Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Blood ; 135(25): 2252-2265, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32181817

RESUMO

Prolonged or enhanced expression of the proto-oncogene Lmo2 is associated with a severe form of T-cell acute lymphoblastic leukemia (T-ALL), designated early T-cell precursor ALL, which is characterized by the aberrant self-renewal and subsequent oncogenic transformation of immature thymocytes. It has been suggested that Lmo2 exerts these effects by functioning as component of a multi-subunit transcription complex that includes the ubiquitous adapter Ldb1 along with b-HLH and/or GATA family transcription factors; however, direct experimental evidence for this mechanism is lacking. In this study, we investigated the importance of Ldb1 for Lmo2-induced T-ALL by conditional deletion of Ldb1 in thymocytes in an Lmo2 transgenic mouse model of T-ALL. Our results identify a critical requirement for Ldb1 in Lmo2-induced thymocyte self-renewal and thymocyte radiation resistance and for the transition of preleukemic thymocytes to overt T-ALL. Moreover, Ldb1 was also required for acquisition of the aberrant preleukemic ETP gene expression signature in immature Lmo2 transgenic thymocytes. Co-binding of Ldb1 and Lmo2 was detected at the promoters of key upregulated T-ALL driver genes (Hhex, Lyl1, and Nfe2) in preleukemic Lmo2 transgenic thymocytes, and binding of both Ldb1 and Lmo2 at these sites was reduced following Cre-mediated deletion of Ldb1. Together, these results identify a key role for Ldb1, a nonproto-oncogene, in T-ALL and support a model in which Lmo2-induced T-ALL results from failure to downregulate Ldb1/Lmo2-nucleated transcription complexes which normally function to enforce self-renewal in bone marrow hematopoietic progenitors.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Autorrenovação Celular , Proteínas de Ligação a DNA/fisiologia , Proteínas com Domínio LIM/fisiologia , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Timócitos/citologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Transferência Adotiva , Animais , Antígenos CD/biossíntese , Transformação Celular Neoplásica , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Deleção de Genes , Técnicas de Introdução de Genes , Proteínas com Domínio LIM/deficiência , Proteínas com Domínio LIM/genética , Linfopoese , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proto-Oncogene Mas , RNA-Seq , Quimera por Radiação , Tolerância a Radiação , Timócitos/metabolismo , Timócitos/efeitos da radiação , Timócitos/transplante
3.
Amyloid ; 18(3): 136-46, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21830877

RESUMO

AA amyloid patients who experience disease progression and develop renal failure have not received sufficient benefit from agents that treat inflammation or infection. We have begun to explore the potential application of antisense oligonucleotides (ASOs) to specifically suppress SAA production and thereby reduce amyloid deposition. Proof-of-concept experiments conducted in mice initially examined ASO ability to reduce serum levels of SAA during an acute inflammatory response. Peak SAA levels in ASO-treated mice were reduced as much as 65% relative to levels in saline-treated mice. The extent of suppression was dose-dependent and influenced by the time interval between ASO administration and inflammatory stimulation. Subsequent experiments tested whether ASO suppression of SAA was sufficient to mitigate amyloid deposition. Amyloidosis was induced by amyloid-enhancing factor and silver nitrate injection; ASO treatment was initiated 1 week later and continued 1× or 3× per week; inflammation was re-triggered by subsequent injection(s) of silver nitrate; mice were sacrificed after 4-5 weeks. Examination of tissues by Congo red staining and SAA/AA immunohistochemistry revealed consistently less amyloid in the organs of ASO-treated mice compared to saline-treated counterparts. These findings provide rationale for further investigation of SAA-specific ASOs as a potential therapy for AA amyloidosis.


Assuntos
Amiloidose/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Placa Amiloide/tratamento farmacológico , Proteína Amiloide A Sérica/metabolismo , Amiloidose/sangue , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Caseínas/farmacologia , Citocinas/sangue , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática , Feminino , Imuno-Histoquímica , Camundongos , Oligonucleotídeos Antissenso/administração & dosagem , Placa Amiloide/sangue , Placa Amiloide/metabolismo , Placa Amiloide/patologia , Nitrato de Prata/farmacologia
4.
Chest ; 126(1): 186-91, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15249461

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV)-induced diseases are mediated through active cytokines released during infection. We hypothesized that RSV infection causes bronchial epithelial monolayer permeability in vitro via induction of vascular endothelial growth factor (VEGF). METHODS: Human bronchial epithelial cells were infected with RSV. In some cultures, VEGF antibody was included to block VEGF response; in other cultures, palivizumab was added to block RSV infection. Permeability was assessed in real-time using electric cell-substrate impedance sensing. VEGF release was assessed using enzyme-linked immunosorbent assay. Gap formation was assessed using live cell imaging. RESULTS: RSV-infected cells demonstrated a decrease in the resistance of the monolayer indicating an increase in permeability; this increase was blocked with VEGF-specific antibody, and palivizumab. Intercellular gap formation developed in RSV-infected epithelial monolayers. CONCLUSION: RSV increases permeability of the bronchial airway epithelial monolayer via VEGF induction.


Assuntos
Brônquios/virologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sinciciais Respiratórios , Fatores de Crescimento do Endotélio Vascular/biossíntese , Brônquios/metabolismo , Células Cultivadas , Ensaio de Imunoadsorção Enzimática , Humanos , Permeabilidade
5.
J Clin Immunol ; 24(4): 426-34, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15163899

RESUMO

Asthma is characterized by infiltration and shedding of the bronchial epithelium. The Th2 cytokines IL-4 and IL-13 are involved in the cellular recruitment and infiltration seen in asthma. The effects of IL-4 and IL-13 on cell-matrix interactions and epithelial shedding are unknown. We hypothesize that bronchial airway epithelial cells (BAEC) express paxillin, a structural focal adhesion protein, and downregulation of paxillin by Th2 cytokines lead to BAEC hyperpermeability. We showed by confocal microscopy the presence of paxillin in BAEC. We demonstrated by Western blot analysis that IL-4 and IL-13 stimulation results in downregulation of paxillin production. IL-4 and IL-13 stimulation decreased epithelial cell-matrix attachment as measured by electrical cell-substrate impedance sensing system (ECIS). Our results suggest that Th2 cytokines IL-4 and IL-13 downregulate paxillin production by BAEC, thereby disrupting the cell-matrix interactions. This may help explain the epithelial shedding and epithelial membrane hyperpermeability that occurs in asthma.


Assuntos
Brônquios/citologia , Proteínas do Citoesqueleto/genética , Células Epiteliais/efeitos dos fármacos , Interleucina-13/farmacologia , Interleucina-4/farmacologia , Fosfoproteínas/genética , Adesão Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Humanos , Paxilina , Permeabilidade/efeitos dos fármacos , Células Th2 , Fator A de Crescimento do Endotélio Vascular/farmacologia
6.
Chest ; 124(2): 682-7, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12907560

RESUMO

STUDY OBJECTIVES: Sialomucin complex (SMC) is a heterodimeric glycoprotein, and is found on the surfaces of the mesothelia of the pleura, pericardium, and peritoneum. Sialomucins play a significant role in adhesion as well as in defense. In this study, we hypothesized that pleural mesothelial cells (PMCs) express SMC and thus prevent the adherence of ovarian cancer cells (HTB-77) to the pleura. METHODS: PMCs were plated, and the adherence of HTB-77 cells was observed using a cytofluor. The PMC monolayer was pretreated with sialidase, and HTB-77 adherence was observed using cytofluor. In another set of HTB-77 cells, adherence was observed when the PMC monolayer was pretreated with supernatants of HTB-77 cells. Last, supernatants of HTB-77 cells were assayed for sialidase activity. RESULTS: The removal of SMC by sialidase greatly increased the adherence of HTB-77 cells to the PMC monolayer, which was statistically significant (p < 0.05). Similar results were obtained when the PMC monolayer was pretreated with the supernatants of HTB-77 cells. Supernatants of HTB-77 cells showed the presence of sialidase. CONCLUSIONS: The presence of SMC on the PMC acts as a defense layer, and its removal by sialidase increases the susceptibility of the PMC layer to the adherence of malignant cells and to increased metastasis. HTB-77 cells also express sialidase, which by its action on the monolayer aids in the adherence of tumor cells to the pleural surface.


Assuntos
Mucinas/fisiologia , Metástase Neoplásica/prevenção & controle , Neoplasias Ovarianas/metabolismo , Pleura/fisiologia , Adesão Celular/fisiologia , Feminino , Humanos , Neuraminidase/metabolismo , Neoplasias Ovarianas/enzimologia , Sialomucinas , Células Tumorais Cultivadas
7.
J Clin Immunol ; 23(1): 1-10, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12645855

RESUMO

In bacterial empyema, the recruited polymorphonuclear leukocytes (PMN) represent important phagocytic cells involved in antibacterial defense. In this study we demonstrate that pleural fluids (PF) obtained from patients with empyema (EMP) contains significantly higher levels of granulocyte colony stimulating factor (GM-CSF), and PMN incubated in empyema (EMP) pleural fluid (PF) showed significantly less apoptosis than congestive heart failure (CHF) PF. Staphylococcus aureus-stimulated PMC released significantly (P < 0.001) higher levels of GM-CSF than resting PMC. Staphylococcus aureus-stimulated PMC (SPMC)-CM significantly (P < 0.001) inhibited PMN apoptosis. In SPMC-CM-incubated PMN the antiapoptotic gene Bcl-xL mRNA and protein expression was up-regulated; Bak mRNA and protein expression was down-regulated compared to control PMN. The active caspases activity significantly decreased. When SPMC-CM and EMP PF were immunodepleted with GM-CSF antibody, PMN apoptosis was significantly higher. The delay in apoptosis of PMN is in part attributable to the release of cytokine GM-CSF by activated PMC. These findings suggest that S. aureus-activated PMC extend PMN life span by modulating Bcl-xL and Bak gene expression and active caspases activity during acute inflammation and empyema.


Assuntos
Apoptose/fisiologia , Empiema Pleural/metabolismo , Neutrófilos/metabolismo , Cavidade Pleural/metabolismo , Caspases/metabolismo , Epitélio/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neutrófilos/imunologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Staphylococcus aureus/imunologia , Proteína Killer-Antagonista Homóloga a bcl-2 , Proteína bcl-X
8.
Oncol Res ; 14(3): 155-61, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14760864

RESUMO

Tumors such as ovarian, lung, and breast have been found to have a predilection for the pleura. Pleural mesothelial cells (PMCs) play an active role in pleural inflammation via release of cytokines. However, mechanisms whereby PMCs defend themselves against invading malignant cells are unknown. In the present study, we hypothesized that PMCs release the antiangiogenic factor endostatin and inhibit malignant cell invasion. We evaluated the endostatin levels in malignant (MAL) and congestive heart failure (CHF) pleural fluids (PF). Endostatin expression by PMC was also demonstrated by Western analysis and confocal microscopy. Our results demonstrate that CHF PF contained significantly higher levels of endostatin when compared with MAL PF. PMCs alone released a significantly greater amount of endostatin when compared with ovarian cancer cells (OCCs). When the PMC were cocultured with OCCs without contact, there was an increase in the endostatin production. However, when the PMCs were cocultured in direct contact with OCCs the endostatin levels significantly decreased. Endostatin production was upregulated in the presence of tumor cells but not when OCCs were adherent to underlying PMC monolayer. Immunofluorescent staining of PMCs for endostatin correlated with endostatin release. These findings suggest that PMCs play a key role in the antiangiogenesis process by producing endostatin in the pleural space, and thus preventing tumor spread and metastasis in the pleura.


Assuntos
Transformação Celular Neoplásica/metabolismo , Endostatinas/metabolismo , Células Epiteliais/metabolismo , Pleura/metabolismo , Western Blotting , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/patologia , Feminino , Insuficiência Cardíaca/metabolismo , Humanos , Microscopia Confocal , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Pleura/citologia , Pleura/patologia
9.
Oncol Res ; 13(2): 71-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12392154

RESUMO

Hyaluronan (HA) is a nonsulfated glycosaminoglycan that is secreted in significant quantities by pleural mesothelial cells (PMC) and malignant mesotheioma cells (MMC). The functional significance of HA deposition in the pleural space has not been fully elucidated. In this study, we hypothesized that low molecular weight but not high molecular weight hyaluronan induces proliferation and migration of MMC, and that the hyaluronan receptor (CD44S) expressed on the mesothelioma cell surface is involved in this process. We evaluated the effect of low molecular weight hyaluronan (LMWHA) and high molecular weight hyaluronan (HMWHA) on four MMC lines (CRL-2081, CRL-5915, CRL-5830, CRL-5820) proliferation and haptotactic migration. We also studied the expression of HA receptor CD44S on MMC by Northern hybridization and flow cytometry. The binding of LMWHA and HMWHA t o MMC surface was determined by FACS analysis using FITC-conjugated hyaluronan. Our results indicate that the MMC line that expressed the highest amount of CD44 receptor showed increased proliferation and haptotactic migration of MMC when stimulated with LMWHA but not HMWHA. Monoclonal antibody against CD44 inhibited proliferation by about 12-40% and migration by 10-35% in the MMC lines that were studied, and thus in part inhibited LMWHA-induced proliferation and migration in MMC. LMWHA binding to MM cell surface was significantly higher than HMWHA. This directly correlated with their CD44 receptor expression. Neutralization of CD44 receptor significantly reduced the LMMHA binding to MMC. These results provide evidence that the interaction between the adhesive protein receptor CD44 and extracellular matrix component (HA) transmits regulatory signals for mediating the locomotion and proliferation of MMC, and thus plays an important role in localized extension of tumor growth.


Assuntos
Receptores de Hialuronatos/fisiologia , Ácido Hialurônico/farmacologia , Mesotelioma/patologia , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Humanos , Receptores de Hialuronatos/genética , Peso Molecular , RNA Mensageiro/análise , Células Tumorais Cultivadas
10.
Oncol Res ; 13(2): 79-85, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12392155

RESUMO

Malignant pleural effusion (MPE) carries a grave prognosis with median survival after diagnosis being 5 months. The major causes of MPE are lung, breast, ovary,and gastric cancer. It is still unclear how cancer cells penetrate the pleural mesothelial monolayer and reach the pleural space. In this study we examined the effect of ovarian epithelial cancer cells on a confluent pleural mesothelial cell (PMC) monolayer. We demonstrate that ovarian cancer cells adhere to the mesothelial monolayer in a time-dependent manner and induce PMC barrier dysfunction as evidenced by a drop in electrical resistance on electrical cell substrate impedance-sensing system (ECIS) and increased protein permeability. Barrier dysfunction is attenuated by addition of vascular endothelial growth factor (VEGF) antibody. Significant release of VEOF was noted when ovarian cancer cells were cocultured with PMC. Electron microscopy demonstrated gap formation in PMC monolayer only at the site of cancer cell attachment with surrounding areas remaining confluent.


Assuntos
Adesão Celular , Neoplasias Ovarianas/patologia , Pleura/citologia , Animais , Técnicas de Cocultura , Fatores de Crescimento Endotelial/biossíntese , Células Epiteliais/metabolismo , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Linfocinas/biossíntese , Camundongos , Permeabilidade , Proteínas/metabolismo , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
11.
J Clin Immunol ; 22(3): 144-52, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-12078856

RESUMO

The repair of an injured bronchial epithelial cell (BEC) monolayer requires proliferation and migration of BECs into the injured area. We hypothesized that BEC monolayer injury results in monocyte chemoattractant protein-1 (MCP-1) production, which initiates the repair process. BECs (BEAS-2B from ATCC) were utilized in this study. MCP-1 interacts with CCR2B receptor (CCR2B), resulting in cell proliferation, haptotaxis, and healing of the monolayer. Reverse transcriptase-polymerase chain reaction (RT-PCR) was employed to verify the presence of CCR2B. CCR2B was not merely present but also inducible by interleukin-2 (IL-2) and lipopolysaccharide (LPS). We demonstrated by immunohistochemistry that BECs express MCP-1 after injury and that receptor expression can be regulated by exposure to IL-2 and LPS. Haptotactic migration of cells was enhanced in the presence of MCP-1 and reduced in the presence of CCR2B antibody. This enhanced or depressed ability of the BECs to perform haptotactic migration was shown to be statistically significant (P < 0.05) when compared to controls. Finally, BECs proliferate in response to MCP-1 as proven by electric cell-substrate impedance sensing (ECIS) technology. MCP-1-specific antibodies were shown to neutralize the MCP-1-mediated BEC proliferation. This cascade of events following injury to the bronchial epithelium may provide insight into the mechanism of the repair process.


Assuntos
Brônquios/citologia , Quimiocina CCL2/fisiologia , Células Epiteliais/patologia , Divisão Celular , Linhagem Celular Transformada , Movimento Celular , Quimiocina CCL2/biossíntese , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Interleucina-2/farmacologia , Lipopolissacarídeos/farmacologia , Receptores CCR2 , Receptores de Quimiocinas/efeitos dos fármacos , Receptores de Quimiocinas/fisiologia
12.
Inflammation ; 26(2): 73-82, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11989790

RESUMO

Current knowledge implicates pleural mesothelial cells as mainly responsible for inflammatory responses in the pleural space. However, a vast body of recent evidence underscores the important role of fibroblasts in the process of inflammation in several types of tissues. We hypothesize that HPFBs (human pleural fibroblasts) play an important role in pleural responses and also when activated by bacterial endotoxin LPS (lipopolysaccharide), IL-1 beta (interleukin-1 beta), or TNF-alpha (tumor necrosis factor-alpha) release of C-C and C-X-C chemokines-specifically, MCP-1 and IL-8. Our results show that pleural fluid-isolated human fibroblasts release IL-8 and MCP-1 upon stimulation with IL-1 beta, TNF-alpha, and LPS in both a concentration- and time-dependent manner. RT-PCR (reverse-transcriptase-polymerase chain reaction) studies have also confirmed IL-8- and MCP-1-specific mRNA expression in activated pleural fibroblasts. On the time-dependent response curve, IL-8 was found in maximum concentrations at 144 hr, whereas MCP-1 continued to increase even after 196 hr following stimulation. IL-1 beta induced the maximum release of IL-8 (800-fold) and MCP-1 (164-fold), as compared to the controls. TNF-alpha induced a 95-fold increase in IL-8 and an 84-fold increase in MCP-1 levels, as compared to the controls. Collectively, our results show that human pleural fibroblasts contribute to the inflammatory cascade in the pleural space.


Assuntos
Quimiocina CCL2/biossíntese , Fibroblastos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-1/farmacologia , Interleucina-8/biossíntese , Pleura/citologia , Fator de Necrose Tumoral alfa/farmacologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Quimiocina CCL2/genética , Quimiotaxia de Leucócito/efeitos dos fármacos , DNA Complementar/genética , Fibroblastos/metabolismo , Humanos , Interleucina-8/genética , Lipopolissacarídeos/farmacologia , Monócitos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , RNA Mensageiro/biossíntese , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...