finitos , Cabeza/fisiopatología , Hematoma Subdural/fisiopatología , Humanos , Masculino , Modelos Teóricos , Rotura/fisiopatología
13.
Cell Tissue Res ; 331(1): 125-34, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17828555

RESUMEN

Bone marrow (BM) develops in mammals by the end of the second/beginning of the third trimester of gestation and becomes a major hematopoietic organ in postnatal life. The alpha-chemokine stromal derived factor-1 (SDF-1) to CXCR4 (G ai-protein-coupled seven transmembrane-spanning chemokine receptor) axis plays a major role in BM colonization by stem cells. By the end of the second trimester of gestation, BM becomes colonized by hematopoietic stem cells (HSC), which are chemoattracted from the fetal liver in a CXCR4-SDF-1-dependent manner. Whereas CXCR4 is expressed on HSC, SDF-1 is secreted by BM stroma and osteoblasts that line BM cavities. Mounting evidence indicates that BM also contains rare CXCR4(+) pluripotent stem cells (PSC). Recently, our group has identified a population of CXCR4(+) very small embryonic like stem cells in murine BM and human cord blood. We hypothesize that these cells are deposited during development in BM as a mobile pool of circulating PSC that play a pivotal role in postnatal tissue turnover, both of non-hematopoietic and hematopoietic tissues.


Asunto(s)
Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Animales , Impresión Genómica , Células Germinativas , Humanos , Células Madre Pluripotentes/citología
14.
Cytotherapy ; 10(2): 182-92, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18368597

RESUMEN

BACKGROUND: In patients transplanted with cord blood (CB), prolonged thrombocytopenia is a major complication. However, this could be alleviated by supplementing the CB graft with ex vivo-expanded megakaryocytic progenitors (CFU-Meg), provided that the homing properties of these cells are not affected negatively by expansion. METHODS AND RESULTS: We assessed the in vitro homing potential of CFU-Meg progenitors expanded from CB and showed that the combination of thrombopoietin (TPO) with interleukin-3 (IL-3) used for expansion not only results in optimal proliferation of CFU-Meg but also protects these cells from apoptosis. Moreover, we found that ex vivo-expanded CFU-Meg maintained expression of the CXCR4 receptor throughout a 9-day culture and were chemoattracted towards a stromal cell-derived factor-1 (SDF-1) gradient. They also expressed matrix metalloproteinase-9 (MMP-9) and membrane-type (MT) 1-MMP, and transmigrated across the reconstituted basement membrane Matrigel. Finally, we observed that SDF-1 up-regulated the expression of both MMP-9 and MT1-MMP in CB CD34(+) cells and ex vivo-expanded CFU-Meg. DISCUSSION: We suggest that CB-expanded CFU-Meg, in particular those from day 3 of expansion, when their proliferation and in vitro homing potential are maximal, could be employed to supplement CB grafts and speed up platelet recovery in transplant recipients.


Asunto(s)
Ensayo de Unidades Formadoras de Colonias , Sangre Fetal/citología , Sangre Fetal/enzimología , Metaloproteinasa 14 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Megacariocitos/citología , Células Madre/citología , Antígenos CD34/metabolismo , Catequina/análogos & derivados , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Factores Quimiotácticos/farmacología , Quimiotaxis/efectos de los fármacos , Colágeno/metabolismo , Combinación de Medicamentos , Sangre Fetal/efectos de los fármacos , Humanos , Interleucina-3/farmacología , Cinética , Laminina/metabolismo , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Megacariocitos/efectos de los fármacos , Megacariocitos/enzimología , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Proteoglicanos/metabolismo , Receptores CXCR4/metabolismo , Células Madre/efectos de los fármacos , Trombopoyetina/farmacología , Regulación hacia Arriba/efectos de los fármacos
15.
Leukemia ; 21(5): 973-82, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17330096

RESUMEN

We hypothesized that the third complement component (C3) cleavage fragments (C3a and (des-Arg)C3a) are involved in stress/inflammation-related thrombocytosis, and investigated their potential role in reactive thrombocytosis induced by bleeding. We found that platelet counts are lower in C3-deficient mice in response to excessive bleeding as compared to normal littermates and that C3a and (des-Arg)C3a enhance stromal-derived factor-1 (SDF-1)-dependent megakaryocyte (Megs) migration, adhesion and platelet shedding. At the molecular level, C3a stimulates in Megs MAPKp42/44 phosphorylation, and enhances incorporation of CXCR4 into membrane lipid rafts increasing the responsiveness of Megs to SDF-1. We found that perturbation of lipid raft formation by statins decreases SDF-1/C3a-dependent platelet production in vitro and in an in vivo model statins ameliorated post-bleeding thrombocytosis. Thus, inhibition of lipid raft formation could find potential clinical application as a means of ameliorating some forms of thrombocytosis.


Asunto(s)
Quimiocinas CXC/fisiología , Complemento C3a/fisiología , Hemorragia/fisiopatología , Trombocitosis/fisiopatología , Trombopoyesis , Quimiocina CXCL12 , Humanos , Microdominios de Membrana/fisiología , Recuento de Plaquetas , Receptores CXCR4/fisiología , Receptores de Complemento/análisis , Transducción de Señal
16.
Leukemia ; 21(5): 860-7, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17344915

RESUMEN

Accumulating evidence demonstrates that adult tissues contain a population of stem cells that express early developmental markers such as stage-specific embryonic antigen and transcription factors Oct-4 and Nanog. These are the markers characteristic for embryonic stem cells, epiblast stem cells and primordial germ cells. The presence of these stem cells in adult tissues including bone marrow, epidermis, bronchial epithelium, myocardium, pancreas and testes supports the concept that adult tissues contain some population of pluripotent stem cells that is deposited in embryogenesis during early gastrulation. In this review we will discuss these data and present a hypothesis that these cells could be direct descendants of the germ lineage. The germ lineage in order to pass genes on to the next generations creates soma and thus becomes a 'mother lineage' for all somatic cell lineages present in the adult body.


Asunto(s)
Células Madre Adultas/química , Células de la Médula Ósea/citología , Células Madre Embrionarias/citología , Factor 3 de Transcripción de Unión a Octámeros/análisis , Células Madre Pluripotentes/citología , Animales , Linaje de la Célula , Humanos
17.
Leukemia ; 21(2): 297-303, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17136117

RESUMEN

Recently, we purified from adult murine bone marrow (BM) a population of CXCR4(+), Oct-4(+) SSEA-1(+), Sca-1(+) lin(-) CD45(-) very small embryonic-like (VSEL) stem cells and hypothesized that similar cells could be also present in human cord blood (CB). Here, we report that by employing a novel two-step isolation procedure -- removal of erythrocytes by hypotonic lysis combined with multiparameter sorting -- we could isolate from CB a population of human cells that are similar to murine BM-derived VSELs, described previously by us. These CB-isolated VSELs (CB-VSEL) are very small (3-5 micro m) and highly enriched in a population of CXCR4(+)AC133(+)CD34(+)lin(-) CD45(-) CB mononuclear cells, possess large nuclei containing unorganized euchromatin and express nuclear embryonic transcription factors Oct-4 and Nanog and surface embryonic antigen SSEA-4. Further studies are needed to see if human CB-isolated VSELs similar to their murine BM-derived counterparts are endowed with pluripotent stem cell properties.


Asunto(s)
Células Madre Embrionarias/citología , Sangre Fetal/citología , Glicoesfingolípidos/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Receptores CXCR4/genética , Animales , Células de la Médula Ósea/citología , Tamaño de la Célula , Células Madre Embrionarias/ultraestructura , Humanos , Recién Nacido , Ratones , Microscopía Electrónica , ARN/genética , ARN/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Antígenos Embrionarios Específico de Estadio
18.
J Clin Invest ; 104(12): 1739-49, 1999 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-10606628

RESUMEN

CD34(+) cells are nonpermissive to infection by HIV strains X4 and R5, despite the fact that many CD34(+) cells express high levels of the viral receptor protein CD4 and the coreceptor CXCR4 on their surface. In these cells, the co-receptor CCR5 protein, which, like CXCR4, is a chemokine receptor, is detected mainly intracellularly. We hypothesized that CD34(+) cells secrete CCR5-binding chemokines and that these factors interfere with HIV R5 interactions with these cells, possibly by binding CCR5 or by inducing its internalization. We found that human CD34(+) cells and CD34(+)KIT(+) cells, which are enriched in myeloid progenitor cells, expressed and secreted the CCR5 ligands RANTES, MIP-1alpha, and MIP-1beta and that IFN-gamma stimulated expression of these chemokines. In contrast, SDF-1, a CXCR4 ligand, was not detectable in the CD34(+)KIT(+) cells, even by RT-PCR. Conditioned media from CD34(+) cell culture significantly protected the T lymphocyte cell line PB-1 from infection by R5 but not X4 strains of HIV. Interestingly, the secretion of endogenous chemokines decreased with the maturation of CD34(+) cells, although ex vivo, expanded megakaryoblasts still secreted a significant amount of RANTES. Synthesis of CCR5-binding chemokines by human CD34(+) cells and megakaryoblasts therefore largely determines the susceptibility of these cells to infection by R5 HIV strains. We postulate that therapeutic agents that induce the endogenous synthesis of chemokines in human hematopoietic cells may protect these cells from HIV infection.


Asunto(s)
Antígenos CD34/análisis , Células de la Médula Ósea/fisiología , Quimiocinas/fisiología , VIH/fisiología , Células Madre Hematopoyéticas/virología , Megacariocitos/fisiología , Quimiocinas/análisis , Humanos , Interferón gamma/farmacología , ARN Mensajero/análisis , Receptores CCR5/análisis , Receptores CXCR4/análisis
19.
J Clin Invest ; 94(1): 320-7, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8040273

RESUMEN

IGF-I has been reported to increase hematopoietic progenitor cell cloning efficiency. To investigate this phenomenon, we studied the IGF-I responsiveness of human marrow cells expressing IGF-I receptor (IGF-IR), a direct strategy not used previously. IGF-IR+ and control CD34+ marrow cells were isolated using immunoaffinity methods. Then, the cells were cloned in methylcellulose containing variable amounts of serum- and lineage-appropriate growth factors supplemented with recombinant human IGF-I. In contrast to CD34+ cells, IGF-IR+ cells never gave rise to CFU-Blast, CFU-Mix, CFU-GM, BFU-E, or CFU-E. To substantiate the suggestion that CD34+ and IGF-IR+ cells were distinct populations, we used reverse transcription PCR to detect IGF-I, EpO, and KIT receptor mRNAs in these cells. The mRNA phenotype of CD34+ cells was EpO (+), KIT (+), and IGF-IR (-), while IGF-IR+ cells were IGF-IR (+), EpO (-), and KIT (-). These results suggested that IGF-IR is either not expressed or expressed at low levels on normal hematopoietic progenitor cells. Functional significance of the latter possibility was tested by exposing CD34+ cells to IGF-IR antisense oligodeoxynucleotides. Colony formation was unaffected by oligodeoxynucleotide disruption of IGF-IR, suggesting that, even if expressed at low level, the receptor's functional significance was doubtful. Nevertheless, when cultured in the presence of IGF-I, IGF-IR+ cells elaborated an activity with mild BFU-E stimulatory effects. Accordingly, if IGF-I plays a role in hematopoietic colony formation, it is probably and results from stimulation of IGF-IR-positive ancillary cells to secrete growth factors. Studies carried out with human leukemia cells yielded similar results.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/farmacología , Secuencia de Bases , División Celular/efectos de los fármacos , Células Cultivadas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Leucemia/patología , Datos de Secuencia Molecular , Receptor IGF Tipo 1/análisis
20.
Leukemia ; 20(9): 1487-95, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16791265

RESUMEN

Normal and malignant cells shed from their surface membranes as well as secrete from the endosomal membrane compartment circular membrane fragments called microvesicles (MV). MV that are released from viable cells are usually smaller in size compared to the apoptotic bodies derived from damaged cells and unlike them do not contain fragmented DNA. Growing experimental evidence indicates that MV are an underappreciated component of the cell environment and play an important pleiotropic role in many biological processes. Generally, MV are enriched in various bioactive molecules and may (i) directly stimulate cells as a kind of 'signaling complex', (ii) transfer membrane receptors, proteins, mRNA and organelles (e.g., mitochondria) between cells and finally (iii) deliver infectious agents into cells (e.g., human immuno deficiency virus, prions). In this review, we discuss the pleiotropic effects of MV that are important for communication between cells, as well as the role of MV in carcinogenesis, coagulation, immune responses and modulation of susceptibility/infectability of cells to retroviruses or prions.


Asunto(s)
Comunicación Celular , Membrana Celular/fisiología , Orgánulos/fisiología , Progresión de la Enfermedad , Humanos , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA