Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Leuk Lymphoma ; 65(9): 1281-1291, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38712673

RESUMO

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.


Assuntos
Anticorpos Biespecíficos , Leucemia Mieloide Aguda , Humanos , Masculino , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/patologia , Leucemia Mieloide Aguda/diagnóstico , Feminino , Pessoa de Meia-Idade , Adulto , Idoso , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/uso terapêutico , Resultado do Tratamento , Adulto Jovem , Dose Máxima Tolerável , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Lectina 3 Semelhante a Ig de Ligação ao Ácido Siálico/metabolismo , Recidiva , Idoso de 80 Anos ou mais , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/patologia , Relação Dose-Resposta a Droga , Síndrome da Liberação de Citocina/etiologia
2.
J Biol Chem ; 287(30): 25419-33, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22654110

RESUMO

The contribution of hyaluronan (HA) to the regulatory network of the hematopoietic microenvironment was studied using knock-out mice of three hyaluronan synthase genes (Has1, Has2, and Has3). The number of hematopoietic progenitors was decreased in bone marrow and increased in extramedullary sites of Prx1-Cre;Has2(flox/flox);Has1(-/-);Has3(-/-) triple knock-out (tKO) mice as compared with wild type (WT) and Has1(-/-);Has3(-/-) double knock-out (dKO) mice. In line with this observation, decreased hematopoietic activity was observed in long term bone marrow cultures (LTBMC) from tKO mice, whereas the formation of the adherent layer and generation of hematopoietic cells in WT and dKO cultures was not different. 4-Methylumbelliferone (4MU) was used to pharmacologically inhibit the production of HA in LTBMC. Treatment with 4MU inhibited HA synthesis, decreased expression of HAS2 and HAS3, and eliminated hematopoiesis in LTBMC, and this effect was alleviated by the addition of exogenous HA. Exogenous HA also augmented the cell motility in LTBMC, which correlated with the HA-stimulated production of chemokines and growth factors. Conditioned media from HA-induced LTBMC enhanced the chemotaxis of hematopoietic stem/progenitor cells (HSPC) in response to SDF-1. Exposure of endothelial cells to 4MU decreased their ability to support HSPC rolling and adhesion. In addition, migration of transplanted HSPC into the marrow of 4MU-pretreated mice was lower than in untreated mice. Collectively, the results suggest that HA depletion reduces the ability of the microenvironment to support HSPC, and confirm a role for HA as a necessary regulatory element in the structure of the hematopoietic microenvironment.


Assuntos
Medula Óssea/metabolismo , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Ácido Hialurônico/metabolismo , Nicho de Células-Tronco/fisiologia , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Células Cultivadas , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Quimiotaxia/efeitos dos fármacos , Quimiotaxia/fisiologia , Glucuronosiltransferase/genética , Glucuronosiltransferase/metabolismo , Hematopoese/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Humanos , Hialuronan Sintases , Ácido Hialurônico/genética , Himecromona/análogos & derivados , Himecromona/farmacologia , Camundongos , Camundongos Knockout , Nicho de Células-Tronco/efeitos dos fármacos
3.
Blood ; 118(20): 5622-30, 2011 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-21940822

RESUMO

Localization of plasmin on macrophages and activation of pro-MMP-9 play key roles in macrophage recruitment in the inflammatory response. These functions are promoted by plasminogen receptors exposing C-terminal basic residues on the macrophage surface. Recently, we identified a novel transmembrane plasminogen receptor, Plg-R(KT), which exposes a C-terminal lysine on the cell surface. In the present study, we investigated the role of Plg-R(KT) in macrophage invasion, chemotactic migration, and recruitment. Plg-R(KT) was prominently expressed in membranes of human peripheral blood monocytes and monocytoid cells. Plasminogen activation by urokinase-type plasminogen activator (uPA) was markedly inhibited (by 39%) by treatment with anti-Plg-R(KT) mAb. Treatment of monocytes with anti-Plg-R(KT) mAb substantially inhibited invasion through the representative matrix, Matrigel, in response to MCP-1 (by 54% compared with isotype control). Furthermore, chemotactic migration was also inhibited by treatment with anti-Plg-R(KT) mAb (by 64%). In a mouse model of thioglycollate-induced peritonitis, anti-Plg-R(KT) mAb markedly inhibited macrophage recruitment (by 58%), concomitant with a reduction in pro-MMP-9 activation in the inflamed peritoneum. Treatment with anti-Plg-R(KT) mAb did not further reduce the low level of macrophage recruitment in plasminogen-null mice. We conclude that Plg-R(KT) plays a key role in the plasminogen-dependent regulation of macrophage invasion, chemotactic migration, and recruitment in the inflammatory response.


Assuntos
Movimento Celular/imunologia , Macrófagos/citologia , Peritonite/imunologia , Plasminogênio/imunologia , Receptores de Superfície Celular/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Materiais Biocompatíveis , Movimento Celular/efeitos dos fármacos , Colágeno , Modelos Animais de Doenças , Combinação de Medicamentos , Feminino , Fibrinolisina/metabolismo , Fibrinolisina/farmacologia , Humanos , Laminina , Macrófagos/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Monócitos/citologia , Monócitos/imunologia , Peritonite/induzido quimicamente , Peritonite/metabolismo , Plasminogênio/genética , Plasminogênio/metabolismo , Proteoglicanas , Receptores de Superfície Celular/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/imunologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Tioglicolatos/toxicidade
4.
Immunology ; 135(4): 287-98, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22117697

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Quimiocinas CC/farmacologia , Macrófagos/imunologia , Monócitos/citologia , Animais , Células Cultivadas , Quimiocinas/metabolismo , Quimiocinas CC/imunologia , Cobaias , Humanos , Interleucina-10/metabolismo , Macrófagos/citologia , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , Fagocitose
5.
J Biomed Biotechnol ; 2012: 250464, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23125524

RESUMO

When plasminogen binds to cells its activation to plasmin is markedly enhanced compared to the reaction in solution. Thus, cells become armed with the broad spectrum proteolytic activity of plasmin. Cell-surface plasmin plays a key role in macrophage recruitment during the inflammatory response. Proteins exposing basic residues on the cell surface promote plasminogen activation on eukaryotic cells. We have used a proteomics approach combining targeted proteolysis with carboxypeptidase B and multidimensional protein identification technology, MudPIT, and a monocyte progenitor cell line to identify a novel transmembrane protein, the plasminogen receptor, Plg-R(KT). Plg-R(KT) exposes a C-terminal lysine on the cell surface in an orientation to bind plasminogen and promote plasminogen activation. Here we review the characteristics of this new protein, with regard to membrane topology, conservation of sequence across species, the role of its C-terminus in plasminogen binding, its function in plasminogen activation, cell migration, and its role in macrophage recruitment in the inflammatory response.


Assuntos
Macrófagos/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Sequência de Aminoácidos , Animais , Sequência Conservada , Humanos , Inflamação/metabolismo , Inflamação/patologia , Lisina/metabolismo , Macrófagos/citologia , Dados de Sequência Molecular , Plasminogênio/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/química , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética
6.
Cureus ; 14(4): e24053, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35573501

RESUMO

Introduction The elevation of aminotransferase levels is regarded as an indicator of hepatocellular injury. The objective of this study was to describe real-world incidence of elevated aminotransferase levels with or without bilirubin elevation among patients treated with immune checkpoint inhibitors (ICIs) for solid tumors. Methods This retrospective cohort study used an electronic health record database representing > 1.5 million active United States (US) cancer patients and included patients diagnosed with any cancer between January 1, 2014 and March 31, 2019, and treated with one or more ICIs such as ipilimumab, tremelimumab, nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab. The frequency, onset, duration, management of grade ≥ 3 elevation of aminotransferase levels with or without bilirubin elevation events, progression rate from isolated elevation of aminotransferase levels (IAT) to elevated aminotransferase levels with elevated bilirubin (ATWB), and mortality were described. Results Overall, 69,140 patients received 85,433 treatment courses. A total of 1,799 (2.11%) IAT and 441 (0.52%) ATWB events were observed during treatment courses. The median onset was 51 and 42 days for IAT and ATWB, respectively, across treatment courses, and the median duration of both was approximately seven days. Approximately 5% (n=96) of IAT events progressed to ATWB in a median time of 11 days. The proportion of patients who received corticosteroids after elevated aminotransferase levels with or without bilirubin was ~37% (n=671/1,799 of IAT and n=147/441 of ATWB) and ~8% discontinued ICI treatment (n=118/1,799 of IAT and n=43/441 of ATWB). About 46% (n=68/147) of ATWB and and 25% (n=172/671) of IAT events treated with steroids led to death within 45 days. Similarly, 49% (n=21/43) of ATWB and 35% (n=42/118) of IAT events leading to treatment discontinuation led to death within 45 days. Conclusions Real-world data from oncology clinics in US suggest low incidence of grade ≥ 3 elevated aminotransferase levels with or without bilirubin elevation following treatment with ICIs. In most cases, ICI treatment was not discontinued and management of elevated aminotransferases consisted of corticosteroid treatment in one-third of cases.

7.
Crit Rev Oncol Hematol ; 175: 103710, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35588936

RESUMO

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.


Assuntos
Leucemia Mieloide Aguda , Células-Tronco Neoplásicas , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Neoplasia Residual/metabolismo , Células-Tronco Neoplásicas/metabolismo , Recidiva , Pesquisa Translacional Biomédica
8.
Am J Clin Oncol ; 45(8): 352-365, 2022 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-35848749

RESUMO

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.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Neoplasias , Alanina Transaminase/uso terapêutico , Aspartato Aminotransferases/uso terapêutico , Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Humanos , Neoplasias/tratamento farmacológico
9.
Protein Expr Purif ; 79(1): 72-80, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21550406

RESUMO

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.


Assuntos
Clonagem Molecular/métodos , Proteína Básica Maior de Eosinófilos/genética , Escherichia coli/genética , Proteínas Recombinantes de Fusão/genética , Quimiocinas CC/genética , Quimiocinas CC/isolamento & purificação , Complemento C5a/genética , Complemento C5a/isolamento & purificação , Proteína Básica Maior de Eosinófilos/isolamento & purificação , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/isolamento & purificação , Expressão Gênica , Células HEK293 , Humanos , Fator Inibidor de Leucemia/genética , Fator Inibidor de Leucemia/isolamento & purificação , Proteínas Recombinantes de Fusão/isolamento & purificação , Solubilidade
10.
J Immunol ; 182(6): 3827-36, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265162

RESUMO

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.


Assuntos
Fatores Quimiotáticos/fisiologia , Complemento C3a/fisiologia , Complemento C5a/fisiologia , Células-Tronco Mesenquimais/enzimologia , Células-Tronco Mesenquimais/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Linhagem Celular , Células Cultivadas , Quimiotaxia de Leucócito/imunologia , Humanos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor da Anafilatoxina C5a , Receptores de Complemento/biossíntese , Receptores de Complemento/genética , Fatores de Tempo
11.
Methods ; 45(2): 159-67, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18593612

RESUMO

This review is focused on methods that are used to derive hematopoietic cells from embryonic stem cells (ESCs). One of the strategies that have been recently used to achieve this goal is an approach of mimicking the hematopoietic niche in vitro by using hematopoiesis-supportive feeder cells, cocktails of soluble hematopoietic growth factors and a variety of matrices. While there is clear evidence that it is possible to derive hematopoietic stem cells (HSCs) and subsequently committed hematopoietic progenitors and mature cells from ESCs, there remains the need to address multiple issues including the efficiency of HSCs derivation in vitro and their proper functionality.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , Linhagem da Célula/genética , Separação Celular/métodos , Meios de Cultura/farmacologia , Meios de Cultura/normas , Cultura em Câmaras de Difusão , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/fisiologia , Hematopoese , Sistema Hematopoético/fisiologia , Humanos , Camundongos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos
12.
Life Sci ; 80(24-25): 2352-60, 2007 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-17512954

RESUMO

Gene expression profiling demonstrated that components of the cholinergic system, including choline acetyltransferase, acetylcholinesterase and nicotinic acetylcholine receptors (nAChRs), are expressed in embryonic stem cells and differentiating embryoid bodies (EBs). Triggering of nAChRs expressed in EBs by nicotine resulted in activation of MAPK and shifts of spontaneous differentiation toward hemangioblast. In vivo, non-neural nAChRs are detected early during development in fetal sites of hematopoiesis. Similarly, in vivo exposure of the developing embryo to nicotine resulted in higher numbers of hematopoietic progenitors in fetal liver. However postpartum, the number of hematopoietic stem/progenitor cells (HSPC) was decreased, suggesting an impaired colonization of the fetal bone marrow with HSPCs. This correlated with increased number of circulating HSPC and decreased expression of CXCR4 that mediates migration of circulating cells into the bone marrow regulatory niche. In addition, protein microarrays demonstrated that nicotine changed the profile of cytokines produced in the niche. While the levels of IL1alpha, IL1beta, IL2, IL9 and IL10 were not changed, the production of hematopoiesis-supportive cytokines including G-CSF, GM-CSF, IL3, IL6 and IGFBP-3 was decreased. This correlated with the decreased repopulating ability of HSPC in vivo and diminished hematopoietic activity in bone marrow cultures treated with nicotine. Interestingly, nicotine stimulated the production of IL4 and IL5, implying a possible role of the cholinergic system in pathogenesis of allergic diseases. Our data provide evidence that the nicotine-induced imbalance of the cholinergic system during gestation interferes with normal development and provides the basis for negative health outcomes postpartum in active and passive smokers.


Assuntos
Acetilcolinesterase/genética , Colina O-Acetiltransferase/genética , Sistema Hematopoético/metabolismo , Receptores Nicotínicos/genética , Acetilcolinesterase/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colina O-Acetiltransferase/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo/métodos , Expressão Gênica/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/metabolismo , Sistema Hematopoético/embriologia , Sistema Hematopoético/crescimento & desenvolvimento , Humanos , Imuno-Histoquímica , Injeções Intravenosas , Antígenos Comuns de Leucócito/análise , Camundongos , Camundongos Endogâmicos BALB C , Nicotina/administração & dosagem , Nicotina/farmacologia , Agonistas Nicotínicos/administração & dosagem , Agonistas Nicotínicos/farmacologia , Fosforilação/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/análise , Receptores CXCR4/metabolismo , Receptores Nicotínicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Stem Cells Dev ; 14(1): 81-91, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15725747

RESUMO

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.


Assuntos
Medula Óssea/efeitos dos fármacos , Feto/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Troca Materno-Fetal , Nicotina/efeitos adversos , Animais , Medula Óssea/embriologia , Células da Medula Óssea/citologia , Feminino , Desenvolvimento Fetal/efeitos dos fármacos , Feto/citologia , Células-Tronco Hematopoéticas/citologia , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/embriologia , Interleucina-6/deficiência , Camundongos , Gravidez
15.
World J Stem Cells ; 7(8): 1090-108, 2015 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-26435769

RESUMO

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.

16.
Adv Cancer Res ; 123: 149-89, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25081529

RESUMO

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.


Assuntos
Células da Medula Óssea/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Ácido Hialurônico/fisiologia , Leucemia/metabolismo , Animais , Aorta/patologia , Medula Óssea/metabolismo , Células da Medula Óssea/citologia , Diferenciação Celular , Movimento Celular , Fibroblastos/citologia , Glucuronosiltransferase/metabolismo , Homeostase , Humanos , Hialuronan Sintases , Ácido Hialurônico/química , Macrófagos/citologia , Camundongos , Camundongos Knockout , Músculo Liso/citologia , Osteoblastos/citologia , Osteoclastos/citologia , Polímeros/química , Ligação Proteica , Células-Tronco/citologia , Fatores de Tempo
17.
Stem Cells Transl Med ; 3(2): 229-40, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24396034

RESUMO

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.


Assuntos
Barreira Hematoencefálica/citologia , Células Endoteliais/citologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/transplante , Acidente Vascular Cerebral/terapia , Veias/citologia , Animais , Comunicação Celular , Movimento Celular/fisiologia , Sobrevivência Celular/fisiologia , Cultura em Câmaras de Difusão , Modelos Animais de Doenças , Células Endoteliais/fisiologia , Feminino , Fucose/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Injeções Intravenosas , Células-Tronco Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Estresse Mecânico , Acidente Vascular Cerebral/patologia , Veias/fisiologia
18.
Int Rev Cell Mol Biol ; 309: 259-302, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24529725

RESUMO

Plasminogen (PLG) is the zymogen of plasmin, the major enzyme that degrades fibrin clots. In addition to its binding and activation on fibrin clots, PLG also specifically interacts with cell surfaces where it is more efficiently activated by PLG activators, compared with the reaction in solution. This results in association of the broad-spectrum proteolytic activity of plasmin with cell surfaces that functions to promote cell migration. Here, we review emerging data establishing a role for PLG, plasminogen receptors and the newly discovered plasminogen receptor, Plg-RKT, in macrophage recruitment in the inflammatory response, and we address mechanisms by which the interplay between PLG and its receptors regulates inflammation.


Assuntos
Macrófagos/metabolismo , Plasminogênio/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Animais , Humanos , Macrófagos/patologia , Dados de Sequência Molecular , Peritonite/patologia , Plasminogênio/química , Proteômica , Receptores de Superfície Celular/química
19.
J Vis Exp ; (77): e50959, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23893091

RESUMO

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.


Assuntos
Movimento Celular/fisiologia , Quimiocinas/farmacologia , Técnicas Citológicas/instrumentação , Células Endoteliais/citologia , Animais , Células da Medula Óssea/citologia , Técnicas Citológicas/métodos , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Camundongos
20.
Front Biosci (Landmark Ed) ; 16(6): 2271-88, 2011 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-21622176

RESUMO

Mesenchymal stem cells (MSC) are multipotent stem cells that hold promise for an expanding list of therapeutic uses, not only due to their ability to differentiate into all connective tissues including bone, fat and cartilage, but additionally due to their trophic and anti-inflammatory effects which contribute to healing and tissue regeneration. Ongoing research is starting to illuminate important aspects of the microenvironmental niche, which supports MSC self-renewal. In this review, we summarize recent findings on cellular structures and molecular pathways that are involved in regulation of MSC self-renewal versus differentiation, and in retention of MSCs within the niche versus mobilization and recruitment to sites of injury. In addition, the contribution of MSCs to the structure and function of hematopoietic and cancerous niches is discussed.


Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Animais , Diferenciação Celular , Proliferação de Células , Citocinas/fisiologia , Matriz Extracelular/fisiologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Modelos Biológicos , Neoplasias/etiologia , Neoplasias/patologia , Neoplasias/terapia , Transdução de Sinais , Cicatrização/fisiologia , Ferimentos e Lesões/patologia , Ferimentos e Lesões/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa