Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Biochim Biophys Acta ; 1849(4): 371-7, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25038585

RESUMEN

Over a decade of intensive investigation of the possible plasticity of mammalian cells has eventually substantiated that mammalian species are endowed with a remarkable capacity to change mature cell fates. We review below the evidence for the occurrence of processes such as dedifferentiation and transdifferentiation within mammalian tissues in vivo, and in cells removed from their protective microenvironment and seeded in culture under conditions poorly resembling their physiological state in situ. Overall, these studies point to one major conclusion: stressful conditions, whether due to in vivo tissue damage or otherwise to isolation of cells from their in vivo restrictive niches, lead to extreme fate changes. Some examples of dedifferentiation are discussed in detail showing that rare cells within the population tend to turn back into less mature ones due to severe cell damage. It is proposed that cell stress, mechanistically sensed by isolation from neighboring cells, leads to dedifferentiation, in an attempt to build a new stem cell reservoir for subsequent regeneration of the damaged tissue. This article is part of a Special Issue entitled: Stress as a fundamental theme in cell plasticity.


Asunto(s)
Desdiferenciación Celular , Fenómenos Fisiológicos Celulares , Estrés Fisiológico/fisiología , Animales , Reprogramación Celular , Humanos , Mamíferos
2.
Stem Cells ; 32(8): 2008-20, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24715711

RESUMEN

Mesenchymal stromal cell populations include a fraction, termed mesenchymal stem cells, exhibiting multipotency. Other cells within this population possess a lesser differentiation range. This was assumed to be due to a mesenchymal cellular cascade topped by a multipotent cell, which gives rise to progeny with diminishing differentiation potentials. Here, we show that mesenchymal cells, a priori exhibiting a limited differentiation potential, may gain new capacities and become multipotent following single-cell isolation. These fate changes were accompanied by upregulation of differentiation promoting genes, many of which also became H4K20me1 methylated. Early events in the process included TGFß and Wnt modulation, and downregulation of hypoxia signaling. Indeed, hypoxic conditions inhibited the observed cell changes. Overall, cell isolation from neighboring partners caused major molecular changes and particularly, a newly established epigenetic state, ultimately leading to the acquisition of new differentiation potentials and an altered cell fate.


Asunto(s)
Diferenciación Celular/fisiología , Separación Celular , Células Madre Mesenquimatosas/citología , Animales , Células de la Médula Ósea/citología , Técnicas de Cultivo de Célula , Linaje de la Célula , Inmunoprecipitación de Cromatina , Células Clonales/citología , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa
3.
Blood ; 113(15): 3530-41, 2009 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18931338

RESUMEN

The default pathway of cell-surface T-cell receptor (TCR) complex formation, and the subsequent transport to the membrane, is thought to entail endoplasmic reticulum (ER) localization followed by proteasome degradation of the unassembled chains. We show herein an alternative pathway: short, incomplete peptide versions of TCRbeta naturally occur in the thymus. Such peptides, which have minimally lost the leader sequence or have been massively truncated, leaving only the very C terminus intact, are sorted preferentially to the mitochondrion. As a consequence of the mitochondrial localization, apoptotic cell death is induced. Structure function analysis showed that both the specific localization and induction of apoptosis depend on the transmembrane domain (TMD) and associated residues at the COOH-terminus of TCR. Truncated forms of TCR, such as the short peptides that we detected in the thymus, may be products of protein degradation within thymocytes. Alternatively, they may occur through the translation of truncated mRNAs resulting from unfruitful rearrangement or from germline transcription. It is proposed that mitochondria serve as a subcellular sequestration site for incomplete TCR molecules.


Asunto(s)
Apoptosis/inmunología , Mitocondrias/metabolismo , Transporte de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Células COS , Supervivencia Celular/inmunología , Chlorocebus aethiops , Reordenamiento Génico de Linfocito T , Proteínas Fluorescentes Verdes/genética , Región Variable de Inmunoglobulina/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Estructura Terciaria de Proteína , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transducción de Señal/inmunología , Timo/citología
4.
Med Sci (Paris) ; 27(3): 303-7, 2011 Mar.
Artículo en Francés | MEDLINE | ID: mdl-21447304

RESUMEN

Stem cells are a non-coherent group of cells that have little in common. Despite the fact that these diverse cell types are regarded as belonging to the same category, they do not share molecular markers. The definition of stemness is therefore descriptive, relating to potentials of the cells rather than to the actual properties that they harbor. This situation is confusing and causes unnecessary debates in this field of research. It is therefore of paramount importance to find a new, molecular definition of stemness, that would consist of the cellular machineries which constitute the stem cell state.


Asunto(s)
Células Madre/fisiología , Animales , Desdiferenciación Celular , Diferenciación Celular , Transdiferenciación Celular , Biología Molecular
5.
J Immunol ; 181(5): 3714-24, 2008 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18714048

RESUMEN

CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs) play a central role in cancer tolerance. However, mechanisms leading to their accumulation in cancer remain unknown. Although the thymus is the main site of Treg development, thymic contribution to Treg expansion in cancer has not been directly examined. Herein, we used two murine models of multiple myeloma (MM), 5T2 MM and 5T33 MM, to examine Treg accumulation in peripheral lymphoid organs, including spleen, lymph nodes, bone marrow, and blood, and to explore thymic Treg development during malignancy. We found that peripheral ratios of suppressive-functional Tregs increased in both models of MM-inflicted mice. We found that thymic ratios of Treg development in MM increased, in strong association with thymus atrophy and altered developmental processes in the thymus. The CD4(+)CD8(+) double-positive population, normally the largest thymocyte subset, is significantly decreased, whereas the CD4(-)CD8(-) double-negative population is increased. Administration of thymocytes from MM-inflicted mice compared with control thymocytes resulted in increased progression of the disease, and this effect was shown to be mediated by Tregs in the thymus of MM-inflicted mice. Our data suggest that increased ratios of Treg development in the thymus may contribute to disease progression in MM-inflicted mice.


Asunto(s)
Atrofia , Mieloma Múltiple/inmunología , Subgrupos de Linfocitos T/patología , Linfocitos T Reguladores/patología , Timo/patología , Animales , Modelos Animales de Enfermedad , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Mieloma Múltiple/etiología
6.
Exp Cell Res ; 315(11): 1904-13, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19328780

RESUMEN

Multiple myeloma is characterized by the malignant growth of immunoglobulin producing plasma cells, predominantly in the bone marrow. The effects of primary human mesenchymal stromal cells on the differentiation phenotype of multiple myeloma cells were studied by co-culture experiments. The incubation of multiple myeloma cells with bone marrow-derived mesenchymal stromal cells resulted in significant reduction of the expression of the predominant plasma cell differentiation markers CD38 and CD138, and cell surface immunoglobulin light chain. While the down-regulation of CD138 by stromal cells was completely dependent on their adhesive interactions with the multiple myeloma cells, interleukin-6 induced specific down-regulation of CD38. Mesenchymal stromal cells or their conditioned media inhibited the growth of multiple myeloma cell line, thereby reducing the overall amounts of secreted light chains. Analysis of primary multiple myeloma bone marrow samples reveled that the expression of CD38 on multiple myeloma cells was not affected by adhesive interactions. The ex vivo propagation of primary multiple myeloma cells resulted in significant increase in their differentiation markers. Overall, the data indicate that the bone marrow-derived mesenchymal stromal cells revert multiple myeloma cells to less differentiated phenotype by the combined activities of adhesive interactions and interleukin-6.


Asunto(s)
Interleucina-6/fisiología , Mesodermo/citología , Mesodermo/fisiología , Mieloma Múltiple/patología , Mieloma Múltiple/fisiopatología , Células del Estroma/citología , Células del Estroma/fisiología , ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/farmacología , Biomarcadores de Tumor/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Adhesión Celular , Diferenciación Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Regulación hacia Abajo , Humanos , Cadenas kappa de Inmunoglobulina/metabolismo , Interleucina-6/antagonistas & inhibidores , Interleucina-6/farmacología , Glicoproteínas de Membrana/metabolismo , Mesodermo/efectos de los fármacos , Pruebas de Neutralización , Fenotipo , Células Plasmáticas/citología , Células Plasmáticas/fisiología , Proteínas Recombinantes/farmacología , Células del Estroma/efectos de los fármacos , Sindecano-1/metabolismo
7.
Stem Cells ; 26(9): 2275-86, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18556513

RESUMEN

Cultured bone marrow stromal cells create an in vitro milieu supportive of long-term hemopoiesis and serve as a source for multipotent mesenchymal progenitor cells defined by their ability to differentiate into a variety of mesodermal derivatives. This study aims to examine whether the capacity to support myelopoiesis is coupled with the multipotency. Our results show that the myelopoietic supportive ability of stromal cells, whether from the bone marrow or from embryo origin, is not linked with multipotency; cell populations that possess multipotent capacity may or may not support myelopoiesis, whereas others, lacking multipotency, may possess full myelopoietic supportive ability. However, upon differentiation, the ability of multipotent mesenchymal progenitors to support myelopoiesis is varied. Osteogenic differentiation did not affect myelopoietic supportive capacity, whereas adipogenesis resulted in reduced ability to support the maintenance of myeloid progenitor cells. These differences were accompanied by a divergence in glycosylation patterns, as measured by binding to lectin microarrays; osteogenic differentiation was associated with an increased level of antennarity of N-linked glycans, whereas adipogenic differentiation caused a decrease in antennarity. Inhibition of glycosylation prior to seeding the stroma with bone marrow cells resulted in reduced capacity of the stromal cells to support the formation of cobblestone areas. Our data show that myelopoietic support is unrelated to the multipotent phenotype of cultured mesenchymal progenitors but is dependent on the choice of differentiation pathway and upon correct glycosylation of the stromal cells.


Asunto(s)
Células de la Médula Ósea/citología , Linaje de la Célula , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Mielopoyesis , Animales , Diferenciación Celular , Células Cultivadas , Glicosilación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células del Estroma/citología
8.
Stem Cells Dev ; 17(1): 93-106, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18225977

RESUMEN

In vitro and in vivo studies implicate a series of cytokines in regulation of lymphohematopoiesis. However, direct indications for a local role of most of these cytokines within the bone marrow is lacking. In the present study, we aimed to test the contribution of a specific cytokine, activin A, a member of the transforming growth factor-beta (TGF-beta) family, to lymphohematopoiesis in mouse bone marrow. We show that mouse embryonic fibroblasts (MEFs) are indistinguishable from multipotent stromal cells (MSCs). Such MEFs overexpressing activin A, supported in vitro myelopoiesis in long-term bone marrow cultures as effectively as control MEFs. In contrast, activin A-overexpressing MEFs interfered with the in vitro generation of B lineage cells in such cultures. Thus, excessive expression in vitro of activin A, by supportive stromal cells, causes preferential maturation of myeloid rather than lymphoid cells. Moreover, the activin A-overexpressing MEFs caused an increased incidence in vivo of relatively immature B lineage cells; upon transplantation through the spleen route, MEFs engrafted the bone marrow specifically. Activin A-overexpressing MEFs accumulated in the bone marrow compartment and slowed down the progression of B cell precursors along the differentiation pathway, while sparing the myeloid population. The assay system described in this paper provides a means to assess the contribution of a wide range of molecules to hematopoiesis without perturbing the constitution of other organs.


Asunto(s)
Activinas/genética , Linfocitos B/citología , Médula Ósea , Linfopoyesis/genética , Células del Estroma/metabolismo , Activinas/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias , Femenino , Fibroblastos , Expresión Génica , Ratones , Células Madre Multipotentes
9.
Mol Biol Cell ; 16(5): 2395-413, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15758027

RESUMEN

Nucleolar segregation is observed under some physiological conditions of transcriptional arrest. This process can be mimicked by transcriptional arrest after actinomycin D treatment leading to the segregation of nucleolar components and the formation of unique structures termed nucleolar caps surrounding a central body. These nucleolar caps have been proposed to arise from the segregation of nucleolar components. We show that contrary to prevailing notion, a group of nucleoplasmic proteins, mostly RNA binding proteins, relocalized from the nucleoplasm to a specific nucleolar cap during transcriptional inhibition. For instance, an exclusively nucleoplasmic protein, the splicing factor PSF, localized to nucleolar caps under these conditions. This structure also contained pre-rRNA transcripts, but other caps contained either nucleolar proteins, PML, or Cajal body proteins and in addition nucleolar or Cajal body RNAs. In contrast to the capping of the nucleoplasmic components, nucleolar granular component proteins dispersed into the nucleoplasm, although at least two (p14/ARF and MRP RNA) were retained in the central body. The nucleolar caps are dynamic structures as determined using photobleaching and require energy for their formation. These findings demonstrate that the process of nucleolar segregation and capping involves energy-dependent repositioning of nuclear proteins and RNAs and emphasize the dynamic characteristics of nuclear domain formation in response to cellular stress.


Asunto(s)
Nucléolo Celular/metabolismo , Transcripción Genética , Secuencia de Bases , Línea Celular , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/ultraestructura , ADN/genética , Dactinomicina/farmacología , Metabolismo Energético , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Microscopía Inmunoelectrónica , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Factor de Empalme Asociado a PTB , ARN Nuclear/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transcripción Genética/efectos de los fármacos , Proteína p14ARF Supresora de Tumor/metabolismo
10.
Cell Death Dis ; 9(6): 695, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29891848

RESUMEN

Fas-L is a TNF family member known to trigger cell death. It has recently become evident that Fas-L can transduce also non-apoptotic signals. Mesenchymal stem cells (MSCs) are multipotent cells that are derived from various adult tissues. Although MSCs from different tissues display common properties they also display tissue-specific characteristics. Previous works have demonstrated massive apoptosis following Fas-L treatment of bone marrow-derived MSCs both in vitro and following their administration in vivo. We therefore set to examine Fas-L-induced responses in adipose-derived stem cells (ASCs). Human ASCs were isolated from lipoaspirates and their reactivity to Fas-L treatment was examined. ASCs responded to Fas-L by simultaneous apoptosis and proliferation, which yielded a net doubling of cell quantities and a phenotypic shift, including reduced expression of CD105 and increased expression of CD73, in association with increased bone differentiation potential. Treatment of freshly isolated ASCs led to an increase in large colony forming unit fibroblasts, likely produced by early stem cell progenitor cells. Fas-L-induced apoptosis and proliferation signaling were found to be independent as caspase inhibition attenuated Fas-L-induced apoptosis without impacting proliferation, whereas inhibition of PI3K and MEK, but not of JNK, attenuated Fas-L-dependent proliferation, but not apoptosis. Thus, Fas-L signaling in ASCs leads to their expansion and phenotypic shift toward a more potent stem cell state. We speculate that these reactions ensure the survival of ASC progenitor cells encountering Fas-L-enriched environments during tissue damage and inflammation and may also enhance ASC survival following their administration in vivo.


Asunto(s)
Tejido Adiposo/citología , Proteína Ligando Fas/farmacología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Antígenos CD/metabolismo , Apoptosis/efectos de los fármacos , Huesos/citología , Inhibidores de Caspasas/farmacología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ensayo de Unidades Formadoras de Colonias , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Fenotipo , Fosfatidilinositol 3-Quinasas/metabolismo , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
11.
J Clin Anesth ; 19(3): 168-74, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17531723

RESUMEN

STUDY OBJECTIVE: To investigate the immunohistochemical localization of betaA subunit of activin A in human term placenta, as a marker for placental infection/inflammation and elevated temperature, in parturients laboring during two analgesic regimens. DESIGN: Prospective, randomized controlled study. SETTING: Delivery room. PATIENTS: 56 healthy, ASA physical status I and II primiparous women in labor. INTERVENTIONS: Parturients were assigned to receive patient-controlled epidural analgesia (PCEA) with 0.2% ropivacaine or patient-controlled intravenous analgesia PCA with meperidine. MEASUREMENTS: Histologic and immunohistochemical placental evaluation for white blood cell infiltration and activin betaA staining were made. Maternal temperature elevation above 37.6 degrees C and leukocytosis above 15,000/microL were recorded. MAIN RESULTS: Temperature was not significantly increased in parturients receiving PCEA over those who received (PCA) with meperidine (31% vs 11%, respectively; P = 0.1). There was also no association between temperature elevation during epidural analgesia and increased white blood cell count (>15,000/microL) or presence of polymorphonuclear and/or lymphocyte aggregation in the placenta. Immunohistochemical staining with antisera against the betaA subunit of activin was present mainly in the placental cytotrophoblast, syncytiotrophoblast, and vascular endothelium, and was not associated with an increase in maternal temperature. No significant difference was noted between the two analgesic techniques with regard to maternal temperature elevation. Intrapartum temperature elevation was not associated with histologic signs of placental inflammation or with expression of activin betaA in the placenta. CONCLUSION: Other mechanisms may be involved in the etiology of temperature elevation during labor.


Asunto(s)
Analgesia Epidural/efectos adversos , Analgesia Obstétrica/efectos adversos , Corioamnionitis/diagnóstico , Fiebre/etiología , Subunidades beta de Inhibinas/análisis , Placenta/química , Adulto , Analgesia Controlada por el Paciente/efectos adversos , Analgésicos Opioides , Biomarcadores/análisis , Corioamnionitis/patología , Femenino , Humanos , Meperidina , Placenta/patología , Embarazo , Estudios Prospectivos
12.
Oncotarget ; 7(41): 67061-67070, 2016 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-27563813

RESUMEN

INTRODUCTION: Multiple myeloma is still incurable in most cases. Polyclonal anti T lymphocyte globulins (ATG) have been reported to kill human myeloma cells in vitro and in mouse models. METHODS: Anti-human-myeloma globulins (AMG) were produced by immunizing rabbits with human myeloma cell lines RPMI-8226 (AMG-8226) or KMS-12-BM (AMG-12-BM). Cytotoxicity of the polyclonal antibodies was analyzed in vitro and in a xenograft NOD-SCID mouse model. RESULTS: Both AMG had stronger cytotoxicity against myeloma cells compared to ATG. In primary T cells, AMG-8226 showed greater complement-dependent cytotoxicity (CDC) than ATG, whereas complement-independent cytotoxicity did not differ. Effects on non-hematopoietic cell lines were also similar. Competitive blocking assays revealed fourfold more antibodies against CD38 in AMG-8226 compared to ATG. Low concentrations of AMG-8226 and ATG increased ADCC. At higher concentrations, ATG inhibited ADCC more potently than AMG-8226. Combinations of ATG and AMG-8226 with melphalan or bortezomib showed additive to synergistic cytotoxicity on myeloma cells. The cytotoxic effects of AMG and ATG were confirmed in the xenograft NOD-SCID mouse model. CONCLUSION: Our data show more potent antimyeloma effects of AMG compared to ATG. These results lay the ground for the development of polyclonal antibodies for the treatment of multiple myeloma.


Asunto(s)
Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Suero Antilinfocítico/farmacología , Antineoplásicos/farmacología , Mieloma Múltiple , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncogene ; 21(13): 2029-36, 2002 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-11960375

RESUMEN

The mesenchyme plays a crucial regulatory role in organ formation and maintenance. However, comprehensive molecular characterization of these cells is lacking. We found unexpectedly that primary mesenchyme, as well as mesenchymal cell clones, express T cell receptor (TCR)alphabeta mRNAs, lacking the variable region. Immunological and genetic evidence support the expression of a corresponding TCRbeta protein. Additionally, mRNAs encoding TCR complex components including CD3 and zeta chain are present. A relatively higher expression of the mesenchymal TCRbeta mRNA by cultured mesenchymal cell clones correlates with fast growth, whereas poorly expressing cells are slow growers and are contact inhibited. The clones that express relatively higher amount of the TCR mRNA exhibit an increased capacity to form tumors in nude mice. However, the expression of this mRNA in the mesenchyme is not per se leading to tumorigenesis, as demonstrated by primary mesenchyme that does not form tumors in mice while expressing moderate amounts of the TCR transcripts. The expression of mesencymal TCRbeta was confined to the G2/M phases of the cell cycle in the MBA-13 mesenchymal cell line. This cell cycle dependent expression, considered together with the correlation between growth properties and the level of TCR expression by cell clones, implies association of mesenchymal TCR with cell growth control.


Asunto(s)
Mesodermo/citología , Mesodermo/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Animales , Ciclo Celular , División Celular , Línea Celular , Citometría de Flujo , Perfilación de la Expresión Génica , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Células Tumorales Cultivadas
14.
Circulation ; 108(7): 863-8, 2003 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-12900340

RESUMEN

BACKGROUND: Systemic delivery of bone marrow-derived mesenchymal stem cells (BM-MSCs) is an attractive approach for myocardial repair. We aimed to test this strategy in a rat model after myocardial infarction (MI). METHODS AND RESULTS: BM-MSCs were obtained from rat bone marrow, expanded in vitro to a purity of >50%, and labeled with 99mTc exametazime, fluorescent dye, LacZ marker gene, or bromodeoxyuridine. Rats were subjected to MI by transient coronary artery occlusion or to sham MI. 99mTc-labeled cells (4x10(6)) were transfused into the left ventricular cavity of MI rats either at 2 or 10 to 14 days after MI and were compared with sham-MI rats or MI rats treated with intravenous infusion. Gamma camera imaging and isolated organ counting 4 hours after intravenous infusion revealed uptake of the 99mTc-labeled cells mainly in the lungs, with significantly smaller amounts in the liver, heart, and spleen. Delivery by left ventricular cavity infusion resulted in drastically lower lung uptake, better uptake in the heart, and specifically higher uptake in infarcted compared with sham-MI hearts. Histological examination at 1 week after infusion identified labeled cells either in the infarcted or border zone but not in remote viable myocardium or sham-MI hearts. Labeled cells were also identified in the lung, liver, spleen, and bone marrow. CONCLUSIONS: Systemic intravenous delivery of BM-MSCs to rats after MI, although feasible, is limited by entrapment of the donor cells in the lungs. Direct left ventricular cavity infusion enhances migration and colonization of the cells preferentially to the ischemic myocardium.


Asunto(s)
Células de la Médula Ósea , Movimiento Celular , Mesodermo/trasplante , Infarto del Miocardio/terapia , Trasplante de Células Madre , Animales , Células de la Médula Ósea/citología , Cateterismo Cardíaco , Separación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Estudios de Factibilidad , Femenino , Cámaras gamma , Corazón/diagnóstico por imagen , Corazón/fisiopatología , Pulmón/irrigación sanguínea , Pulmón/citología , Mesodermo/citología , Infarto del Miocardio/diagnóstico por imagen , Especificidad de Órganos , Cintigrafía , Ratas , Ratas Sprague-Dawley , Tecnecio , Resultado del Tratamiento , Función Ventricular
15.
Blood ; 111(12): 5422-3, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18544692
16.
Stem Cell Rev Rep ; 11(6): 826-40, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26250539

RESUMEN

Mesenchymal stem cells (MSCs) serve as supporting and regulatory cells, by providing tissues with multiple factors and are also known for their immunosuppressive capabilities. Our laboratory had previously shown that MSCs expressed toll-like receptor (TLR) 2 and are activated by its ligand Pam3Cys. TLR2 is an important component of the innate immune system, as it recognizes bacterial lipopeptides, thus priming a pro-inflammatory immune response. This study showed that Pam3Cys attached extensively to cells of both wild-type and TLR2 deficient cultured MSCs, thus, independently of TLR2. The TLR2 independent binding occurred through the adsorption of the palmitoyl moieties of Pam3Cys. It was further showed that Pam3Cys was transferred from cultured MSCs to immune cells. Moreover, Pam3Cys provided to the immune cells induced a pro-inflammatory response in vitro and in vivo. Overall, it is demonstrated herein that a TLR2 ligand bound to MSCs also through a TLR2 independent mechanism. Furthermore, the ligand incorporated by MSCs is subsequently released to stimulate an immune response both in vitro and in vivo. It is thus suggested that during bacterial infection, stromal cells may retain a reservoir of the TLR2 ligands, in a long-term manner, and release them slowly to maintain an immune response.


Asunto(s)
Lipoproteínas/metabolismo , Células Madre Mesenquimatosas/inmunología , Células Madre Mesenquimatosas/metabolismo , Receptor Toll-Like 2/agonistas , Animales , Proliferación Celular , Células Cultivadas , Femenino , Inflamación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica , Receptor Toll-Like 2/genética , Receptor Toll-Like 2/inmunología
17.
FEBS Lett ; 531(2): 109-14, 2002 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-12417296

RESUMEN

Proteins are often referred to in accordance with the activity with which they were first associated or the organelle in which they were initially identified. However, a variety of nuclear factors act in multiple molecular reactions occurring simultaneously within the nucleus. This review describes the functions of the nuclear factors PSF (polypyrimidine tract-binding protein-associated splicing factor) and p54(nrb)/NonO. PSF was initially termed a splicing factor due to its association with the second step of pre-mRNA splicing while p54(nrb)/NonO was thought to participate in transcriptional regulation. Recent evidence shows that the simplistic categorization of PSF and its homolog p54(nrb)/NonO to any single nuclear activity is not possible and in fact these proteins exhibit multi-functional characteristics in a variety of nuclear processes.


Asunto(s)
Proteínas Nucleares/fisiología , Proteínas de Unión al ARN/fisiología , Animales , Anhidrasas Carbónicas/metabolismo , Núcleo Celular/metabolismo , Replicación del ADN , Regulación de la Expresión Génica , Humanos , Ratones , Modelos Genéticos , Neoplasias/etiología , Factor de Empalme Asociado a PTB , ARN/metabolismo , Procesamiento Postranscripcional del ARN , Factores de Empalme Serina-Arginina , Transcripción Genética
18.
Ann N Y Acad Sci ; 996: 245-60, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12799303

RESUMEN

The negative control of B cell generation is only partially resolved. We assessed the role of activin A in regulation of B lymphopoiesis in view of its specific inhibitory effects on tumor B lineage cells. Activin A is constitutively expressed in mouse hemopoietic organs and in cultured mesenchymal cell lines. We observed an inverse relationship between activin A titer and B lineage cell production. In the spleen, the red pulp exhibited a relatively higher abundance of the protein as compared with the lymphoid follicles, wherein B cell accumulation occurs. Furthermore, a specific shut off in activin A expression was observed in bone marrow and spleen following in vivo induction of B lymphocyte polyclonal activation. We further substantiated these in vivo observations by in vitro studies of primary bone marrow cultures, in which the expression of functional activin A was found to be diminished prior to the onset of B lymphopoiesis. The reduction in functional activin A is shown to concomitantly occur with spontaneous induction of the expression of activin A specific inhibitors. We therefore propose that the mesenchymal organ stroma expresses activin A that negatively controls B cell lymphopoiesis.


Asunto(s)
Activinas/metabolismo , Linfocitos B/citología , Subunidades beta de Inhibinas/metabolismo , Linfopoyesis , Células del Estroma/metabolismo , Activinas/genética , Animales , Médula Ósea/metabolismo , Células Cultivadas , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Subunidades beta de Inhibinas/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Especificidad de Órganos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/metabolismo , Factores de Tiempo
19.
Stem Cells Dev ; 23(6): 676-86, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24200242

RESUMEN

Umbilical cord blood (UCB) is a good source of hematopoietic progenitors with increasing implementation in the clinical transplant setting. This study evaluates the molecular mechanisms of progenitor resistance to apoptosis triggered by Fas cross-linking. CD34(+) and lineage-negative progenitors survive short-term ex vivo incubation and are not induced into apoptosis by Fas cross-linking. Furthermore, brief exposure of UCB cells to Fas-ligand for 24-48 h does not impair quantitative severe combine immune deficiency (SCID) reconstitution activity and appears to foster myelomonocyte reconstitution. The transcriptome of Fas receptor-positive CD34(+) cells that survived an apoptotic challenge showed significant transcriptional upregulation of caspase-8, mucosa-associated lymphoid tissue lymphoma translocation gene-1 (MALT1), HtrA2, and GSK3ß in addition to higher levels of c-FLICE inhibitory protein (FLIP), Bcl-2, and cytosolic inhibitor of apoptosis protein (cIAP) in all Fas-positive cells. Most prominent is the transcriptional upregulation of several key components the NFκB1 pathway including the membrane receptors TGF-ß, interleukin-1 (IL-1), and TCR, the associated factor TNF receptor-associated factor-6 (TRAF6), and the converting enzymes TGF-ß-activated kinase-1 (TAK1), double-stranded RNA-activated protein kinase (PKR), and α-catalytic subunit of IκB kinase (IKKα), that promote activation and nuclear translocation of this transcription factor. These data indicate that hematopoietic progenitors are not insensitive to apoptosis but are actively shielded from the extrinsic and intrinsic apoptotic pathways. This may occur through inherent transcriptional upregulation of the entire NFκB pathway in the presence of competent apoptotic signaling.


Asunto(s)
Apoptosis , Proteína Ligando Fas/metabolismo , Células Madre Hematopoyéticas/citología , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Caspasas/metabolismo , Células Cultivadas , Células Madre Hematopoyéticas/metabolismo , Humanos , FN-kappa B/efectos de los fármacos , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo
20.
Stem Cell Rev Rep ; 10(3): 376-88, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24664302

RESUMEN

The outstanding heterogeneity of stem cell populations is a major obstacle on the way to their clinical application. It is therefore paramount to identify the molecular mechanisms that underlay this heterogeneity. Individually derived bone marrow mesenchymal stromal cells (MSCs) preparations, studied here, diverged markedly in various properties, despite of being all tripotent in their differentiation potential. Microarray analysis showed that MSC diversity is evident also in highly variable gene expression patterns. Differentially expressed genes were significantly enriched in toll-like receptors (TLRs) and differentiation pathways. Marked differences were observed in LPS binding protein (LBP) and transforming growth factor (TGF)ß1 expression. These differences correlated with MSC functionality. Therefore, the possible contribution of these molecules to MSC diversity was examined. In the TLR signaling pathway, LBP levels predicted the ability of specific MSCs to secrete interleukin (IL)-6 in response to LPS. A relatively higher expression of TGFß1 endowed MSCs with a capacity to respond to IL-1ß by reduced osteogenic differentiation. This study thus demonstrates major diversity within MSC isolates, which appears early on following derivation and persists following long-term culture. MSC heterogeneity results from highly variable transcriptome. Differential expression of LBP and TGFß1, along with other genes, in different MSC preparations, produces the variable responses to external stimuli.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Proteínas Portadoras/metabolismo , Interleucina-1beta/fisiología , Glicoproteínas de Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Receptores Toll-Like/agonistas , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/inmunología , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Receptores Toll-Like/metabolismo , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA