Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Leuk Lymphoma ; : 1-11, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38712673

RESUMEN

AMG 330, a bispecific T-cell engager (BiTE®) that binds CD33 and CD3 on T cells facilitates T-cell-mediated cytotoxicity against CD33+ cells. This first-in-human, open-label, dose-escalation study evaluated the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of AMG 330 in adults with relapsed/refractory acute myeloid leukemia (R/R AML). Amongst 77 patients treated with AMG 330 (0.5 µg/day-1.6 mg/day) on 14-day or 28-day cycles, maximum tolerated dose was not reached; median duration of treatment was 29 days. The most frequent treatment-related adverse events were cytokine release syndrome (CRS; 78%) and rash (30%); 10% of patients experienced grade 3/4 CRS. CRS was mitigated with stepwise dosing of AMG 330, prophylactic dexamethasone, and early treatment with tocilizumab. Among 60 evaluable patients, eight achieved complete remission or morphologic leukemia-free state; of the 52 non-responders, 37% had ≥50% reduction in AML bone marrow blasts. AMG 330 is a promising CD33-targeted therapeutic strategy for R/R AML.

2.
Am J Clin Oncol ; 45(8): 352-365, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35848749

RESUMEN

Monitoring for liver injury remains an important aspect of drug safety assessment, including for oncotherapeutics. When present, drug-induced liver injury may limit the use or result in the discontinuation of these agents. Drug-induced liver injury can exhibit with a wide spectrum of clinical and biochemical manifestations, ranging from transient asymptomatic elevations in aminotransferases (TAEAT) to acute liver failure. Numerous oncotherapeutics have been associated with TAEAT, with published reports indicating a phenomenon in which patients may be asymptomatic without overt liver injury despite the presence of grade ≥3 aminotransferase elevations. In this review, we discuss the occurrence of TAEAT in the context of oncology clinical trials and clinical practice, as well as the clinical relevance of this phenomenon as an adverse event in response to oncotherapeutics and the related cellular and molecular mechanisms that may underlie its occurrence. We also identify several gaps in knowledge relevant to the diagnosis and the management of TAEAT in patients receiving oncotherapeutics, and identify areas warranting further study to enable the future development of consensus guidelines to support clinical decision-making.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas , Neoplasias , Alanina Transaminasa/uso terapéutico , Aspartato Aminotransferasas/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Humanos , Neoplasias/tratamiento farmacológico
3.
Crit Rev Oncol Hematol ; 175: 103710, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35588936

RESUMEN

Relapse is common in acute myeloid leukemia (AML) and thought to be due to resistance of underlying leukemic stem cells (LSCs) to current standard therapies, although a lack of tools to measure the quantity and quality of these cells in patients precludes the clinical testing of this concept. This review discusses the current knowledge of LSC properties and appraises strategies aimed to bring the therapeutic targeting of LSCs to the bedside to improve patient outcomes. We highlight pathways and targets of interest and summarize available information on drugs that might eradicate LSCs. Future research is needed to close identified gaps in knowledge and provide evidence for the clinical efficacy of LSC-directed therapies to support the development of treatments that eliminate residual disease and prevent relapse, thereby increasing the cure rates of patients with AML.


Asunto(s)
Leucemia Mieloide Aguda , Células Madre Neoplásicas , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Neoplasia Residual/metabolismo , Células Madre Neoplásicas/metabolismo , Recurrencia , Investigación Biomédica Traslacional
4.
World J Stem Cells ; 7(8): 1090-108, 2015 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-26435769

RESUMEN

The complement pathway is best known for its role in immune surveillance and inflammation. However, its ability of opsonizing and removing not only pathogens, but also necrotic and apoptotic cells, is a phylogenetically ancient means of initiating tissue repair. The means and mechanisms of complement-mediated tissue repair are discussed in this review. There is increasing evidence that complement activation contributes to tissue repair at several levels. These range from the chemo-attraction of stem and progenitor cells to areas of complement activation, to increased survival of various cell types in the presence of split products of complement, and to the production of trophic factors by cells activated by the anaphylatoxins C3a and C5a. This repair aspect of complement biology has not found sufficient appreciation until recently. The following will examine this aspect of complement biology with an emphasis on the anaphylatoxins C3a and C5a.

5.
Adv Cancer Res ; 123: 149-89, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25081529

RESUMEN

The fate of both endogenous and transplanted stem cells is dependent on the functional status of the regulatory local microenvironment, which is compromised by disease and therapeutic intervention. The glycosaminoglycan hyaluronan (HA) is a critical component of the hematopoietic microenvironment. We summarize recent advances in our understanding of the role of HA in regulating mesenchymal stem cells, osteoblasts, fibroblasts, macrophages, and endothelium in bone marrow (BM) and their crosstalk within the hematopoietic microenvironment. HA not only determines the volume, hydration, and microfluidics of the BM interstitial space, but also, via interactions with specific receptors, regulates multiple cell functions including differentiation, migration, and production of regulatory factors. The effects of HA are dependent on the polymer size and are influenced by the formation of complexes with other molecules. In healthy BM, HA synthases and hyaluronidases form a molecular network that maintains extracellular HA levels within a discrete physiological window, but HA homeostasis is often perturbed in pathological conditions, including hematological malignancies. Recent studies have suggested that HA synthases may have functions beyond HA production and contribute to the intracellular regulatory machinery. We discuss a possible role for HA synthases, intracellular and extracellular HA in the malignant BM microenvironment, and resistance to therapy.


Asunto(s)
Células de la Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ácido Hialurónico/fisiología , Leucemia/metabolismo , Animales , Aorta/patología , Médula Ósea/metabolismo , Células de la Médula Ósea/citología , Diferenciación Celular , Movimiento Celular , Fibroblastos/citología , Glucuronosiltransferasa/metabolismo , Homeostasis , Humanos , Hialuronano Sintasas , Ácido Hialurónico/química , Macrófagos/citología , Ratones , Ratones Noqueados , Músculo Liso/citología , Osteoblastos/citología , Osteoclastos/citología , Polímeros/química , Unión Proteica , Células Madre/citología , Factores de Tiempo
6.
Stem Cells Transl Med ; 3(2): 229-40, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24396034

RESUMEN

Human neural stem cells (hNSCs) hold great potential for treatment of a wide variety of neurodegenerative and neurotraumatic conditions. Heretofore, administration has been through intracranial injection or implantation of cells. Because neural stem cells are capable of migrating to the injured brain from the intravascular space, it seemed feasible to administer them intravenously if their ability to circumvent the blood-brain barrier was enhanced. In the present studies, we found that interactions of hNSCs in vitro on the luminal surface of human umbilical vein endothelial cells was enhanced following enforced expression of cutaneous lymphocyte antigen on cell surface moieties by incubation of hNSCs with fucosyltransferase VI and GDP-fucose (fhNSCs). Interestingly, ex vivo fucosylation of hNSCs not only did not improve the cells homing into the brain injured by stroke following intravenous administration but also increased mortality of rats compared with the nonfucosylated hNSC group. Efforts to explain these unexpected findings using a three-dimensional flow chamber device revealed that transmigration of fhNSCs (under conditions of physiological shear stress) mediated by stromal cell-derived factor 1α was significantly decreased compared with controls. Further analysis revealed that hNSCs poorly withstand physiological shear stress, and their ability is further decreased following fucosylation. In addition, fhNSCs demonstrated a higher frequency of cellular aggregate formation as well as a tendency for removal of fucose from the cell surface. In summary, our findings suggest that the behavior of hNSCs in circulation is different from that observed with other cell types and that, at least for stroke, intravenous administration is a suboptimal route, even when the in vitro rolling ability of hNSCs is optimized by enforced fucosylation.


Asunto(s)
Barrera Hematoencefálica/citología , Células Endoteliales/citología , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Accidente Cerebrovascular/terapia , Venas/citología , Animales , Comunicación Celular , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Cámaras de Difusión de Cultivos , Modelos Animales de Enfermedad , Células Endoteliales/fisiología , Femenino , Fucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inyecciones Intravenosas , Células-Madre Neurales/fisiología , Ratas , Ratas Sprague-Dawley , Estrés Mecánico , Accidente Cerebrovascular/patología , Venas/fisiología
7.
J Vis Exp ; (77): e50959, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23893091

RESUMEN

Extravasation of circulating cells from the bloodstream plays a central role in many physiological and pathophysiological processes, including stem cell homing and tumor metastasis. The three-dimensional flow chamber device (hereafter the 3D device) is a novel in vitro technology that recreates physiological shear stress and allows each step of the cell extravasation cascade to be quantified. The 3D device consists of an upper compartment in which the cells of interest circulate under shear stress, and a lower compartment of static wells that contain the chemoattractants of interest. The two compartments are separated by porous inserts coated with a monolayer of endothelial cells (EC). An optional second insert with microenvironmental cells of interest can be placed immediately beneath the EC layer. A gas exchange unit allows the optimal CO2 tension to be maintained and provides an access point to add or withdraw cells or compounds during the experiment. The test cells circulate in the upper compartment at the desired shear stress (flow rate) controlled by a peristaltic pump. At the end of the experiment, the circulating and migrated cells are collected for further analyses. The 3D device can be used to examine cell rolling on and adhesion to EC under shear stress, transmigration in response to chemokine gradients, resistance to shear stress, cluster formation, and cell survival. In addition, the optional second insert allows the effects of crosstalk between EC and microenvironmental cells to be examined. The translational applications of the 3D device include testing of drug candidates that target cell migration and predicting the in vivo behavior of cells after intravenous injection. Thus, the novel 3D device is a versatile and inexpensive tool to study the molecular mechanisms that mediate cellular extravasation.


Asunto(s)
Movimiento Celular/fisiología , Quimiocinas/farmacología , Técnicas Citológicas/instrumentación , Células Endoteliales/citología , Animales , Células de la Médula Ósea/citología , Técnicas Citológicas/métodos , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Ratones
8.
Immunology ; 135(4): 287-98, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22117697

RESUMEN

The observation that human monocytes cultured in the presence of the chemokine CCL18 showed increased survival, led us to profile cytokine expression in CCL18-stimulated versus control cultures. CCL18 caused significantly increased expression of chemokines (CXCL8, CCL2, CCL3 and CCL22), interleukin-10 (IL-10) and platelet-derived growth factor, but no up-regulation of M1 cytokines IL-1ß or IL-12. CCL18-stimulated monocytes matured into cells with morphological resemblance to IL-4-stimulated macrophages, and expressed the monocyte marker CD14 as well the M2 macrophage markers CD206 and 15-lipoxygenase, but no mature dendritic cell markers (CD80, CD83 or CD86). Functionally, CCL18-stimulated macrophages showed a high capacity for unspecific phagocytosis and for pinocytosis, which was not associated with an oxidative burst. These findings suggest that CCL18-activated macrophages stand at the cross-roads between inflammation and its resolution. The chemokines that are produced in response to CCL18 are angiogenic and attract various leucocyte populations, which sustain inflammation. However, the capacity of these cells to remove cellular debris without causing oxidative damage and the production of the anti-inflammatory IL-10 will initiate termination of the inflammatory response. In summary, CCL18 induces an M2 spectrum macrophage phenotype in the absence of IL-4.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Quimiocinas CC/farmacología , Macrófagos/inmunología , Monocitos/citología , Animales , Células Cultivadas , Quimiocinas/metabolismo , Quimiocinas CC/inmunología , Cobayas , Humanos , Interleucina-10/metabolismo , Macrófagos/citología , Ratones , Monocitos/efectos de los fármacos , Monocitos/inmunología , Fagocitosis
9.
Protein Expr Purif ; 79(1): 72-80, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21550406

RESUMEN

An expression method has been developed to produce soluble cationic polypeptides in Escherichia coli while avoiding inclusion body deposition. For this technique the recombinant product is linked through a thrombin or factor Xa susceptible bond to the amino-terminal domain of the precursor of eosinophil major basic protein (MBP). This N-terminal domain is strongly acidic and is apparently able to shield eosinophils from the potentially injurious activities of MBP. It was reasoned that constructs of this acidic domain with small heterologous cationic proteins expressed in E. coli could result in soluble expression while preventing trafficking and packaging into insoluble inclusion bodies. This has been demonstrated using four examples: complement C5a, CCL18, fibroblast growth factor-ß, and leukemia inhibitory factor, whose isoelectric points range from 8.93 to 9.59. Further general applicability of this technique has been shown by using two different expression systems, one which encodes an amino-terminal oligo-histidine leash, and another that codes for an amino-terminal glutathione-S-transferase. Thus the utility of coupling MAP to cationic polypeptides for the purpose of soluble heterologous protein expression in E. coli has been demonstrated.


Asunto(s)
Clonación Molecular/métodos , Proteína Mayor Básica del Eosinófilo/genética , Escherichia coli/genética , Proteínas Recombinantes de Fusión/genética , Quimiocinas CC/genética , Quimiocinas CC/aislamiento & purificación , Complemento C5a/genética , Complemento C5a/aislamiento & purificación , Proteína Mayor Básica del Eosinófilo/aislamiento & purificación , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/aislamiento & purificación , Expresión Génica , Células HEK293 , Humanos , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/aislamiento & purificación , Proteínas Recombinantes de Fusión/aislamiento & purificación , Solubilidad
10.
Mol Cancer Ther ; 9(11): 3024-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20833754

RESUMEN

Bone marrow hypoplasia and pancytopenia are among the most undesirable sequelae of chemotherapy for the treatment of cancer. We recently showed that hyaluronan (HA) facilitates hematopoietic recovery in tumor-free animals receiving chemotherapeutic agents. However, following a chemotherapeutic regimen in tumor-bearing animals, it is possible that residual tumor cells might respond to systemic injections of HA. Thus, in this study, we investigated the effect of HA on the regrowth of residual tumor cells following chemotherapy. As a model, we used the HCT-8 human colon carcinoma cell line, which expresses the HA receptor CD44, binds exogenous HA, and is susceptible to a chemotherapy protocol containing irinotecan and 5-fluorouracil in a human/mouse xenograft model. HCT-8 cells were implanted in severe combined immunodeficient mice, followed by irinotecan/5-fluorouracil treatment. After three rounds of chemotherapy, residual tumors were allowed to regrow in the presence or absence of HA. The dynamics of tumor regrowth in the group treated with HA was slower compared with the control group. By week 5 after tumor implantation, the difference in the size of regrown tumors was statistically significant and correlated with lower proliferation and higher apoptosis in HA-treated tumors as compared with controls. This finding provides evidence that HA treatment does not stimulate but delays the growth of residual cancer cells, which is an important parameter in establishing whether the use of HA can enhance current chemotherapeutic strategies.


Asunto(s)
Carcinoma/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Ácido Hialurónico/farmacología , Recurrencia Local de Neoplasia/prevención & control , Animales , Carcinoma/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quimioterapia Adyuvante , Neoplasias del Colon/patología , Citostáticos/administración & dosificación , Citostáticos/farmacología , Esquema de Medicación , Femenino , Humanos , Ácido Hialurónico/administración & dosificación , Ratones , Ratones SCID , Modelos Biológicos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Stem Cells ; 5(1): 9-21, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20861924

RESUMEN

Hyaluronan (HA) is an important component of the microenvironment in bone marrow, but its role in regulation of the development of hematopoietic cells is not well understood. To address the role of HA in regulation of human embryonic stem cell (hESC) differentiation into the hematopoietic lineage, we screened for genes encoding components of the HA pathway. Using gene arrays, we found that HA synthases and HA receptors are expressed in both undifferentiated and differentiating hESCs. Enzymatic degradation of HA resulted in decreased numbers of hematopoietic progenitors and lower numbers of CD45+ cells generated in HA-deprived embryoid bodies (EBs). In addition, deprivation of HA resulted in the inhibition of generation of CD31+ cells, stromal fibroblast-like cells and contracting myocytes in EBs. RT-PCR and immunocytochemistry revealed that HA deprivation did not influence the dynamics of OCT4 expression, but decreased the expression of BRY, an early mesoderm marker, and BMP2, a later mesoderm marker in differentiating EBs. In addition, the endoderm markers α-FP and SOX17 were decreased, whereas the expression of the ectoderm markers GFAP and FGF5 was higher in HA-deprived cultures. Our findings indicate that endogenously produced HA contributes to the network that regulates the differentiation of hESC and the generation of mesodermal lineage in general and hematopoietic cells specifically.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Sistema Hematopoyético/citología , Ácido Hialurónico/fisiología , Biomarcadores/metabolismo , Linaje de la Célula , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/metabolismo , Humanos , Técnicas para Inmunoenzimas , Mesodermo/citología , Mesodermo/efectos de los fármacos , Mesodermo/metabolismo , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
J Immunol ; 182(6): 3827-36, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19265162

RESUMEN

Mesenchymal stem cells (MSCs) have a great potential for tissue repair, especially if they can be delivered efficiently to sites of tissue injury. Since complement activation occurs whenever there is tissue damage, the effects of the complement activation products C3a and C5a on MSCs were examined. Both C3a and C5a were chemoattractants for human bone marrow-derived MSCs, which expressed both the C3a receptor (C3aR) and the C5a receptor (C5aR; CD88) on the cell surface. Specific C3aR and C5aR inhibitors blocked the chemotactic response, as did pertussis toxin, indicating that the response was mediated by the known anaphylatoxin receptors in a G(i) activation-dependent fashion. While C5a causes strong and prolonged activation of various signaling pathways in many different cell types, the response observed with C3a is generally transient and weak. However, we show herein that in MSCs both C3a and C5a caused prolonged and robust ERK1/2 and Akt phosphorylation. Phospho-ERK1/2 was translocated to the nucleus in both C3a and C5a-stimulated MSCs, which was associated with subsequent phosphorylation of the transcription factor Elk, which could not be detected in other cell types stimulated with C3a. More surprisingly, the C3aR itself was translocated to the nucleus in C3a-stimulated MSCs, especially at low cell densities. Since nuclear activation/translocation of G protein-coupled receptors has been shown to induce long-term effects, this novel observation implies that C3a exerts far-reaching consequences on MSC biology. These results suggest that the anaphylatoxins C3a and C5a present in injured tissues contribute to the recruitment of MSCs and regulation of their behavior.


Asunto(s)
Factores Quimiotácticos/fisiología , Complemento C3a/fisiología , Complemento C5a/fisiología , Células Madre Mesenquimatosas/enzimología , Células Madre Mesenquimatosas/inmunología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Línea Celular , Células Cultivadas , Quimiotaxis de Leucocito/inmunología , Humanos , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Anafilatoxina C5a , Receptores de Complemento/biosíntesis , Receptores de Complemento/genética , Factores de Tiempo
13.
J Stem Cells ; 4(3): 147-60, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20232600

RESUMEN

Embryonic stem cells (ESCs) offer a powerful in vitro model to study mechanisms implicated in cell fate decision. Developmental pathways by which pluripotent ESCs become committed to specific lineages are reflected in dynamic changes of signaling and transcriptional programs. However, the mechanisms that govern the regulatory intracellular networks underlying lineage fate decisions and differentiation programs remain poorly understood and differ significantly in different species. In this review we analyze the current understanding of the signaling mechanisms and transcriptional regulation of differentiation of murine and human ESCs into the mesoderm.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Células Madre Mesenquimatosas/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Humanos , Transducción de Señal/genética , Factores de Transcripción/metabolismo
14.
J Stem Cells ; 4(4): 191-202, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20720593

RESUMEN

Hyaluronan (HA) is expressed by cells in bone marrow where it contributes to the regulation of hematopoietic homeostasis. In this study, we have demonstrated that exogenous low molecular weight HA (LMW HA) polymers mobilize leukocytes, but not hematopoietic progenitor cells, to peripheral blood within a 3 hour time period following HA administration. Mobilization of leukocytes correlated with increased extracellular MMP-9 concentrations induced by LMW HA, but not high molecular weight (HMW) HA. In contrast, HMW HA up-regulated TIMP-1 expression in bone marrow cells. In vitro, HMW HA did not influence SDF-1 - mediated chemotaxis of hematopoietic progenitors, whereas LMW HA polymers demonstrated inhibitory activity. These findings suggest that the effects of HA on cell motility depend on the size of the HA polymers and on the type of target cells.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Ácido Hialurónico/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Movimiento Celular/fisiología , Células Cultivadas , Glucuronosiltransferasa/metabolismo , Células Madre Hematopoyéticas/fisiología , Receptores de Hialuranos/metabolismo , Hialuronano Sintasas , Ácido Hialurónico/química , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C
15.
J Stem Cells ; 4(4): 203-15, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20720594

RESUMEN

The efficient migration of mesenchymal stem cells (MSCs) to diseased tissues is required for the fulfillment of their regenerative potential. Recruitment of circulating cells into the damaged tissues is regulated by a complex network, which includes the non-neural cholinergic system. We found that human MSCs (hMSCs) express nicotinic acetylcholine receptor subunits alpha 7, beta 2 and beta 4. The receptor agonist nicotine caused calcium (Ca(++)) influx into hMSCs suggesting that the calcium ion channel alpha 7 homopolymer mediated this response. While high concentrations of nicotine (10(5)M) induced hMSC apoptosis, physiological concentrations (10(7)M) did not interfere with cell survival. At non-toxic concentrations, nicotine increased spontaneous migration of hMSCs, whereas chemotaxis of hMSCs toward C3a and bFGF in vitro and migration of intravenously infusion hMSCs into bone marrow and spleen in vivo were inhibited. The antagonist for the alpha 7 homopolymer, bungarotoxin, blocked the inhibitory effect of nicotine on chemotactic factor-induced migration of hMSCs. These findings reveal an involvement of the non-neural cholinergic system in regulation of hMSC migration.


Asunto(s)
Movimiento Celular/fisiología , Células Madre Mesenquimatosas/fisiología , Subunidades de Proteína/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas/farmacología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Nicotina/metabolismo , Nicotina/farmacología , Agonistas Nicotínicos/metabolismo , Agonistas Nicotínicos/farmacología , Subunidades de Proteína/genética , Receptores Nicotínicos/genética , Receptor Nicotínico de Acetilcolina alfa 7
16.
Stem Cells ; 24(11): 2367-72, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17071855

RESUMEN

Understanding the mechanisms by which stem cells home precisely to regions of injury or degeneration is of importance to both basic and applied regenerative medicine. Optimizing regenerative processes may depend on identifying the range of molecules that subserve stem cell trafficking. The "rolling" of extravasating cells on endothelium under conditions of physiological flow is the first essential step in the homing cascade and determines cell adhesion and transmigration. Using a laminar flow chamber to simulate physiological shear stress, we explored an aspect of this process by using human neural stem cells (hNSCs). We observed that the interactions between hNSCs and tumor necrosis factor-alpha (TNF-alpha)-stimulated human endothelium (simulating an inflamed milieu) are mediated by a subclass of integrins--alpha2, alpha6, and beta1, but not alpha4, alphav, or the chemokine-mediated pathway CXCR4-stromal cell-derived factor-1alpha--suggesting not only that the mechanisms mediating hNSC homing via the vasculature differ from the mechanisms mediating homing through parenchyma, but also that each step invokes a distinct pathway mediating a specialized function in the hNSC homing cascade. (TNF-alpha stimulation also upregulates vascular cell adhesion molecule-1 expression on the hNSCs themselves and increases NSC-endothelial interactions.) The selective use of integrin subgroups to mediate homing of cells of neuroectodermal origin may also be used to ensure that cells within the systemic circulation are delivered to the pathological region of a given organ to the exclusion of other, perhaps undesired, organs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Inflamación/metabolismo , Integrinas/metabolismo , Neuronas/metabolismo , Adhesión Celular/efectos de los fármacos , Línea Celular , Movimiento Celular/efectos de los fármacos , Células Madre Embrionarias/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Humanos , Inmunohistoquímica , Inflamación/genética , Integrina alfa2/metabolismo , Integrina alfa6/metabolismo , Integrina beta1/metabolismo , Neuronas/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Mecánico , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/biosíntesis , Molécula 1 de Adhesión Celular Vascular/inmunología
17.
Blood ; 108(12): 3722-9, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16888095

RESUMEN

Chemokines play a role in regulating hematopoietic stem cell function, including migration, proliferation, and retention. We investigated the involvement of CCL18 in the regulation of bone marrow hematopoiesis. Treatment of human long-term bone marrow cultures (LTBMCs) with CCL18 resulted in significant stimulation of hematopoiesis, as measured by the total number of hematopoietic cells and their committed progenitors produced in culture. Monocytes/macrophages, whose survival was almost doubled in the presence of CCL18 compared with controls, were the primary cells mediating this effect. Conditioned media from CCL18-treated mature monocytes fostered colony-promoting activity that increased the number of colonies formed by hematopoietic progenitor cells. Gene expression profiling of CCL18-stimulated monocytes demonstrated more than 200 differentially expressed genes, including those regulating apoptosis (caspase-8) and proliferation (IL-6, IL-15, stem cell factor [SCF]). Up-regulation of these cytokines was confirmed on the protein expression level. The contribution of SCF and IL-6 in CCL18-mediated stimulatory activity for hematopoiesis was confirmed by SCF- and IL-6-blocking antibodies that significantly inhibited the colony-promoting activity of CCL18-stimulated conditioned medium. In addition to the effect on monocytes, CCL18 facilitated the formation of the adherent layer in LTBMCs and increased the proliferation of stromal fibroblast-like cells.


Asunto(s)
Médula Ósea/metabolismo , Quimiocinas CC/metabolismo , Hematopoyesis/fisiología , Monocitos/metabolismo , Biosíntesis de Proteínas/fisiología , Regulación hacia Arriba/fisiología , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimiocinas CC/farmacología , Ensayo de Unidades Formadoras de Colonias/métodos , Medios de Cultivo Condicionados , Citocinas/biosíntesis , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica/métodos , Hematopoyesis/efectos de los fármacos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Biosíntesis de Proteínas/efectos de los fármacos , Células Madre/citología , Células Madre/metabolismo , Células del Estroma/citología , Células del Estroma/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
Br J Haematol ; 129(2): 257-65, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15813854

RESUMEN

The function of endothelial cells that contribute to the regulation of haematopoietic stem/progenitor cells (HSPC) migration from peripheral blood into bone marrow can be influenced by extrinsic factors including nicotine. Therefore, the effect of nicotine on HSPC extravasation was studied. Using a parallel laminar flow chamber, we demonstrated an increase in the number of HSPC adhering to the nicotine-exposed endothelium under conditions of physiological shear stress in vitro. Nicotine-induced adhesion of HSPC was inhibited by mecamylamine, a non-selective nicotinic acetylcholine receptor (nAchR) antagonist. The enhanced adhesive interactions of HSPC with nicotine-exposed endothelial monolayers coincided with the nicotine-induced activation of endothelial cells. Nicotine induced fast cytoskeletal reorganization and formation of filopodia in endothelial cells through interaction with the non-neuronal nAchR expressed by these cells. In addition, nicotine treatment stimulated rapid phosphorylation of Erk1/2 and p-38 in endothelial cells. Finally, nicotine inhibited the stroma derived factor-1-mediated transendothelial migration of HSPC. Decreased migration of HSPC correlated with diminished matrix metalloproteinase-9 activity secreted by bone marrow cells and decreased expression of CD44 on the surface of endothelial cells. Overall, our data suggest that exposure to nicotine causes endothelial cell dysfunction and leads to the pathological arrest of HSPC on endothelium, interfering with their proper migration process.


Asunto(s)
Células Endoteliales/metabolismo , Células Madre Hematopoyéticas/metabolismo , Nicotina/farmacología , Receptores Colinérgicos/metabolismo , Animales , Western Blotting/métodos , Adhesión Celular , Movimiento Celular , Técnicas de Cocultivo , Ensayo de Unidades Formadoras de Colonias , Electroforesis en Gel de Poliacrilamida , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C , Microscopía Fluorescente , Estrés Mecánico
19.
Stem Cells Dev ; 14(1): 81-91, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15725747

RESUMEN

Environmental factors, including cigarette smoke components, can cross the placental barrier and accumulate in amniotic fluid and fetal tissue, and, therefore, interfere with the normal course of ontogenesis. Although cigarette smoke contains numerous compounds, the most adverse effects on mammalian tissues have been associated with nicotine. The aim of this study was to investigate the effect of intrauterine exposure to nicotine on hematopoiesis during fetal development and postpartum. Intrauterine exposure of mice to nicotine resulted in a more than two-fold reduction of the delayed- type hypersensitivity (DTH) response and a 2.5-fold decrease in the number of plaque forming cell (PFC) in offspring after 1 month of postnatal life, and correlated with low counts of mature lymphocytes and lymphoid progenitors in hematopoietic tissues. Neonates exposed to nicotine during gestation showed a significant decrease in the number of bone marrow hematopoietic progenitors, as measured by colony-forming unit (CFU) and long-term culture initiating cell (LTC-IC) assays, and decreased concentration of interleukin-6 (IL-6) in their serum. Analysis of the fetal bone marrow (E15) obtained from nicotine-exposed fetuses demonstrated a lower number of hematopoietic progenitors, whereas their number in the fetal liver was not significantly changed. Our data provide evidence that by targeting the nicotinic acetylcholine receptor (nAChR) nicotine interferes with the fetal development of the hematopoietic system. Inferior colonization of the fetal bone marrow by hematopoietic stem/progenitor cells (HSPC) subsequently results in an imbalance of mature blood and immune cell production after birth.


Asunto(s)
Médula Ósea/efectos de los fármacos , Feto/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Intercambio Materno-Fetal , Nicotina/efectos adversos , Animales , Médula Ósea/embriología , Células de la Médula Ósea/citología , Femenino , Desarrollo Fetal/efectos de los fármacos , Feto/citología , Células Madre Hematopoyéticas/citología , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/embriología , Interleucina-6/deficiencia , Ratones , Embarazo
20.
Stem Cells ; 22(4): 544-55, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15277700

RESUMEN

The fate of hematopoietic stem cells (HSCs) is determined by microenvironmental niches, but the molecular structure of these local networks is not yet completely characterized. Our recent observation that glycosaminoglycan hyaluronic acid (HA), a major component of the bone marrow extracellular matrix, is required for in vitro hematopoiesis led us to suggest a role for HA in structuring the hematopoietic niche. Accordingly, HA deprivation induced by various treatments might lead to an imbalance of normal HSC homeostasis. Since 5-fluorouracil (5-FU) administration sharply decreases the amount of cell surface-associated HA in bone marrow, we examined whether the administration of exogenous HA enhances suppressed hematopoiesis in 5-FU-treated mice. HA administered to mice following 5-FU infusion facilitated the recovery of leukocytes and thrombocytes in the peripheral blood. Intravenously infused HA was found in the bone marrow, where it bound endothelial cells and resident macrophages and increased expression of the hematopoiesis-supportive cytokines interleukin-1 and interleukin-6. In agreement with these observations, enhanced hematopoietic activity was detected in the bone marrow, as measured by elevated counts of long-term culture-initiating cells (LTC-ICs), committed progenitors, and the total number of mature bone marrow cells. Overall, our results suggest that HA is required for regulation of the hematopoiesis-supportive function of bone marrow accessory cells and, therefore, participates in hematopoietic niche assembly.


Asunto(s)
Fluorouracilo/farmacología , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Ácido Hialurónico/farmacología , Ácido Hialurónico/fisiología , Animales , Técnicas de Cultivo de Célula , Ensayo de Unidades Formadoras de Colonias , Recuento de Eritrocitos , Femenino , Hematócrito , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA