Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 21(1): 631, 2023 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-37717019

RESUMO

BACKGROUND: Increasing evidence suggests that hepatocellular carcinoma (HCC) stem cells (LCSCs) play an essential part in HCC recurrence, metastasis, and chemotherapy and radiotherapy resistance. Multiple studies have demonstrated that stemness-related genes facilitate the progression of tumors. However, the mechanism by which stemness-related genes contribute to HCC is not well understood. Here, we aim to construct a stemness-related score (SRscores) model for deeper analysis of stemness-related genes, assisting with the prognosis and individualized treatment of HCC patients.Further, we found that the gene LPCAT1 was highly expressed in tumor tissues by immunohistochemistry, and sphere-forming assay revealed that knockdown of LPCAT1 inhibited the sphere-forming ability of hepatocellular carcinoma cells. METHODS: We used the TCGA-LIHC dataset to screen stemness-related genes of HCC from the MSigDB database. Prognosis, tumor microenvironment, immunological checkpoints, tumor immune dysfunction, rejection, treatment sensitivity, and putative biological pathways were examined. Random forest created the SRscores model. The anti-PD-1/anti-CTLA4 immunotherapy, tumor mutational burden, medication sensitivity, and cancer stem cell index were compared between the high- and low-risk score groups. We also examined risk scores for different cell types using single-cell RNA sequencing data and correlated transcription factor activity in cancer stem cells with SRscores genes. Finally, we tested core marker expression and biological functions. RESULTS: Patients can be divided into two subtypes (Cluster1 and Cluster2) based on the TCGA-LIHC dataset's identification of 11 stemness-related genes. Additionally, a SRscores was developed based on subtypes. Cluster2 and the group with the lowest SRscores had superior survival and immunotherapy response than Cluster1 and the group with the highest SRscores. The group with a high SRscores was significantly more enriched in classical tumor pathways than the group with a low SRscores. Multiple transcription factors and SRscores genes are correlated. The core gene LPCAT1 is highly expressed in rat liver cancer tissues and promotes tumor cell sphere formation. CONCLUSION: A SRscores model can be utilized to predict the prognosis of HCC patients as well as their response to immunotherapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Ratos , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Imunoterapia , Bioensaio , Linhagem Celular , Microambiente Tumoral
2.
Antioxidants (Basel) ; 12(5)2023 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-37237856

RESUMO

Weightlessness in space leads to bone loss, muscle atrophy, and impaired immune defense in astronauts. Mesenchymal stem cells (MSCs) play crucial roles in maintaining the homeostasis and function of the tissue. However, how microgravity affects the characteristics MSCs and the related roles in the pathophysiological changes in astronauts remain barely known. Here we used a 2D-clinostat device to simulate microgravity. Senescence-associated-ß-galactosidase (SA-ß-gal) staining and the expression of senescent markers p16, p21, and p53 were used to evaluate the senescence of MSCs. Mitochondrial membrane potential (mΔΨm), reactive oxygen species (ROS) production, and ATP production were used to evaluate mitochondrial function. Western blot and immunofluorescence staining were used to investigate the expression and localization of Yes-associated protein (YAP). We found that simulated microgravity (SMG) induced MSC senescence and mitochondrial dysfunction. Mito-TEMPO (MT), a mitochondrial antioxidant, restored mitochondrial function and reversed MSC senescence induced by SMG, suggesting that mitochondrial dysfunction mediates SMG-induced MSC senescence. Further, it was found that SMG promoted YAP expression and its nuclear translocation in MSCs. Verteporfin (VP), an inhibitor of YAP, restored SMG-induced mitochondrial dysfunction and senescence in MSCs by inhibiting YAP expression and nuclear localization. These findings suggest that YAP inhibition alleviates SMG-induced MSC senescence via targeting mitochondrial dysfunction, and YAP may be a potential therapeutic target for the treatment of weightlessness-related cell senescence and aging.

3.
Exp Cell Res ; 355(2): 172-181, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28392353

RESUMO

Migration of bone marrow-derived mesenchymal stem cells (BMSCs) plays an important role in many physiological and pathological settings, including wound healing. During the migration of BMSCs through interstitial tissues, the movement of the nucleus must be coordinated with the cytoskeletal dynamics, which in turn affects the cell migration efficiency. Our previous study indicated that osteopontin (OPN) significantly promotes the migration of rat BMSCs. However, the nuclear behaviors and involved molecular mechanisms in OPN-mediated BMSC migration are largely unclear. In the present study, using an atomic force microscope (AFM), we found that OPN could decrease the nuclear stiffness of BMSCs and reduce the expression of lamin A/C, which is the main determinant of nuclear stiffness. Increased lamin A/C expression attenuates BMSC migration by increasing nuclear stiffness. Decreased lamin A/C expression promotes BMSC migration by decreasing nuclear stiffness. Furthermore, OPN promotes BMSC migration by diminishing lamin A/C expression and decreasing nuclear stiffness via the FAK-ERK1/2 signaling pathway. This study provides strong evidence for the role of nuclear mechanics in BMSC migration as well as new insight into the molecular mechanisms of OPN-promoted BMSC migration.


Assuntos
Células da Medula Óssea/citologia , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteopontina/farmacologia , Animais , Núcleo Celular/metabolismo , Células-Tronco Mesenquimais/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Ratos , Ratos Sprague-Dawley
4.
Sci Rep ; 6: 18809, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26742689

RESUMO

We have previously reported that a synthetic mechano-growth factor (MGF) C-terminal E-domain with 25 amino acids (MGF-C25E) promotes rat tenocyte migration through the FAK-ERK1/2 signaling pathway. However, the role of the nucleus in MGF-C25E-promoted tenocyte migration and the molecular mechanisms involved remain unclear. In this study, we demonstrate that MGF-C25E increases the Young's modulus of tenocytes through the FAK-ERK1/2 signaling pathway. This increase is not accompanied by an obvious change in the expression of Lamin A/C but is accompanied by significant chromatin condensation, indicating that MGF-C25E-induced chromatin condensation may contribute to the increased nuclear stiffness. Moreover, DNA methylation is observed in MGF-C25E-treated tenocytes. Inhibition of DNA methylation suppresses the elevation in chromatin condensation, in nuclear stiffness, and in tenocyte migration induced by MGF-C25E. The inhibition of the focal adhesion kinase (FAK) or extracellular signal regulated kinase 1/2 (ERK1/2) signals represses MGF-C25E-promoted DNA methylation. It also abolishes chromatin condensation, nuclear stiffness, and cell migration. Taken together, our results suggest that MGF-C25E promotes tenocyte migration by increasing nuclear stiffness via the FAK-ERK1/2 signaling pathway. This provides strong evidence for the role of nuclear mechanics in tenocyte migration and new insight into the molecular mechanisms of MGF-promoted tenocyte migration.


Assuntos
Núcleo Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/genética , Fator de Crescimento Insulin-Like I/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Tenócitos/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Núcleo Celular/química , Núcleo Celular/metabolismo , Cromatina/química , Cromatina/efeitos dos fármacos , Cromatina/metabolismo , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Flavonoides/farmacologia , Quinase 1 de Adesão Focal/metabolismo , Regulação da Expressão Gênica , Dureza/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/biossíntese , Masculino , Microscopia de Força Atômica , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Cultura Primária de Células , Domínios Proteicos , Inibidores de Proteínas Quinases/farmacologia , Quinolonas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Sulfonas/farmacologia , Tenócitos/citologia , Tenócitos/metabolismo
5.
J Biomech ; 49(1): 45-52, 2016 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-26627368

RESUMO

Biomechanical properties and cytoskeletal organization of cancer cells are known to be closely related with their aggressive phenotype. In this study, based on atomic force microscopy (AFM), we aimed to evaluate the mechanical property of liver cancer stem-like cells (LCSCs) and compare it with human hepatoma cells (HHCs). LCSCs were enriched from human hepatoma cell line MHCC97H through a sphere culture system. AFM nanoindentation was investigated as a method for measuring the cell stiffness, and reflecting by Young׳s modulus. Microfilament bundles of F-actin were observed with immunofluorescence staining by confocal microscopy. We found that LCSCs show lower Young׳s modulus and higher migration ability compared to MHCC97H cells. Moreover, the decrease in Young׳s modulus is accompanied with a dramatic decline in F-actin content. These results demonstrated a close relationship between the cell Young׳s modulus and metastatic potential of HHCs, which suggest that Young׳s modulus detected by AFM can be used to evaluate metastatic potential of cancer cells.


Assuntos
Microscopia de Força Atômica , Células-Tronco Neoplásicas/citologia , Actinas/metabolismo , Antineoplásicos/química , Fenômenos Biomecânicos , Carcinoma Hepatocelular/patologia , Linhagem Celular , Movimento Celular , Ensaios de Seleção de Medicamentos Antitumorais , Módulo de Elasticidade , Humanos , L-Lactato Desidrogenase , Neoplasias Hepáticas/patologia , Metástase Neoplásica , Fenótipo
6.
Biochem Biophys Res Commun ; 441(1): 202-7, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24140053

RESUMO

Mechano-growth factor (MGF) generated by cells in response to mechanical stimulation has been identified as a mechano effector molecule, playing a key role in regulating mesenchymal stem cell (MSC) function, including proliferation and migration. However, the mechanism(s) underlying how MGF-induced MSC migration occurs is still unclear. In the present study, MGF motivated migration of rat MSCs (rMSCs) in a concentration-dependent manner and optimal concentration of MGF at 50 ng/mL (defined as MGF treatment in this paper) was demonstrated. Notably, enhancement of mechanical properties that is pertinent to cell migration, such as cell traction force and cell stiffness were found to respond to MGF treatment. Furthermore, MGF increased phosphorylation of extracellular signal-regulated kinase (ERK), ERK inhibitor (i.e., PD98059) suppressed ERK phosphorylation, and abolished MGF-induced rMSC migration were found, demonstrating that ERK is involved molecule for MGF-induced rMSC migration. These in vitro evidences of MGF-induced rMSC migration and its direct link to altering rMSC mechanics and activating the ERK pathway, uncover the underlying biomechanical and biological mechanisms of MGF-induced rMSC migration, which may help find MGF-based application of MSC in clinical therapeutics.


Assuntos
Fenômenos Biomecânicos/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/enzimologia , Animais , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Ativação Enzimática/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
7.
Cell Biochem Biophys ; 65(3): 455-62, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23086356

RESUMO

The use of mesenchymal stem cells (MSCs) for therapeutic applications has attracted great attention because MSCs home to and engraft to injured tissues after in vivo administration. The expression of osteopontin (OPN) is elevated in response to injury and inflammation, and its role on rat bone marrow-derived mesenchymal stem cells (rMSCs)-directed migration has been elucidated. However, the signaling pathways through the activation of which OPN promotes rMSCs migration and the involvement of cell mechanics during OPN-mediating rMSCs migration have not been well studied. In this study, we found that OPN activated focal adhesion kinase (FAK) and extracellular signal-regulated kinase (ERK) signaling pathways by the ligation of integrin ß1 in rMSCs. Inhibitors of FAK and ERK pathways inhibited OPN-induced rMSCs migration, indicating the possible involvement of FAK and ERK activation in OPN-induced migration in rMSCs. In addition, atomic force microscopy analysis showed that OPN reduced cell stiffness in rMSCs via integrin ß1, FAK, and ERK pathways, suggesting that the promotion of rMSCs migration might partially be contributing to the decrease in cell stiffness stimulated by OPN. To further examine the role of OPN on cell motility and stiffness, actin cytoskeleton of rMSCs was observed. The reduced well-defined F-actin filaments and the promoted formation of pseudopodia in rMSCs induced by OPN explained the reduction in cell stiffness and the increase in cell migration. The current study data have shown for the first time that OPN binding to integrin ß1 promotes rMSCs migration through the activation of FAK and ERK pathways, which may be attributed to the change in cell stiffness caused by the reduction in the amount of organized actin cytoskeleton.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Integrina beta1/metabolismo , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteopontina/farmacologia , Citoesqueleto de Actina/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Módulo de Elasticidade/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Microscopia de Força Atômica , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
9.
Biochim Biophys Acta ; 1800(3): 380-4, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19914350

RESUMO

BACKGROUND: Dicer is a multidomain ribonuclease III enzyme involved in the biogenesis of microRNAs (miRNAs) and small interfering RNAs (siRNAs); depletion of Dicer was found to impair the migration of endothelial cells. METHODS: siRNA transfection, cell migration assay, real-time RT-PCR, chromatin immunoprecipitation, Western blotting, ELISA, caspase-3 activity assay, and annexin-V-FITC assay were utilized. RESULTS: Knockdown of Dicer impairs the migratory capacity of HEK293T cells and induces fibronectin-1. The upregulation of fibronectin-1 is dependent on Egr1. Fibronectin-1/Dicer double-knockdown cells showed a marked increase in apoptosis compared with fibronectin-1 single knockdown cells. CONCLUSIONS: Decreased Dicer expression induces fibronectin-1 expression via an Egr1-dependent manner. GENERAL SIGNIFICANCE: Our data suggest that upregulation of fibronectin-1 protects Dicer knockdown HEK293T cells against apoptosis.


Assuntos
Proteína 1 de Resposta de Crescimento Precoce/biossíntese , Fibronectinas/genética , Técnicas de Silenciamento de Genes , Rim/fisiologia , Ribonuclease III/genética , Apoptose , Western Blotting , Linhagem Celular , Movimento Celular/fisiologia , Cromatina/genética , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Fibronectinas/fisiologia , Expressão Gênica , Humanos , Rim/embriologia , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
10.
Cytotechnology ; 61(1-2): 1-10, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19842053

RESUMO

Recent evidences have suggested that humoral factors released from the appropriate co-cultured cells influenced the expansion and differentiation of mesenchymal stem cells (MSCs). However, little is known about the proliferation and differentiation of MSCs subjected to co-culture condition with tenocytes. In this study, we aimed to establish a co-culture system of MSCs and tenocytes and investigate the proliferation and tendon/ligament related gene expression of MSCs. MTT assay was used to detect the expansion of MSCs. Semi-quantitative RT-PCR was performed to investigate the expression of proliferation associated c-fos gene and tendon/ligament related genes, including type I collagen (Col I), type III collagen (Col III), tenascin C and scleraxis. Significant increase in MSCs expansion was observed after 3 days of co-culture with tenocytes. The c-fos gene expression was found distinctly higher than for control group on day 4 and day 7 of co-culture. The mRNA expression of four tendon/ligament related genes was significantly up-regulated after 14 days of co-culture with tenocytes. Thus, our research indicates that indirect co-culture with tenocytes promotes the proliferation and mRNA expression of tendon/ligament related genes in MSCs, which suggests a directed differentiation of MSCs into tendon/ligament.

11.
Med Microbiol Immunol ; 198(1): 27-32, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18685862

RESUMO

Hepatitis B virus (HBV) replicates in most tumor tissues of patients with HBV-associated hepatocellular carcinoma (HCC). In the present study, we have shown that the expression of HBV in the HCC cell lines, HepG2 and Huh7, down-regulated the expression of MHC class I-related molecule A (MICA), a ligand of the NKG2D receptor. Inhibition of HBV expression by small interference RNAs (siRNAs) in HepG2.2.15, a cell line that constitutively expresses HBV, induced up-regulation of MICA. The up-regulation of MICA increased the lysis of HepG2.2.15 cells by NK cells. Our results suggest that HBV compromises the innate immune system in HCC patients and that inhibition of HBV replication by siRNAs may enhance the antitumor immune response.


Assuntos
Antivirais/farmacologia , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/fisiologia , Hepatócitos/virologia , Antígenos de Histocompatibilidade Classe I/biossíntese , RNA Interferente Pequeno/farmacologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Células Matadoras Naturais/imunologia
12.
Biochem Biophys Res Commun ; 370(4): 578-83, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18395517

RESUMO

5-Aza-2'-deoxycytidine (5-aza-dC), a DNA methyltransferase inhibitor, exerts antitumor activity through induction of cell cycle arrest, apoptosis and DNA damage. In this study, we showed that MHC class I-related chain B (MICB), a ligand of the NKG2D receptor expressed by natural killer cells and activated CD8(+) T cells, was upregulated following 5-aza-dC treatment. The upregulation of MICB was accompanied by promoter DNA demethylation and DNA damage. Furthermore, the upregulation of MICB was partially prevented by pharmacological or genetic inhibition of ataxia telangiectasia mutated (ATM) kinase. Our results suggest that promoter DNA demethylation, in combination with DNA damage, contribute to the upregulation of MICB induced by 5-aza-dC.


Assuntos
Azacitidina/análogos & derivados , Metilases de Modificação do DNA/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Azacitidina/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Citotoxicidade Imunológica/efeitos dos fármacos , Dano ao DNA , Metilação de DNA/efeitos dos fármacos , Decitabina , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores Imunológicos/metabolismo , Receptores de Células Matadoras Naturais , Transcrição Gênica/efeitos dos fármacos
13.
Colloids Surf B Biointerfaces ; 58(2): 271-7, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17499488

RESUMO

Tissues and cells in the body are continuously exposed to a complex mechanical environment. Mechanical stimulations are critical to morphological, developmental and functional states of living cells, and the fashion of the mechanical stimulation applied to the cells is supposed to be extremely important for the induced cell response and function. In this study, we investigated whether mechanical stretch regulates and promotes proliferation of rat bone marrow mesenchymal stem cells (rMSCs) in vitro. rMSCs from rat bone marrow were isolated, purified and subjected to a cyclic equiaxial stretch treatment, and then MTT assay was adopted and expression of c-fos gene was measured by RT-PCR to access cell proliferation. The results demonstrated that OD values of rMSCs increased in a time-dependent and magnitude-dependent manner after exposure to 1 Hz stretch within 15-60 min and 2-8% strain. Expression of c-fos gene in rMSCs subjected to stretch treatment (1 Hz, 8% strain and 60 min) is significantly higher than that of unstimulated control cells. These results suggest that mechanical stretch plays an important role in regulating the cell growth and proliferation, and an appropriate mechanical stretch treatment could promote proliferating capacity of rMSCs.


Assuntos
Células da Medula Óssea/citologia , Proliferação de Células , Células-Tronco Mesenquimais/citologia , Animais , Células Cultivadas , Ratos , Ratos Sprague-Dawley , Estresse Mecânico
14.
Colloids Surf B Biointerfaces ; 48(1): 1-5, 2006 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-16458489

RESUMO

Oxymatrine, a natural quinolizidine alkaloid, has been known having cytotoxic and chemopreventive effects on various cancer cells. To investigate the possible mechanism of oxymatrine's role on cancer cells, in the present study, we examined further the effects of oxymatrine on the growth, proliferation, apoptosis and expression of bcl-2 and p53 gene in human hepatoma SMMC-7721 cells in vitro. Our results show that oxymatrine notably inhibits the growth and proliferation of SMMC-7721 cells and it present a dose-dependence and time-dependence manner within definite reacting dose and time. Oxymatrine block SMMC-7721 cells in G2/M and S phase; prevent cells entering into G0/G1 phase. It results in an obvious accumulation of G2/M and S phase cells while decrease of G0/G1 phase cells. Oxymatrine induce apoptosis of SMMC-7721 cells and apoptotic rate amount to about 60% after treatment with 1.0 mg/ml oxymatrine for 48 h. We also find that oxymatrine down-regulate expression of bcl-2 gene while up-regulate expression of p53 gene. These results demonstrate that oxymatrine inhibit the proliferation and induce apoptosis of human hepatoma SMMC-7721 cells, and suggest that this effect was mediated probably by a significant cell cycle blockage in G2/M and S phase, down-regulation of bcl-2 and up-regulation of p53.


Assuntos
Alcaloides/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Alcaloides/química , Antineoplásicos/química , Carcinoma Hepatocelular/metabolismo , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Fase G2 , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Cinética , Neoplasias Hepáticas/metabolismo , Conformação Molecular , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Quinolizinas , Fase S , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...