Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
BMC Cancer ; 22(1): 997, 2022 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-36127634

RESUMO

BACKGROUND: Severe graft versus host disease (GVHD) is the main reason for non-relapse mortality following allogeneic hematopoietic cell transplantation (HCT). We investigated the serum protein profiles of patients who had undergone HCT to identify predictive biomarkers of severe acute GVHD (aGVHD). METHODS: Serum samples were collected for 30 patients from day - 7 to day + 14 of HCT. The serum levels of plasma beta2-microglobulin (ß2-MG), soluble vascular cell adhesion molecule-1 (sVCAM-1), platelet factor 4, and TNFSF-14 were measured by ELISA as potential biomarkers following 310 cytokine profiling array. RESULTS: The median age of the study patients was 53.5 years (range, 19-69). All grade and grade 2-4 aGVHD developed in 21 (70.0%) and 17 (56.7%) patients, respectively. Compared with their baseline levels on day - 7, ß2-MG and sVCAM-1 were significantly increased on day + 14 of the HCT procedure (P = 0.028 and P < 0.001, respectively). Patients with a grade 2-4 severe aGVHD showed a significantly higher sVCAM-1 level at baseline (day-7) and at day + 14, compared with the other group with a grade 1 aGVHD or no aGVHD (P = 0.028 and P = 0.035, respectively). CONCLUSION: Higher sVCAM- levels at baseline and on day + 14 in HCT patients could be a significant predictive biomarker of severe aGVHD.


Assuntos
Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Adulto , Idoso , Biomarcadores , Doença Enxerto-Hospedeiro/diagnóstico , Doença Enxerto-Hospedeiro/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Humanos , Pessoa de Meia-Idade , Fator Plaquetário 4 , Molécula 1 de Adesão de Célula Vascular , Adulto Jovem
2.
BMC Cancer ; 20(1): 285, 2020 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-32252668

RESUMO

BACKGROUND: Leukemia stem cells (LSCs) in play an important role in the initiation, relapse, and progression of acute myeloid leukemia (AML), and in the development of chemotherapeutic drug resistance in AML. Studies regarding the detection of LSCs and the development of novel therapies for targeting them are extensive. The identification of LSCs and targeting therapies for them has been continuously under investigation. METHODS: We examined the levels of CD45dimCD34+CD38-CD133+ cells in bone marrow samples from patients with hematological malignancies and healthy controls, using four-color flow cytometry. RESULTS: Interestingly, the CD45dimCD34+CD38-CD133+ cells were highly expressed in the bone marrow of patients with AML compared to that in healthy controls (HC). Moreover, the proportions of CD45dimCD34+CD38-CD133+ cells were also examined in diverse hematological malignancies, including AML, CML, DLBCL, MM, MDS, HL, ALL, and CLL. LSCs were prominently detected in the BMCs isolated from patients with AML and CML, but rarely in BMCs isolated from patients with DLBCL, MM, MDS, ALL, CLL, and HL. Additionally, the high CD45dimCD34+CD38-CD133+ cell counts in AML patients served as a significantly poor risk factor for overall and event free survival. CONCLUSIONS: Therefore, our results suggest that CD45dimCD34+CD38-CD133+ cells in AML might potentially serve as LSCs. In addition, this cell population might represent a novel therapeutic target in AML.


Assuntos
Antígeno AC133/metabolismo , ADP-Ribosil Ciclase 1/metabolismo , Antígenos CD34/metabolismo , Medula Óssea/patologia , Leucemia Mieloide Aguda/patologia , Antígenos Comuns de Leucócito/metabolismo , Glicoproteínas de Membrana/metabolismo , Células-Tronco Neoplásicas/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/metabolismo , Medula Óssea/metabolismo , Feminino , Citometria de Fluxo/métodos , Humanos , Imunofenotipagem/métodos , Leucemia Mieloide Aguda/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida
3.
BMC Cancer ; 20(1): 1193, 2020 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-33276759

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) is a heterogeneous disease that frequently relapses after standard chemotherapy. Therefore, there is a need for the development of novel chemotherapeutic agents that could treat AML effectively. Radotinib, an oral BCR-ABL tyrosine kinase inhibitor, was developed as a drug for the treatment of chronic myeloid leukemia. Previously, we reported that radotinib exerts increased cytotoxic effects towards AML cells. However, little is known about the effects of combining radotinib with Ara-C, a conventional chemotherapeutic agent for AML, with respect to cell death in AML cells. Therefore, we investigated combination effects of radotinib and Ara-C on AML in this study. METHODS: Synergistic anti-cancer effects of radotinib and Ara-C in AML cells including HL60, HEL92.1.7, THP-1 and bone marrow cells from AML patients have been examined. Diverse cell biological assays such as cell viability assay, Annexin V-positive cells, caspase-3 activity, cell cycle distribution, and related signaling pathway have been performed. RESULTS: The combination of radotinib and Ara-C was found to induce AML cell apoptosis, which involved the mitochondrial pathway. In brief, combined radotinib and Ara-C significantly induced Annexin V-positive cells, cytosolic cytochrome C, and the pro-apoptotic protein Bax in AML cells including HL60, HEL92.1.7, and THP-1. In addition, mitochondrial membrane potential and Bcl-xl protein were markedly decreased by radotinib and Ara-C. Moreover, this combination induced caspase-3 activity. Cleaved caspase-3, 7, and 9 levels were also increased by combined radotinib and Ara-C. Additionally, radotinib and Ara-C co-treatment induced G0/G1 arrest via the induction of CDKIs such as p21 and p27 and the inhibition of CDK2 and cyclin E. Thus, radotinib/Ara-C induces mitochondrial-dependent apoptosis and G0/G1 arrest via the regulation of the CDKI-CDK-cyclin cascade in AML cells. In addition, our results showed that combined treatment with radotinib and Ara-C inhibits AML cell growth, including tumor volumes and weights in vivo. Also, the combination of radotinib and Ara-C can sensitize cells to chemotherapeutic agents such as daunorubicin or idarubicin in AML cells. CONCLUSIONS: Therefore, our results can be concluded that radotinib in combination with Ara-C possesses a strong anti-AML activity.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Citarabina/farmacologia , Leucemia Mieloide Aguda/patologia , Inibidores de Proteínas Quinases/farmacologia , Pirazinas/farmacologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Daunorrubicina/farmacologia , Sinergismo Farmacológico , Células HL-60 , Humanos , Idarubicina/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Nus , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Distribuição Aleatória , Método Simples-Cego , Organismos Livres de Patógenos Específicos , Células Tumorais Cultivadas
4.
Tumour Biol ; 41(5): 1010428319848612, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31074363

RESUMO

Aurora kinases play critical roles in regulating several processes pivotal for mitosis. Radotinib, which is approved in South Korea as a second-line treatment for chronic myeloid leukemia, inhibits the tyrosine kinase BCR-ABL and platelet-derived growth factor receptor. However, the effects of radotinib on Aurora kinase expression in acute myeloid leukemia are not well studied. Interestingly, the cytotoxicity of acute myeloid leukemia cells was increased by radotinib treatment. Radotinib significantly decreased the expression of cyclin-dependent kinase 1 and cyclin B1, the key regulators of G2/M phase, and inhibited the expression of Aurora kinase A and Aurora kinase B in acute myeloid leukemia cells. In addition, radotinib decreased the expression and binding between p-Aurora kinase A and TPX2, which are required for spindle assembly. Furthermore, it reduced Aurora kinase A and polo-like kinase 1 phosphorylation and suppressed the expression of α-, ß-, and γ-tubulin in acute myeloid leukemia cells. Furthermore, radotinib significantly suppressed the key regulators of G2/M phase including cyclin B1 and Aurora kinase A in a xenograft animal model. Therefore, our results suggest that radotinib can abrogate acute myeloid leukemia cell growth both in vitro and in vivo and may serve as a candidate agent or a chemosensitizer for treating acute myeloid leukemia.


Assuntos
Aurora Quinase A/antagonistas & inibidores , Benzamidas/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/patologia , Mitose/efeitos dos fármacos , Pirazinas/farmacologia , Animais , Apoptose , Aurora Quinase A/metabolismo , Ciclo Celular , Proliferação de Células , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Camundongos Nus , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
PLoS One ; 18(8): e0289798, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37552689

RESUMO

Liver transplantation is the most effective treatment option for patients with acute or chronic liver failure. However, the applicability and effectiveness of this modality are often limited by a shortage of donors, surgical complications, high medical costs, and the need for continuing immunosuppressive therapy. An alternative approach is liver cell transplantation. LIGHT (a member of the tumor necrosis factor superfamily) could be a promising candidate for promoting the differentiation of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) into hepatocyte-like cells. In this study, we investigated the effect of LIGHT on hBM-MSC differentiation into hepatocyte-like cells. Our previous results showed that LIGHT receptor lymphotoxin-ß receptor (LTßR) is constitutively expressed on the surface of hBM-MSCs. Upon treatment with recombinant human LIGHT (rhLIGHT), the phenotype of hBM-MSCs changed to round or polygonal cells. In addition, the cells exhibited high levels of hepatocyte-specific markers, including albumin, cytokeratin-18 (CK-18), CK-19, cytochrome P450 family 1 subfamily A member 1 (CYP1A1), CYP1A2, CYP3A4, SRY-box transcription factor 17 (SOX17), and forkhead box A2 (FOXA2). These results indicate that rhLIGHT enhances the differentiation of hBM-MSCs into functional hepatocyte-like cells. Furthermore, rhLIGHT-induced hepatocyte-like cells showed a higher ability to store glycogen and uptake indocyanine green compared with control cells, indicating functional progression. Additionally, treatment with rhLIGHT increased the number, viability, and proliferation of cells by inducing the S/G2/M phase and upregulating the expression of various cyclin and cyclin dependent kinase (CDK) proteins. We also found that the hepatogenic differentiation of hBM-MSCs induced by rhLIGHT was mediated by the activation of signal transducer and activator of transcription 3 (STAT3) and STAT5 pathways. Overall, our findings suggest that LIGHT plays an essential role in promoting the hepatogenic differentiation of hBM-MSCs. Hence, LIGHT may be a valuable factor for stem cell therapy.


Assuntos
Medula Óssea , Células-Tronco Mesenquimais , Humanos , Células da Medula Óssea , Diferenciação Celular , Hepatócitos/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Células Cultivadas , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia
6.
Prostate ; 72(12): 1351-8, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22228081

RESUMO

BACKGROUND: Nod1 and Nod2 are cytosolic receptors which are responsible for sensing bacterial peptidoglycan derivatives. In this study, we determined whether Nod1 and Nod2 are involved in the innate immune responses of prostate epithelial cells. METHODS: The expression of Nod1 and Nod2 was examined by RT-PCR and immunohistochemistry. ELISA was performed to determine the production of cytokines/chemokines. Activation of NF-κB and MAPK was examined using western blot analysis. RESULTS: The Nod1 gene was distinctly expressed in all tested cells including DU145, PC3, and TRAMP-C2 cells, whereas Nod2 expression was weak. Both Nod1 and Nod2 proteins were expressed in normal mouse prostate epithelia with difference of expression levels. Tri-DAP (Nod1 agonist), but not MDP (Nod2), increased the production of IL-8 (or KC) and IL-6 in prostate epithelial cells. Tri-DAP and MDP could upregulate the gene expression of COX-2 and activate NF-κB and MAPK. In addition, Tri-DAP and MDP synergized with TLR agonists to induce the production of IL-8/KC or IL-6 in PC3 and TRAMP-C2 cells. We finally showed that Nod1 and Nod2 were also expressed in a wide range of prostate lesions including prostate intraepithelial neoplasm (PIN), phyllodes-like tumor, and adenocarcinoma in TRAMP (transgenic adenocarcinoma of the mouse prostate) mice, even though the expression level of Nod1 and Nod2 was different. CONCLUSION: These results indicate that Nod1 and Nod2 may play important roles in the innate immune response of prostate epithelial cells and the development and progression of prostate cancer.


Assuntos
Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Imunidade Inata , Proteína Adaptadora de Sinalização NOD1/metabolismo , Proteína Adaptadora de Sinalização NOD2/metabolismo , Próstata/imunologia , Próstata/metabolismo , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/metabolismo , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Linhagem Celular Tumoral , Progressão da Doença , Células Epiteliais/citologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Adaptadora de Sinalização NOD1/fisiologia , Proteína Adaptadora de Sinalização NOD2/fisiologia , Próstata/citologia , Neoplasias da Próstata/patologia
7.
PLoS One ; 17(5): e0265958, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35503759

RESUMO

Multiple myeloma (MM) is a hematological cancer causing from accumulated abnormal plasma cells. STAT3 overexpression in MM appears to be mediated by a variety of factors, and it may be associated with an adverse prognosis and play a role in microenvironment-dependent treatment resistance. Unfortunately, MM remains an incurable disease, as relapse is very common. Therefore, there is urgent need to develop new treatment options for MM. Radotinib is a novel anti-cancer drug, currently approved in South Korea for the treatment of chronic myeloid leukemia patients. It is an oral, multitargeted inhibitor of receptor tyrosine kinases, including BCR-ABL, c-KIT, PDGFR, and Src family kinases. However, little is known about the effects of radotinib on multiple myeloma cells. However, little is known about the effects of radotinib on multiple myeloma cells. But even tinip almost not known about the impact of multiple myeloma cells. Moreover, nothing is known about how it affects STAT3 and JAK2. In this study, we analyzed the effect of radotinib on multiple myeloma cells. Herein, Moreover, nothing is known about how it. Moreover, not all is known about how the affects STAT3 and JAK2. We investigated the effect of radotinib on the STAT3 signaling pathway in MM cells, including several MM cell lines and mouse models. So we investigated the effect of radotinib on MM cells, including several MM cell lines and mouse models. Interestingly, radotinib induced apoptosis, and inhibited cell proliferation in MM cells including RPMI-8226, MM.1S, U266B1, and IM-9 cells. Moreover, radotinib treatment significantly increased the number Annexin V-positive cells and G0/G1-phase cells. In addition, radotinib treatment in various MM cells strongly suppressed the activity and expression of STAT3 and JAK2 proteins. We also observed that diverse proteins related to the STAT3 signaling pathway, including c-Myc, Bcl-xL, Mcl-1, cyclin D1 and cyclin D3, were powerfully inhibited by radotinib treatment in MM cells. Furthermore, radotinib significantly suppressed MM cell growth in a xenograft animal model using IM-9 cells. In conclusion, radotinib may play an important role as a candidate agent for MM treatment.


Assuntos
Mieloma Múltiplo , Animais , Apoptose , Benzamidas , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Pirazinas , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Microambiente Tumoral
8.
Sci Rep ; 11(1): 13198, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34168229

RESUMO

Multiple myeloma (MM) is a hematological cancer resulting from accumulated abnormal plasma cells. Unfortunately, MM remains an incurable disease, as relapse is very common. Therefore, there is urgent need to develop new treatment options for MM. Radotinib is a novel anti-cancer drug, currently approved in South Korea for the treatment of chronic myeloid leukemia patients. Its mechanism of action involves inhibition of the tyrosine kinase Bcr-Abl and the platelet-derived growth factor receptor. Generally, the mechanism of inhibition of non-receptor tyrosine kinase c-Abl has played an essential role in the inhibition of cancer progression. However, little is known regarding the effects of the c-Abl inhibitor, radotinib on MM cells. In this study, we analyzed the effect of radotinib on multiple myeloma cells. Interestingly, radotinib caused apoptosis in MM cells including RPMI-8226, MM.1S, and IM-9 cells, even in the absence of c-kit expression in 2 of these lines. Radotinib treatment significantly increased the number Annexin V-positive cells and decreased the mitochondrial membrane potential in MM cells. Additionally, we observed that cytochrome C was localized in the cytosol of radotinib-treated MM cells. Moreover, radotinib decreased the expression of Bcl-2 and Bcl-xL, and increased the expression of Bax and Bak in MM cells. Furthermore, radotinib promoted caspase pathway activation by inducing the expression and activity of caspase-3, -7, and -9. Expression of cleaved PARP-1 was also increased by radotinib treatment in various MM cells. In addition, radotinib significantly suppressed MM cell growth in a xenograft animal model using RPMI-8226 cells, and killed ex vivo myeloma cells from patients. In conclusion, radotinib may play an important role as a candidate agent or chemosensitizer for the treatment of MM.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Mitocôndrias/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Pirazinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Caspases/metabolismo , Humanos , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
9.
PLoS One ; 16(2): e0247368, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33606781

RESUMO

Osteoporosis is a progressive systemic skeletal disease associated with decreased bone mineral density and deterioration of bone quality, and it affects millions of people worldwide. Currently, it is treated mainly using antiresorptive and osteoanabolic agents. However, these drugs have severe adverse effects. Cell replacement therapy using mesenchymal stem cells (MSCs) could serve as a treatment strategy for osteoporosis in the future. LIGHT (HVEM-L, TNFSF14, or CD258) is a member of the tumor necrosis factor superfamily. However, the effect of recombinant LIGHT (rhLIGHT) on osteogenesis in human bone marrow-derived MSCs (hBM-MSCs) is unknown. Therefore, we monitored the effects of LIGHT on osteogenesis of hBM-MSCs. Lymphotoxin-ß receptor (LTßR), which is a LIGHT receptor, was constitutively expressed on the surface of hBM-MSCs. After rhLIGHT treatment, calcium and phosphate deposition in hBM-MSCs, stained by Alizarin red and von Kossa, respectively, significantly increased. We performed quantitative real-time polymerase chain reaction to examine the expressions of osteoprogenitor markers (RUNX2/CBFA1 and collagen I alpha 1) and osteoblast markers (alkaline phosphatase, osterix/Sp7, and osteocalcin) and immunoblotting to assess the underlying biological mechanisms following rhLIGHT treatment. We found that rhLIGHT treatment enhanced von Kossa- and Alizarin red-positive hBM-MSCs and induced the expression of diverse differentiation markers of osteogenesis in a dose-dependent manner. WNT/ß-catenin pathway activation strongly mediated rhLIGHT-induced osteogenesis of hBM-MSCs, accelerating the differentiation of hBM-MSCs into osteocytes. In conclusion, the interaction between LIGHT and LTßR enhances osteogenesis of hBM-MSCs. Therefore, LIGHT might play an important role in stem cell therapy.


Assuntos
Receptor beta de Linfotoxina/genética , Receptor beta de Linfotoxina/metabolismo , Células-Tronco Mesenquimais/citologia , Osteogênese , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Marcadores Genéticos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Proteínas Recombinantes/farmacologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/genética , Via de Sinalização Wnt/efeitos dos fármacos
10.
Infect Immun ; 77(5): 2168-76, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19237524

RESUMO

The tumor necrosis factor receptor family molecule 4-1BB (CD137) has diverse roles in adaptive and innate immune responses. However, little is known of its role in bacterial infections. Previously, we showed that 4-1BB-deficient mice have enhanced susceptibility to Listeria monocytogenes infection, and mice pretreated with agonistic anti-4-1BB antibody (3E1) were much more resistant to L. monocytogenes infection than mice treated with control antibody. In this study, we report that stimulating 4-1BB by administering 3E1 in the early phase of L. monocytogenes infection is critical for promoting the survival of mice by inducing rapid infiltration of neutrophils and monocytes into L. monocytogenes-infected livers. The levels of tumor necrosis factor alpha, interleukin 6, and monocyte chemoattractant protein 1 in the livers of 3E1-treated mice increased as early as 30 min postinfection and peaked by 1 to 2 h, while those in mice treated with control antibody started to increase only at 16 h postinfection. Monocytes and neutrophils from the 3E1-treated mice had higher levels of activation markers, phagocytic activity, and reactive oxygen species than those from control mice. In vitro stimulation of 4-1BB induced the production of the inflammatory cytokines/chemokines of neutrophils, but not those of monocytes. These results suggest that 4-1BB stimulation of neutrophils in the early phase of L. monocytogenes infection causes rapid production of inflammatory cytokines/chemokines and that the subsequent infiltration of neutrophils and monocytes is crucial for eliminating the infecting L. monocytogenes.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Listeria monocytogenes/imunologia , Listeriose/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/agonistas , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Animais , Quimiocina CCL2/análise , Feminino , Interleucina-6/análise , Fígado/química , Fígado/imunologia , Fígado/patologia , Camundongos , Camundongos Endogâmicos BALB C , Fagocitose , Espécies Reativas de Oxigênio/metabolismo , Análise de Sobrevida , Fator de Necrose Tumoral alfa/análise
11.
J Cell Biochem ; 107(1): 123-33, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19241441

RESUMO

LIGHT acted as a new player in the atherogenesis. The dried, unripe fruit of Evodia Fructus (EF) has long been used as a traditional Chinese herbal medicine, and is currently widely used for the treatment of headache, abdominal pain, vomiting, colds and reduced blood circulation. Evodiamine and rutaecarpine are active components of EF. In this study, we investigated the inhibitory effect of evodiamine and rutaecarpine on LIGHT-induced migration in human monocytes. Evodiamine and rutaecarpine decreased the LIGHT-induced production of ROS, IL-8, monocyte chemoattractant protein-1 (MCP-1), TNF-alpha, and IL-6, as well as the expression of chemokine receptor (CCR) 1, CCR2 and ICAM-1 and the phosphorylation of the ERK 1/2 and p38 MAPK. Furthermore, NADPH oxidase assembly inhibitor, AEBSF, blocked LIGHT-induced migration and activation of CCR1, CCR2, ICAM-1, and MAPK such as ERK and p38 in a manner similar to evodiamine and rutaecarpine. These findings indicate that the inhibitory effects of evodiamine and rutaecarpine on LIGHT-induced migration and the activation of CCR1, CCR2, ICAM-1, ERK, and p38 MAPK occurs via decreased ROS production and NADPH oxidase activation. Taken together, these results indicate that evodiamine and rutaecarpine have the potential for use as an anti-atherosclerosis agent.


Assuntos
Alcaloides Indólicos/farmacologia , Monócitos/efeitos dos fármacos , NADPH Oxidases/efeitos dos fármacos , Extratos Vegetais/farmacologia , Quinazolinas/farmacologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Citometria de Fluxo , Humanos , Immunoblotting , Molécula 1 de Adesão Intercelular/efeitos dos fármacos , Molécula 1 de Adesão Intercelular/metabolismo , Monócitos/metabolismo , NADPH Oxidases/metabolismo , Fosfoproteínas/efeitos dos fármacos , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR2/efeitos dos fármacos , Receptores CCR2/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
J Vasc Res ; 46(4): 375-86, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19142017

RESUMO

BACKGROUND: Vascular smooth-muscle cell proliferation plays an important role in atherosclerosis and restenosis. Rhein is an active component extracted from rhubarb. In this study, rhein was found to exert potent inhibitory effects against tumor necrosis factor (TNF)-alpha-induced human aortic smooth-muscle cells (HASMCs) proliferation. METHOD: These effects were associated with induced apoptosis, including the induction of Annexin V-positive cells, the cleavage of poly(ADP-ribose)polymerase (PARP), and caspases 3, 8 and 9. RESULTS: Inhibitors of caspases 3, 8 and 9 were efficiently blocked by rhein-induced apoptosis in TNF-alpha-treated HASMCs. In addition, treatment with rhein resulted in the release of cytochrome c into the cytosol, a loss of mitochondrial membrane potential (DeltaPsi(m)), a decrease in Bcl-2 and Bcl-xL and an increase in Bax and Bak expression. However, rhein-mediated apoptosis was blocked by a mitochondrial membrane depolarization inhibitor. These findings indicate that rhein-induced apoptosis occurred via a mitochondrial pathway. Furthermore, the inhibition of mitochondrial membrane depolarization was efficiently blocked by rhein-induced caspase-9 activity, which indicates that the rhein-induced caspase activation signal was downstream of the mitochondrial pathway. Taken together, the results of this study show that rhein inhibits TNF-alpha-induced HASMC proliferation via mitochondria-dependent apoptosis and that rhein has the potential to act as an anti-atherosclerosis agent.


Assuntos
Antraquinonas/farmacologia , Apoptose/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Proliferação de Células/efeitos dos fármacos , Mitocôndrias Musculares/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Ácido Bongcréquico/farmacologia , Inibidores de Caspase , Caspases/metabolismo , Células Cultivadas , Inibidores de Cisteína Proteinase/farmacologia , Citocromos c/metabolismo , Relação Dose-Resposta a Droga , Humanos , Potencial da Membrana Mitocondrial , Mitocôndrias Musculares/metabolismo , Mitocôndrias Musculares/patologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Proteínas Recombinantes/metabolismo
13.
J Cell Biochem ; 105(1): 70-80, 2008 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-18494000

RESUMO

Vascular smooth-muscle cell (VSMC) proliferation plays a vital role in hypertension, atherosclerosis and restenosis. It has been reported that emodin, an active component extracted from rhubarb, can stop the growth of cancer cells; however, it is not known if emodin exerts similar anti-atherogenic effects in TNF-alpha treated human aortic smooth-muscle cells (HASMC). In this study, emodin treatment showed potent inhibitory effects in TNF-alpha-induced HASMC proliferation that were associated with induced apoptosis, including the cleavage of poly ADP-ribose polymerase (PARP). Moreover, inhibitors of caspase-3, -8 and -9 (Ac-DEVD-CHO, Z-IETD-FMK and Z-LEHD-FMK) efficiently blocked emodin-induced apoptosis in TNF-alpha treated HASMC. Therefore, emodin-induced cell death occurred via caspase-dependent apoptosis. Emodin treatment resulted in the release of cytochrome c into cytosol and a loss of mitochondrial membrane potential (DeltaPsi(m)), as well as a decrease in the expression of an anti-apoptotic protein (Bcl-2) and an increase in the expression of an a pro-apoptotic protein (Bax). Emodin-mediated apoptosis was also blocked by a mitochondrial membrane depolarization inhibitor, which indicates that emodin-induced apoptosis occurred via a mitochondrial pathway. Taken together, the results of this study showed that emodin inhibits TNF-alpha-induced HASMC proliferation via caspase- and a mitochondrial-dependent apoptotic pathway. In addition, these results indicate that emodin has potential as an anti-atherosclerosis agent.


Assuntos
Aorta/metabolismo , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Emodina/farmacologia , Mitocôndrias/enzimologia , Miócitos de Músculo Liso/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Aorta/citologia , Aorta/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Humanos , Quinase I-kappa B/metabolismo , Mitocôndrias/efeitos dos fármacos , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Biochem Biophys Res Commun ; 371(4): 834-40, 2008 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-18468509

RESUMO

Homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus (HSV) glycoprotein D (gD) for herpes virus entry mediator (HVEM; TR2) (LIGHT), a ligand of herpes virus entry mediator (HVEM), increased reactive oxygen species (ROS) and enhanced the destruction of bacteria in human monocytes. In this study, rhLIGHT was found to increase the expression of the chemokine receptors, chemokine receptor 1 (CCR1) and CCR2, as well as to accelerate the migration activity of human monocytes. Additionally, rhLIGHT was found to increase ROS via NADPH oxidase p47(phox) phosphorylation, which was found to be required for LIGHT-induced NF-kappaB activation, CCR1 and CCR2 expression, migration and IL-8 and TNF-alpha production. Taken together, these results indicate that NADPH oxidase activation is required for rhLIGHT-induced migration in human monocytes.


Assuntos
Movimento Celular , Monócitos/imunologia , NADPH Oxidases/metabolismo , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/fisiologia , Ativação Enzimática , Humanos , Proteínas I-kappa B/metabolismo , Interleucina-8/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/enzimologia , Inibidor de NF-kappaB alfa , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR1/metabolismo , Receptores CCR2/metabolismo , Proteínas Recombinantes/farmacologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Immunol Lett ; 120(1-2): 57-64, 2008 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-18675302

RESUMO

Inflammation is an important event in the development of vascular diseases such as hypertension, atherosclerosis, and restenosis. In addition, the stimulation of Toll-like receptor 4 (TLR4) by lipopolysaccharide (LPS) induces the release of critical proinflammatory cytokines that activate potent immune responses. In this study, LPS was found to induce TLR4 expression and increased nitric oxide (NO) production by increasing the expression of inducible nitric oxide synthase (iNOS). Furthermore, LPS was found to induce interleukin (IL)-8 and vascular endothelial growth factor (VEGF) production, as well as intracellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) expression. Taken together, these results indicate that LPS induces inflammatory responses in HASMC. Moreover, NOS inhibitor (L-NAME) and anti-TLR 4mAb reduced the LPS-induced NO, IL-8 and VEGF production and ICAM-1 expression. Additionally, TLR4 expression was reduced by NOS inhibitor. Taken together, these results indicate that LPS-induced inflammatory responses are regulated by TLR4 expression and NO production.


Assuntos
Aorta/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Miócitos de Músculo Liso/efeitos dos fármacos , Óxido Nítrico/biossíntese , Receptor 4 Toll-Like/imunologia , Anticorpos Monoclonais/farmacologia , Aorta/citologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Humanos , Molécula 1 de Adesão Intercelular/imunologia , Interleucina-8/imunologia , Miócitos de Músculo Liso/citologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/imunologia , Molécula 1 de Adesão de Célula Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/imunologia
16.
Phytomedicine ; 49: 66-74, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30217263

RESUMO

BACKGROUND: Rhein (4, 5-dihydroxyanthraquinone-2-carboxylic acid), a natural anthraquinone derivative, is a traditional Chinese herb that has been used as a medication in many Asian countries. It has been used as a laxative and stomach drug for a long time in both China and Korea. It is well-known to have many pharmacological activities, such as anti-cancer, anti-bacterial, anti-fungal, anti-oxidant, anti-atherogenic, anti-angiogenic, anti-fibrosis, anti-inflammatory, hepatoprotective, and nephroprotective properties. However, little is known about how rhein may affect the differentiation activities in acute promyelocytic leukemia (APL) cells. PURPOSE: The present study was designed to examine the anti-leukemic effects of rhein against APL cells and to explore the underlying mechanism. METHODS: Cell viability was investigated by MTS assay. To examine the differentiation activities in APL cells, the cell surface molecules (CD11b, CD14, CCR1 and CCR2), phagocytosis, reactive oxygen species (ROS) were determined by flow cytometry. Also, induction of caspase-3 activity and reduction of mitochondrial membrane potential (MMP) were determined by flow cytometry. RNA and protein expressions were determined by qRT-PCR and western blotting, respectively. RESULTS: In this study we assessed the role of rhein in treating APL. Interestingly, rhein potentiated all-trans retinoic acid (ATRA)-induced macrophage differentiation in NB4 cells by inducing changes in morphology, expression of the differentiation markers CD11b and CD14, ROS production, phagocytic activity, and expression of CCR1 and CCR2. Signaling through CD11b was found to be dependent on ERK activation. Additionally, rhein induced APL cell death by activating apoptosis and suppressing the mTOR pathway. CONCLUSION: Therefore, we suggest that a combination of rhein and ATRA carries strong therapeutic potential through the beneficial differentiation of APL cells. Moreover, rhein causes cell death via the activation of apoptosis and suppression of survival signals in APL cells. In combination with the ability of rhein to promote functional macrophage differentiation in APL, these properties suggest that a combined treatment of rhein and ATRA has great potential as an anti-leukemic therapy for APL.


Assuntos
Antraquinonas/farmacologia , Antineoplásicos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Leucemia Promielocítica Aguda/patologia , Tretinoína/farmacologia , Apoptose/efeitos dos fármacos , Antígeno CD11b/metabolismo , Caspase 3/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Receptores de Lipopolissacarídeos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Potencial da Membrana Mitocondrial , Fagocitose , Espécies Reativas de Oxigênio/metabolismo , Receptores CCR1/metabolismo , Receptores CCR2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
17.
J Leukoc Biol ; 79(2): 330-8, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16275888

RESUMO

Human monocytes and neutrophils play major roles in clearing bacteria from human blood and tissues. We found that the herpes virus entry mediator (HVEM) was highly expressed in monocytes and neutrophils, and its interaction with "homologous to lymphotoxins, shows inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM/tumor necrosis factor (TNF)-related 2" (LIGHT) enhanced bactericidal activity against Listeria monocytogenes and Staphylococcus aureus. The LIGHT-HVEM interaction increased levels of phagocytosis, interleukin (IL)-8, TNF-alpha, nitric oxide (NO), and reactive oxygen species (ROS) in monocytes and neutrophils. Anti-HVEM monoclonal antibody was able to block LIGHT-induced bactericidal activity, cytokine production (IL-8 and TNF-alpha), and ROS generation. Moreover, inhibition of ROS and NO production blocked LIGHT-induced bactericidal activity. Our results indicate that the LIGHT/HVEM interaction in monocytes and neutrophils contributes to antibacterial activity.


Assuntos
Linfotoxina-alfa/farmacologia , Proteínas de Membrana/fisiologia , Monócitos/imunologia , Neutrófilos/imunologia , Receptores do Fator de Necrose Tumoral/imunologia , Receptores Virais/imunologia , Fator 2 Associado a Receptor de TNF/farmacologia , Fator de Necrose Tumoral alfa/fisiologia , Proteínas do Envelope Viral/farmacologia , Anticorpos Monoclonais/farmacologia , Humanos , Interleucina-8/biossíntese , Listeria monocytogenes/efeitos dos fármacos , Listeria monocytogenes/imunologia , Linfotoxina-alfa/antagonistas & inibidores , Linfotoxina-alfa/imunologia , Monócitos/efeitos dos fármacos , Neutrófilos/efeitos dos fármacos , Óxido Nítrico/biossíntese , Fagocitose/efeitos dos fármacos , Fagocitose/imunologia , Espécies Reativas de Oxigênio/imunologia , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Membro 14 de Receptores do Fator de Necrose Tumoral , Receptores Virais/antagonistas & inibidores , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/imunologia , Fator 2 Associado a Receptor de TNF/antagonistas & inibidores , Fator 2 Associado a Receptor de TNF/imunologia , Membro 14 da Superfamília de Ligantes de Fatores de Necrose Tumoral , Fator de Necrose Tumoral alfa/biossíntese , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/imunologia
18.
Exp Hematol ; 34(4): 407-13, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16569587

RESUMO

Bacterial infections are major life-threatening complications in patients receiving cytotoxic drugs. These infections generally occur during periods of neutropenia. It has been suggested that the incidence of neutropenia correlates with the incidence of infections. A synthetic hexapeptide, WKYMVm, which stimulates phosphoinositide hydrolysis in leukocytes, has been shown to activate microbicidal activities of human polymorphonuclear neutrophils. In this study, we evaluate whether WKYMVm stimulates bactericidal activity in neutrophils obtained from patients who received chemotherapy for solid tumors when they were neutropenic. Eight patients and 11 healthy controls were recruited for the study. Patient neutrophils, on day 0 and at 2 weeks after chemotherapy, were collected. Expression of the WKYMVm peptide receptor, on leukocytes, was analyzed by fluorescein-activated cell sorting. Neutrophil bactericidal assays were performed using both reactive oxygen species generation and intracellular killing. Expression of the WKYMVm peptide receptor on leukocytes showed no difference in the treated patients compared to healthy controls. WKYMVm increased bactericidal activities, in a dose-dependent fashion, of control neutrophils compared to treated patient neutrophils obtained on day 0. WKYMVm markedly stimulated bactericidal activity of treated patient neutrophils obtained at 2 weeks after chemotherapy compared to treated patient neutrophils obtained on day 0. WKYMVm augmented neutrophil bactericidal activity was noted at low concentration but was suppressed at higher concentrations of 5-fluorouracil. WKYMVm augmented neutrophil bactericidal activity was not suppressed by cisplatin. WKYMVm has the potential for increasing neutrophil bactericidal activity in chemotherapy-treated cancer patients.


Assuntos
Neoplasias/metabolismo , Neutropenia/metabolismo , Ativação de Neutrófilo/efeitos dos fármacos , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Adulto , Idoso , Infecções Bacterianas/tratamento farmacológico , Infecções Bacterianas/etiologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/patologia , Atividade Bactericida do Sangue/efeitos dos fármacos , Células Cultivadas , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Relação Dose-Resposta a Droga , Antagonismo de Drogas , Feminino , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Neutropenia/tratamento farmacológico , Neutropenia/etiologia , Neutropenia/patologia , Neutrófilos/patologia , Oligopeptídeos/uso terapêutico , Receptores de Superfície Celular/metabolismo
19.
Eur J Pharmacol ; 804: 52-56, 2017 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-28322836

RESUMO

Previously, we reported that radotinib, a BCR-ABL1 tyrosine kinase inhibitor, induced cytotoxicity in acute myeloid leukemia (AML) cells. However, the effects of radotinib in the subpopulation of c-KIT-positive AML cells were unclear. We observed that low-concentration radotinib had more potent cytotoxicity in c-KIT-positive cells than c-KIT-negative cells from AML patients. To address this issue, cell lines with high c-KIT expression, HEL92.1.7, and moderate c-KIT expression, H209, were selected. HEL92.1.7 cells were grouped into intermediate and high c-KIT expression populations. The cytotoxicity of radotinib against the HEL92.1.7 cell population with intermediate c-KIT expression was not different from that of the population with high c-KIT expression. When H209 cells were grouped into c-KIT expression-negative and c-KIT expression-positive populations, radotinib induced cytotoxicity in the c-KIT-positive population, but not the c-KIT-negative population. Thus, radotinib induces cytotoxicity in c-KIT-positive cells, regardless of the c-KIT expression intensity. Therefore, radotinib induces significant cytotoxicity in c-KIT-positive AML cells, suggesting that radotinib is a potential target agent for the treatment of c-KIT-positive malignancies including AML.


Assuntos
Benzamidas/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirazinas/farmacologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Benzamidas/uso terapêutico , Linhagem Celular Tumoral , Feminino , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , Pirazinas/uso terapêutico
20.
Sci Rep ; 7(1): 15278, 2017 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-29127384

RESUMO

Dasatinib and radotinib are oral BCR-ABL tyrosine kinase inhibitors that were developed as drugs for the treatment of chronic myeloid leukemia. We report here that the c-KIT (CD117) targeting with dasatinib and radotinib promotes acute myeloid leukemia (AML) cell death, and c-KIT endocytosis is essential for triggering c-KIT-positive AML cell death by dasatinib and radotinib during the early stages. In addition, dasatinib and radotinib reduce heat shock protein 90ß (HSP90ß) expression and release Apaf-1 in c-KIT-positive AML cells. Finally, this activates a caspase-dependent apoptotic pathway in c-KIT-positive AML cells. Moreover, the inhibition of c-KIT endocytosis by dynamin inhibitor (DY) reversed cell viability and c-KIT expression by dasatinib and radotinib. HSP90ß expression was recovered by DY in c-KIT-positive AML cells as well. Furthermore, the effect of radotinib on c-KIT and HSP90ß showed the same pattern in a xenograft animal model using HEL92.1.7 cells. Therefore, dasatinib and radotinib promote AML cell death by targeting c-KIT. Taken together, these results indicate that dasatinib and radotinib treatment have a potential role in anti-leukemic therapy on c-KIT-positive AML cells.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Dasatinibe/farmacologia , Sistemas de Liberação de Medicamentos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Proteínas Proto-Oncogênicas c-kit/análise , Pirazinas/farmacologia , Animais , Fator Apoptótico 1 Ativador de Proteases/genética , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Linhagem Celular Tumoral , Feminino , Proteínas de Choque Térmico HSP90/biossíntese , Proteínas de Choque Térmico HSP90/genética , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Nus , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA