Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Phytomedicine ; 128: 155477, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38489890

RESUMO

BACKGROUND: The alleviating effect of paeoniflorin (Pae) on liver fibrosis has been established; however, the molecular mechanism and specific target(s) underlying this effect remain elusive. PURPOSE: This study was to investigate the molecular mechanism underlying the regulatory effect of Pae on hepatic stellate cells (HSCs) activation in liver fibrosis, with a specific focus on the role of Pae in modulating histone methylation modifications. METHODS: The therapeutic effect of Pae was evaluated by establishing in vivo and in vitro models of carbon tetrachloride (CCl4)-induced mice and transforming growth factor ß1 (TGF-ß1)-induced LX-2 cells, respectively. Molecular docking, surface plasmon resonance (SPR), chromatin immunoprecipitation-quantitative real time PCR (ChIP-qPCR) and other molecular biological methods were used to clarify the molecular mechanism of Pae regulating HSCs activation. RESULTS: Our study found that Pae inhibited HSCs activation and histone trimethylation modification in liver of CCl4-induced mice and LX-2 cells. We demonstrated that the inhibitory effect of Pae on the activation of HSCs was dependent on peroxisome proliferator-activated receptor γ (PPARγ) expression and enhancer of zeste homolog 2 (EZH2). Mechanistically, Pae directly binded to EZH2 to effectively suppress its enzymatic activity. This attenuation leaded to the suppression of histone H3K27 trimethylation in the PPARγ promoter region, which induced upregulation of PPARγ expression. CONCLUSION: This investigative not only sheds new light on the precise targets that underlie the remission of hepatic fibrogenesis induced by Pae but also emphasizes the critical significance of EZH2-mediated H3K27 trimethylation in driving the pathogenesis of liver fibrosis.


Assuntos
Tetracloreto de Carbono , Proteína Potenciadora do Homólogo 2 de Zeste , Glucosídeos , Células Estreladas do Fígado , Histonas , Cirrose Hepática , Monoterpenos , PPAR gama , Animais , Glucosídeos/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , PPAR gama/metabolismo , Monoterpenos/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/metabolismo , Histonas/metabolismo , Camundongos , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/induzido quimicamente , Masculino , Humanos , Camundongos Endogâmicos C57BL , Metilação , Fator de Crescimento Transformador beta1/metabolismo , Linhagem Celular , Simulação de Acoplamento Molecular
2.
Pathol Res Pract ; 251: 154885, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37862922

RESUMO

In accordance with the World Health Organization, cancer is the second leading cause of death in patients. In recent years, the number of cancer patients has been growing, and the occurrence of cancer in people is becoming more common, primarily due to lifestyle factors. Yin Yang 1 (YY1) is a transcription factor that is widespread throughout. It is a zinc finger protein, falling under the GLI-Kruppel class. YY1 is known to regulate transcriptional activation and repression of various genes associated with different cellular processes such as DNA repair, autophagy, cell survival and apoptosis, and cell division. Meanwhile, EZH2 is a histone-lysine N-methyltransferase enzyme encoded by gene 7 in humans. Its main function involves catalyzing the addition of methyl groups to histone H3 at lysine 27 (H3K27me3), and it is involved in regulating CD8 + T cell fate and function. It is a subunit of a Polycomb repressor complex 2 (PRC2). The EZH2 gene encodes for an enzyme that is involved in histone methylation and transcriptional repression. It adds methyl groups to lysine 27 on histone H3 (H3K27me3) with the help of the cofactor S-adenosyl-L-methionine. In addition to its role in epigenetic regulation, EZH2 also acts as a regulator of CD8+ T cell fate and function. EZH2 has been implicated in T Cell Receptor (TCR) signaling via the regulation of actin polymerization. In fact, EZH2 is involved in numerous signaling pathways that lead to tumorigenesis. EZH2 is mutated in cancer and shows overexpression. Due to its mutation and overexpression, the cells that help combat cancer are suppressed and carcinogenicity is promoted. The association of EZH2 and YY1 poses an intriguing mechanism in relation to cancer.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste , Neoplasias , Humanos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/genética , Complexo Repressor Polycomb 2/genética , Lisina , Epigênese Genética , Yin-Yang , Neoplasias/genética , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
3.
Adv Sci (Weinh) ; 10(25): e2207549, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37401236

RESUMO

LncRNAs play a critical role in oral squamous cell carcinoma (OSCC) progression. However, the function and detailed molecular mechanism of most lncRNAs in OSCC are not fully understood. Here, a novel nuclear-localized lncRNA, DUXAP9 (DUXAP9), that is highly expressed in OSCC is identified. A high level of DUXAP9 is positively associated with lymph node metastasis, poor pathological differentiation, advanced clinical stage, worse overall survival, and worse disease-specific survival in OSCC patients. Overexpression of DUXAP9 significantly promotes OSCC cell proliferation, migration, invasion, and xenograft tumor growth and metastasis, and upregulates N-cadherin, Vimentin, Ki67, PCNA, and EZH2 expression and downregulates E-cadherin in vitro and in vivo, whereas knockdown of DUXAP9 remarkably suppresses OSCC cell proliferation, migration, invasion, and xenograft tumor growth in vitro and in vivo in an EZH2-dependent manner. Yin Yang 1 (YY1) is found to activate the transcriptional expression of DUXAP9 in OSCC. Furthermore, DUXAP9 physically interacts with EZH2 and inhibits EZH2 degradation via the suppression of EZH2 phosphorylation, thereby blocking EZH2 translocation from the nucleus to the cytoplasm. Thus, DUXAP9 can serve as a promising target for OSCC therapy.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , RNA Longo não Codificante , Humanos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Yin-Yang , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Bucais/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Quinase CDC2
4.
J Med Chem ; 66(11): 7629-7644, 2023 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-37203326

RESUMO

The first-generation enhancer of zeste homologue 2 (EZH2) inhibitors suffer from several limitations, such as high dosage, cofactor S-adenosylmethionine (SAM) competition, and acquired drug resistance. Development of covalent EZH2 inhibitors that are noncompetitive with cofactor SAM offers an opportunity to overcome these disadvantages. The structure-based design of compound 16 (BBDDL2059) as a highly potent and selective covalent inhibitor of EZH2 is presented in this context. 16 inhibits EZH2 enzymatic activity at sub-nanomolar concentrations and achieves low nanomolar potencies in cell growth inhibition. The kinetic assay revealed that 16 is noncompetitive with the cofactor SAM, providing the basis for its superior activity over noncovalent and positive controls by reducing competition with cofactor SAM and offering a preliminary proof for its covalent inhibition nature. Mass spectrometric analysis and washout experiments firmly establish its covalent inhibition mechanism. This study demonstrates that covalent inhibition of EZH2 can offer a new opportunity for the development of promising new-generation drug candidates.


Assuntos
Lisina , S-Adenosilmetionina , S-Adenosilmetionina/farmacologia , S-Adenosilmetionina/química , Proteína Potenciadora do Homólogo 2 de Zeste , Complexo Repressor Polycomb 2 , Proliferação de Células , Linhagem Celular Tumoral
5.
Med Oncol ; 40(7): 185, 2023 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-37212947

RESUMO

Non-Small Cell Lung Cancer (NSCLC) is the leading cause of death in all countries alike. In the current study, we have found out that Histone H3Lys4trimethylation is abnormal on YY1 in CD4+T Helper (TH) cells of NSCLC patients which is evident by Histone H3Lys27 trimethylation mediated via EZH2. We investigated the status of Yin Yang 1 (YY1) and the involvement of certain transcription factors that lead to tumorigenesis after depleting endogenous EZH2 in vitro by CRISPR/Cas9 in the CD4+TH1-or-TH2-polarized cells isolated initially as CD4+TH0 cells from the PBMC of the control subjects and patients suffering from NSCLC. After depletion of endogenous EZH2, RT-qPCR based mRNA expression analysis showed that there was an increase in the expression of TH1 specific genes and a decrease in the expression of TH2 specific genes in NSCLC patients CD4+TH cells. We can conclude that this group of NSCLC patients may have the tendency at least in vitro to elucidate adaptive/protective immunity through the depletion of endogenous EZH2 along with the reduction in the expression of YY1. Moreover, depletion of EZH2 not only suppressed the CD4+CD25+FOXP3+Regulatory T cells (Treg) but also it aided the generation of CD8+Cytotoxic T Lymphocytes (CTL) which were involved in killing of the NSCLC cells. Thus the transcription factors involved in EZH2 mediated T cell differentiation linked to malignancies offers us an appealing avenue of targeted therapeutic intervention for NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Fatores de Transcrição/genética , Histonas/metabolismo , Neoplasias Pulmonares/patologia , Epigênese Genética , Leucócitos Mononucleares , Yin-Yang , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Diferenciação Celular/genética , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
6.
Am J Chin Med ; 51(4): 1041-1066, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37120706

RESUMO

Metastasis of osteosarcoma is an important adverse factor affecting patients' survival, and cancer stemness is the crucial cause of distant metastasis. Capsaicin, the main component of pepper, has been proven in our previous work to inhibit osteosarcoma proliferation and enhance its drug sensitivity to cisplatin at low concentrations. This study aims to further explore the anti-osteosarcoma effect of capsaicin at low concentrations (100[Formula: see text][Formula: see text]M, 24[Formula: see text]h) on stemness and metastasis. The stemness of human osteosarcoma (HOS) cells was decreased significantly by capsaicin treatment. Additionally, the capsaicin treatment's inhibition of cancer stem cells (CSCs) was dose-dependent on both sphere formation and sphere size. Meanwhile, capsaicin inhibited invasion and migration, which might be associated with 25 metastasis-related genes. SOX2 and EZH2 were the most two relevant stemness factors for capsaicin's dose-dependent inhibition of osteosarcoma. The mRNAsi score of HOS stemness inhibited by capsaicin was strongly correlated with most metastasis-related genes of osteosarcoma. Capsaicin downregulated six metastasis-promoting genes and up-regulated three metastasis-inhibiting genes, which significantly affected the overall survival and/or disease-free survival of patients. In addition, the CSC re-adhesion scratch assay demonstrated that capsaicin inhibited the migration ability of osteosarcoma by inhibiting its stemness. Overall, capsaicin exerts a significant inhibitory effect on the stemness expression and metastatic ability of osteosarcoma. Moreover, it can inhibit the migratory ability of osteosarcoma by suppressing its stemness via downregulating SOX2 and EZH2. Therefore, capsaicin is expected to be a potential drug against osteosarcoma metastasis due to its ability to inhibit cancer stemness.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Capsaicina/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Osteossarcoma/tratamento farmacológico , Osteossarcoma/genética , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/farmacologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXB1/farmacologia
7.
J Gastroenterol Hepatol ; 38(3): 441-450, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36652457

RESUMO

BACKGROUND AND AIM: Whether vitamin D3 (VD3) supplementation is associated with improved liver fibrosis is controversial. METHODS: Liver fibrosis models were treated with VD3, active VD (1,25-OH2 Vitamin D3), or collaboration with GSK126 (Ezh2 inhibitor), respectively. Hepatic stellate cells (HSCs) were co-cultured with hepatocytes and then stimulated with TGF-ß. Autophagy of hepatocytes was determined after the intervention of 1,25-OH2 Vitamin D3 and GSK126. Also, the active status of HSCs and the mechanism with 1,25-OH2 Vitamin D3 and GSK126 intervention were detected. RESULTS: 1,25-OH2 Vitamin D3, but not VD3, is involved in anti-fibrosis and partially improves liver function, which might be associated with related enzymes and receptors (especially CYP2R1), leading to decreased of its biotransformation. GSK126 plays a synergistic role in anti-fibrosis. The co-culture system showed increased hepatocyte autophagy after HSCs activation. Supplementation with 1,25-OH2 Vitamin D3 or combined GSK126 reduced these effects. Further studies showed that 1,25-OH2 Vitamin D3 promoted H3K27 methylation of DKK1 promoter through VDR/Ezh2 due to the weakening for HSCs inhibitory signal. CONCLUSIONS: VD3 bioactive form 1,25-OH2 Vitamin D3 is responsible for the anti-fibrosis, which might have bidirectional effects on HSCs by regulating histone modification. The inhibitor of Ezh2 plays a synergistic role in this process.


Assuntos
Colecalciferol , Proteína Potenciadora do Homólogo 2 de Zeste , Inibidores Enzimáticos , Células Estreladas do Fígado , Cirrose Hepática , Humanos , Colecalciferol/metabolismo , Colecalciferol/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/farmacologia , Células Estreladas do Fígado/metabolismo , Hepatócitos/metabolismo , Fígado/patologia , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Fator de Crescimento Transformador beta/metabolismo , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico
8.
Altern Ther Health Med ; 29(2): 42-49, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36239569

RESUMO

Context: Endometriosis refers to the appearance of ectopic endometrioid tissue outside the uterus. Low PCDH10 expression has been associated with enhancer of zeste homolog 2 (EZH2), which catalyzes histone 3 (H3K27me3). H3K27me3 is an epigenetic marker associated with endometriosis. Objective: The study intended to explore the influence of protocadherin 10 (PCDH10) on the invasion and migration of endometrial stromal cells in endometriosis as well as its mechanism. Design: The research team designed a laboratory study using endometrial tissue. Setting: The study took place in Department of Obstetrics and Gynecology at South University of Science and Technology Hospital in Shenzhen, Guangdong Province, China. Participants: Participants were 10 patients with ovarian endometriosis (ovarian chocolate cysts) who were undergoing surgical treatment at the hospital between January and December 2019. The endometrial tissue of those participants became the endometriosis group. Other participants with normal endometrial tissue became the controls (n=10). Outcome Measures: The research team collected tissues from participants and used immunofluorescence, real-time quantitative polymerase chain reaction (qPCR), and Western blot assay to determine the expression levels of PCDH10, enhancer of zeste homolog 2 (EZH2), and histone H3 (H3K27me3). The team cultured endometrial stromal cells from participants primarily to detect the effects of silencing EZH2 on PCDH10 and H3K27me3 expression. The team used a Transwell assay and scratch test to examine the influence of silencing EZH2 on invasion and migration of endometrial stromal cells and applied chromatin immunoprecipitation to determine H3K27me3 enrichment in the PCDH10 gene promoter region. Results: PCDH10 in heterotopic endometrial tissues of endometriosis patients had low expression, while EZH2 and H3K27me3 were highly expressed. Silencing EZH2 inhibited EZH2 protein expression, increased PCDH10 expression, and inhibited invasion and migration of endometrial stromal cells by increasing PCDH10 expression. Silencing EZH2 also reduced H3K27me3 enrichment in PCDH10 promoter region. Conclusions: Low PCDH10 expression may be associated with high EZH2 expression and H3K27me3 enrichment in endometriosis patients, which promotes the migration and invasion of endometrial stromal cells. This connection provides a theoretical basis for the treatment of endometriosis.


Assuntos
Endometriose , Proteína Potenciadora do Homólogo 2 de Zeste , Feminino , Humanos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/metabolismo , Metilação , Endometriose/genética , Endometriose/metabolismo , Células Estromais/metabolismo , Protocaderinas
9.
Phytomedicine ; 106: 154390, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35994849

RESUMO

BACKGROUND: Epigenetics regulating gene expression plays important role in kidney fibrosis. Natural products originating from diverse sources including plants and microorganisms are capable to influence epigenetic modifications. Gambogenic acid (GNA) is a caged xanthone extracted from gamboge resin, exudation of Garcinia hanburyi Hook.f., and the effect of GNA on kidney fibrosis with its underlying mechanism on epigenetics remains unknown. PURPOSE: This study aimed to explore the role of GNA against kidney fibrogenesis by histone methylation mediating gene expression. METHODS: Two experimental mice of unilateral ureteral obstruction (UUO) and folic acid (FA) were given two dosages of GNA (3 and 6 mg/kg/d). TGF-ß1 was used to stimulate mouse tubular epithelial (TCMK-1) cells and siRNAs were transfected to verify the underlying mechanisms of GNA. Histological changes were evaluated by HE, MASSON stainings, immunohistochemistry and immunofluorescence. Western blot and qPCR were used to measure protein/gene transcription levels. RESULTS: GNA dose-dependently alleviated UUO-induced kidney fibrosis and FA-induced kidney early fibrosis, indicated by the pathology and fibrotic factor changes (α-SMA, collagen I, collagen VI, and fibronectin). Mechanically, GNA reduced enhancer of zeste homolog 2 (EZH2) and H3K27me3, promoted Smad7 transcription, and inhibited TGF-ß/Smad3 fibrotic signaling in injured kidneys. Moreover, with TGF-ß1-induced EZH2 increasing, GNA suppressed α-SMA, fibronectin and collagen levels in tubular epithelial TCMK-1 cells. Although partially decreasing EZH2, GNA did not influence fibrotic signaling in Smad7 siRNA-transfected TCMK-1 cells. CONCLUSION: Epigenetic inhibition of EZH2 by GNA ameliorated kidney fibrogenesis via regulating Smad7-meidated TGF-ß/Smad3 signaling.


Assuntos
Produtos Biológicos , Nefropatias , Obstrução Ureteral , Xantonas , Animais , Produtos Biológicos/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Fibronectinas/metabolismo , Fibrose , Ácido Fólico/metabolismo , Histonas/metabolismo , Rim , Nefropatias/metabolismo , Camundongos , RNA Interferente Pequeno/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/patologia , Xantenos , Xantonas/farmacologia
10.
Ann Clin Lab Sci ; 52(1): 27-32, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35181615

RESUMO

OBJECTIVE: Diffuse large B-cell lymphoma (DLBCL) is the most aggressive form of non-Hodgkin's lymphoma. METHODS: We applied morphoproteomics to a tissue microarray of DLBCL cases to uncover commonalities in its biology with therapeutic implications. Morphoproteomic analysis of 9 individual cases of classic DLBCL included immunohistochemical probes to detect silent mating type information regulation 2 homolog 1 (SIRT1), enhancer of Zeste homolog 2 (EZH2) and C-X-C chemokine receptor 4 (CXCR4) and their cellular compartmentalization by bright field microscopy. RESULTS: We detected the expression of SIRT1 in the majority (>50%) of the tumoral nuclei of 8 of 9 cases of DLBCL and of EZH2 expression in the majority (>50%) of the tumoral nuclei in 9 of the 9 cases; and the expression of the tumoral stem cell marker, CXCR4 in the cytoplasmic and/or plasmalemmal compartment at >50% of the tumor cells in all 9 cases of DLBCL. The morphoproteomic findings of SIRT1 and EZH2 expression in DLBCL, for the most part, parallel the morphoproteomic findings in B-cell acute lymphoblastic leukemia. This concordance has pharmacogenomic and therapeutic implications. Similarly, the fact that EZH2 can enhance the expression of tumoral stem cell marker, CXCR4 implies that there is a block in differentiation in DLBCL. CONCLUSION: By targeting the Sirt1, EZH2 and CXCR4 pathways using relatively non-toxic adjuvant therapeutic agents such as metformin, melatonin, curcumin, sulforaphane, vitamin D3 and plerixafor, we should be able to target the biology of DLBCL.


Assuntos
Compostos Heterocíclicos , Linfoma Difuso de Grandes Células B , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Mobilização de Células-Tronco Hematopoéticas , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/genética , Receptores CXCR4/genética , Sirtuína 1/genética , Sirtuína 1/metabolismo
11.
Lab Invest ; 102(2): 185-193, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34802040

RESUMO

Brain tumors are the leading cause of cancer-related death in children. Tazemetostat is an FDA-approved enhancer of zeste homolog (EZH2) inhibitor. To determine its role in difficult-to-treat pediatric brain tumors, we examined EZH2 levels in a panel of 22 PDOX models and confirmed EZH2 mRNA over-expression in 9 GBM (34.6 ± 12.7-fold) and 11 medulloblastoma models (6.2 ± 1.7 in group 3, 6.0 ± 2.4 in group 4) accompanied by elevated H3K27me3 expression. Therapeutic efficacy was evaluated in 4 models (1 GBM, 2 medulloblastomas and 1 ATRT) via systematically administered tazemetostat (250 and 400 mg/kg, gavaged, twice daily) alone and in combination with cisplatin (5 mg/kg, i.p., twice) and/or radiation (2 Gy/day × 5 days). Compared with the untreated controls, tazemetostat significantly (Pcorrected < 0.05) prolonged survival times in IC-L1115ATRT (101% at 400 mg/kg) and IC-2305GBM (32% at 250 mg/kg, 45% at 400 mg/kg) in a dose-dependent manner. The addition of tazemetostat with radiation was evaluated in 3 models, with only one [IC-1078MB (group 4)] showing a substantial, though not statistically significant, prolongation in survival compared to radiation treatment alone. Combining tazemetostat (250 mg/kg) with cisplatin was not superior to cisplatin alone in any model. Analysis of in vivo drug resistance detected predominance of EZH2-negative cells in the remnant PDOX tumors accompanied by decreased H3K27me2 and H3K27me3 expressions. These data supported the use of tazemetostat in a subset of pediatric brain tumors and suggests that EZH2-negative tumor cells may have caused therapy resistance and should be prioritized for the search of new therapeutic targets.


Assuntos
Neoplasias Encefálicas/terapia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adolescente , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzamidas/administração & dosagem , Benzamidas/farmacologia , Compostos de Bifenilo/administração & dosagem , Compostos de Bifenilo/farmacologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Quimiorradioterapia , Criança , Cisplatino/administração & dosagem , Terapia Combinada/métodos , Avaliação Pré-Clínica de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Inibidores Enzimáticos/administração & dosagem , Feminino , Perfilação da Expressão Gênica/métodos , Humanos , Lactente , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Morfolinas/administração & dosagem , Morfolinas/farmacologia , Piridonas/administração & dosagem , Piridonas/farmacologia , Dosagem Radioterapêutica
12.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34948250

RESUMO

Hypoxia is a major obstacle to gastric cancer (GC) therapy and leads to chemoresistance as GC cells are frequently exposed to the hypoxia environment. Apigenin, a flavonoid found in traditional medicine, fruits, and vegetables and an HDAC inhibitor, is a powerful anti-cancer agent against various cancer cell lines. However, detailed mechanisms involved in the treatment of GC using APG are not fully understood. In this study, we investigated the biological activity of and molecular mechanisms involved in APG-mediated treatment of GC under hypoxia. APG promoted autophagic cell death by increasing ATG5, LC3-II, and phosphorylation of AMPK and ULK1 and down-regulating p-mTOR and p62 in GC. Furthermore, our results show that APG induces autophagic cell death via the activation of the PERK signaling, indicating an endoplasmic reticulum (ER) stress response. The inhibition of ER stress suppressed APG-induced autophagy and conferred prolonged cell survival, indicating autophagic cell death. We further show that APG induces ER stress- and autophagy-related cell death through the inhibition of HIF-1α and Ezh2 under normoxia and hypoxia. Taken together, our findings indicate that APG activates autophagic cell death by inhibiting HIF-1α and Ezh2 under hypoxia conditions in GC cells.


Assuntos
Apigenina/metabolismo , Neoplasias Gástricas/metabolismo , Adenilato Quinase/metabolismo , Apigenina/farmacologia , Apoptose , Morte Celular Autofágica/efeitos dos fármacos , Autofagia , Proteína 5 Relacionada à Autofagia/metabolismo , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Morte Celular , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Estresse do Retículo Endoplasmático , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Fosforilação , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Neoplasias Gástricas/fisiopatologia , Serina-Treonina Quinases TOR/metabolismo
13.
Aging (Albany NY) ; 13(21): 24101-24116, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34739394

RESUMO

Currently, conventional treatment is not sufficient to improve the survival of glioma patients. Hence, adopting novel personalized treatment programs is imperative. Curcumol, a Chinese herbal medicine extract from the roots of Rhizoma Curcumae, has attracted significant interest due to its beneficial pharmacological activities. The current study revealed that curcumol inhibited the proliferation, metastasis, self-renewal ability, and TMZ resistance in glioma cells in vitro and in vivo. Next, the potential molecular mechanisms of curcumol in inhibiting glioma were investigated. We found that the long non-coding RNA (lncRNA) FOXD2-As1 might contribute to the effects of curcumol on glioma cells. Enforced expression of FOXD2-As1 attenuated the curcumol-induced reduction in glioma cell proliferation, metastasis, self-renewal ability, and TMZ resistance. Moreover, the forced expression of FOXD2-As1 reversed the inhibitory effect of curcumol on the binding ability of EZH2 and H3K27me3 modification in the promoter regions of anti-oncogenes. Our results showed for the first time that curcumol is effective in inhibiting malignant biological behaviors and TMZ-resistance of glioma cells by suppressing FOXD2-As1-mediated EZH2 activation. Our study offers the possibility of exploiting curcumol as a promising therapeutic agent for glioma treatment and may provide an option for the clinical application of this natural herbal medicine.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Glioma/metabolismo , RNA Longo não Codificante/genética , Sesquiterpenos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/genética , Humanos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Temozolomida/farmacologia
14.
Sci Rep ; 11(1): 21396, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34725436

RESUMO

Both EZH2 and its homolog EZH1 function as histone H3 Lysine 27 (H3K27) methyltransferases and repress the transcription of target genes. Dysregulation of H3K27 trimethylation (H3K27me3) plays an important role in the development and progression of cancers such as hepatocellular carcinoma (HCC). This study investigated the relationship between the expression of EZH1/2 and the level of H3K27me3 in HCC. Additionally, the role of EZH1/2 in cell growth, tumorigenicity, and resistance to sorafenib were also analyzed. Both the lentiviral knockdown and the pharmacological inhibition of EZH1/2 (UNC1999) diminished the level of H3K27me3 and suppressed cell growth in liver cancer cells, compared with EZH1 or EZH2 single knockdown. Although a significant association was observed between EZH2 expression and H3K27me3 levels in HCC samples, overexpression of EZH1 appeared to contribute to enhanced H3K27me3 levels in some EZH2lowH3K27me3high cases. Akt suppression following sorafenib treatment resulted in an increase of the H3K27me3 levels through a decrease in EZH2 phosphorylation at serine 21. The combined use of sorafenib and UNC1999 exhibited synergistic antitumor effects in vitro and in vivo. Combination treatment canceled the sorafenib-induced enhancement in H3K27me3 levels, indicating that activation of EZH2 function is one of the mechanisms of sorafenib-resistance in HCC. In conclusion, sorafenib plus EZH1/2 inhibitors may comprise a novel therapeutic approach in HCC.


Assuntos
Antineoplásicos/uso terapêutico , Benzamidas/uso terapêutico , Carcinoma Hepatocelular/terapia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Indazóis/uso terapêutico , Neoplasias Hepáticas/terapia , Piperazinas/uso terapêutico , Complexo Repressor Polycomb 2/antagonistas & inibidores , Piridonas/uso terapêutico , Sorafenibe/uso terapêutico , Idoso , Animais , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Terapia Genética , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos SCID , Pessoa de Meia-Idade , Complexo Repressor Polycomb 2/genética
15.
Phytomedicine ; 91: 153706, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34517264

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most common cause of cancer-related death worldwide. Curcumin (C) has been extensively investigated in different types of malignancies, including hepatocellular carcinoma, but its physicochemical properties have significantly influenced its clinical use. Several approaches are being explored to enhance curcumin's therapeutic response, including its combination with various drugs. PURPOSE: This study aimed to evaluate the anti-tumor effect of curcumin (C) in combination with F2 (N-n-butyl haloperidol iodide) on hepatocellular carcinoma and its potential underlying mechanism in vitro and in vivo. METHODS: Cell proliferation was evaluated by CCK-8 and colony formation assays, and apoptosis was measured by flow cytometry. The migratory and invasive abilities of Hep3B and SMMC-7721 cells were measured by wound-healing and matrigel transwell assays. In order to investigate the molecular pathways, various experiments such as western blotting, qPCR, RNA-seq, immunostaining and transfection were performed. To evaluate the anti-HCC effects in vivo, a xenograft tumor model was used. RESULTS: Our findings showed that the combination of curcumin (C) & F2 (F2C) strongly inhibited malignant proliferation and migration in SMMC-7721 and Hep3B cells. The F2C treatment downregulates enhancer of zeste homolog 2 (EZH2) transcription and protein expression, which is key epigenetic regulator responsible for HCC development. Moreover, the inhibition of EZH2 by F2C led to Wnt/ß-catenin signaling inhibition by decreasing tri-methylation of histone H3 at lysine 27 (H3K27me3) and long non-coding RNA H19 expression. The inhibition of F2C was associated with the suppression of tumorigenicity in xenograft HCC models. CONCLUSION: These findings suggested that, F2C inhibited HCC formation, migration and its modulatory mechanism seemed to be associated with downregulation of EZH2, silencing Wnt/ß-catenin signaling by interacting with H19, suggesting that F2C may be a promising drug in the clinical treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Curcumina , Haloperidol/análogos & derivados , Neoplasias Hepáticas , RNA Longo não Codificante , Via de Sinalização Wnt/efeitos dos fármacos , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Proliferação de Células , Curcumina/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação Neoplásica da Expressão Gênica , Haloperidol/farmacologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/genética , Camundongos Nus , RNA Longo não Codificante/genética , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Biochem Mol Toxicol ; 35(10): e22870, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34350670

RESUMO

Scutellarein, a flavone found in the perennial herb Scutellaria baicalensis, has antitumorigenic activity in multiple human cancers. However, whether scutellarein can attenuate ovarian cancer (OC) is unclear. This study investigated the effects of scutellarein in OC. In vitro cell viability was assessed using MTT assay whereas proliferation was assessed using 5-ethynyl-2'-deoxyuridine and colony formation assays. Cell apoptosis was detected by an Annexin V-fluorescein isothiocyanate/propidium iodide assay. Wound-healing and Transwell assays were used to determine cell migration and invasion. The differential expression of enhancer of zeste homolog 2 (EZH2) and forkhead box protein O1 (FOXO1) was measured by Quantitative real-time PCR and western blot analysis. We found that scutellarein inhibited viability, migration, invasion of A2780 and SKOV-3 cells, and reduced the expression of EZH2 in OC cells. In addition, FOXO1 was downregulated in OC tissues and cells and negatively regulated by EZH2. Also, scutellarein inhibited tumor growth and metastasis in vivo. In conclusion, scutellarein alleviates OC by the regulation of EZH2/FOXO1 signaling.


Assuntos
Antineoplásicos/administração & dosagem , Apigenina/farmacologia , Apoptose/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Proteína Forkhead Box O1/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Compostos Fitoquímicos/farmacologia , Fitoterapia/métodos , Scutellaria baicalensis/química , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Ovarianas/patologia , Transdução de Sinais/genética , Transfecção , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
17.
J Ethnopharmacol ; 280: 114413, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34265379

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Curcuma kwangsiensis S. G. Lee & C. F. Liang (Guangxi ezhu, in Chinese) has been used as a traditional Chinese medicine (TCM) for approximately 2000 years. Curcumol is one of the major bioactive components of this herb, which has been demonstrated possesses anti-cancer properties, and was recorded in the Chinese Pharmacopoeia 2020 edition. However, most studies mainly focused on the superficial anti-cancer activity, the underlying mechanism remains poorly understood. AIM OF THE STUDY: In the present study, we aimed to investigate the anti-tumor effect of Curcumol on hepatocellular carcinoma (HCC), and elucidate its underlying mechanism from the perspective of epigenetic modification. MATERIALS AND METHODS: The potential anti-cancer properties of Curcumol were evaluated in HepG2 and SMMC-7721 cells. Its effects on cell growth, cell cycle, apoptosis and migration were examined in these HCC cells. Moreover, the lncRNA HOX transcript antisense intergenic RNA (Hotair) and histone methylatic modification were detected by qPCR and Western blotting assays. RESULTS: In the present study, Curcumol was illustrated to suppress cell growth in HCC cells via inducing apoptosis and cell cycle arrest. And it was also found that Curcumol inhibited the invasion and metastasis of HCC as well. As for the mechanism investigation, it was showed that lncRNA Hotair was significantly downregulated by Curcumol in HCC cells. As is well known, Hotair recruited histone methyltransferase enhancer of zeste homolog 2 (EZH2) to exert transcriptional regulation. Our results showed that EZH2 were downregulated by Curcumol in HCC cells, and thus disrupted the trimethylation of H3K9 and H3K27 which were specifically catalyzed by EZH2. CONCLUSIONS: In conclude, our results demonstrated that Curcumol suppressed tumor growth and metastasis via an Hotair/EZH2/histone modification regulatory axis.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Histonas/metabolismo , Neoplasias Hepáticas/tratamento farmacológico , RNA Longo não Codificante/metabolismo , Sesquiterpenos/uso terapêutico , Sobrevivência Celular/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Metilação , Estrutura Molecular , RNA Longo não Codificante/genética , Sesquiterpenos/química
18.
Theranostics ; 11(14): 6891-6904, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34093860

RESUMO

Rationale: Tanshinone, a type of diterpenes derived from salvia miltiorrhiza, is a particularly promising herbal medicine compound for the treatment of cancers including acute myeloid leukemia (AML). However, the therapeutic function and the underlying mechanism of Tanshinone in AML are not clear, and the toxic effect of Tanshinone limits its clinical application. Methods: Our work utilizes human leukemia cell lines, zebrafish transgenics and xenograft models to study the cellular and molecular mechanisms of how Tanshinone affects normal and abnormal hematopoiesis. WISH, Sudan Black and O-Dianisidine Staining were used to determine the expression of hematopoietic genes on zebrafish embryos. RNA-seq analysis showed that differential expression genes and enrichment gene signature with Tan I treatment. The surface plasmon resonance (SPR) method was used with a BIAcore T200 (GE Healthcare) to measure the binding affinities of Tan I. In vitro methyltransferase assay was performed to verify Tan I inhibits the histone enzymatic activity of the PRC2 complex. ChIP-qPCR assay was used to determine the H3K27me3 level of EZH2 target genes. Results: We found that Tanshinone I (Tan I), one of the Tanshinones, can inhibit the proliferation of human leukemia cells in vitro and in the xenograft zebrafish model, as well as the normal and malignant definitive hematopoiesis in zebrafish. Mechanistic studies illustrate that Tan I regulates normal and malignant hematopoiesis through direct binding to EZH2, a well-known histone H3K27 methyltransferase, and inhibiting PRC2 enzymatic activity. Furthermore, we identified MMP9 and ABCG2 as two possible downstream genes of Tan I's effects on EZH2. Conclusions: Together, this study confirmed that Tan I is a novel EZH2 inhibitor and suggested MMP9 and ABCG2 as two potential therapeutic targets for myeloid malignant diseases.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Abietanos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Hematopoese/efeitos dos fármacos , Leucemia/tratamento farmacológico , Leucemia/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Imunoprecipitação da Cromatina , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Hematopoese/genética , Histonas/metabolismo , Humanos , Leucemia/enzimologia , Leucemia/genética , Metaloproteinase 9 da Matriz/genética , Proteínas de Neoplasias/genética , Complexo Repressor Polycomb 2/metabolismo , Ligação Proteica , RNA-Seq , Salvia miltiorrhiza/química , Ressonância de Plasmônio de Superfície , Transcriptoma/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
19.
Future Oncol ; 17(17): 2127-2140, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33709777

RESUMO

Epigenetic alterations are major drivers of follicular lymphomagenesis, and these alterations are frequently caused by mutations in or upregulation of EZH2, a histone methyltransferase responsible for PRC2-mediated gene repression. EZH2 hyperactivation increases proliferation of B cells and prevents them from exiting the germinal center, favoring lymphomagenesis. The first FDA-approved EZH2 inhibitor is tazemetostat, which is orally available and targets both mutant and wild-type forms of the protein to induce cell cycle arrest and apoptosis of lymphoma cells in preclinical models. Phase II trials have shown objective response rates of 69% for patients with lymphoma-carrying EZH2 mutations and 35% for those with wild-type EZH2 without major toxicity, leading to tazemetostat approval for this cancer by the US FDA in June 2020.


Lay abstract Follicular lymphoma (FL) is a subtype of B-cell cancer. Initial prognosis of this disease is favorable as first-line treatments provide responses lasting 10 years on average. However, most patients will experience relapse and subsequent treatments are not as efficient nor as well tolerated as the first ones. An important driver of FL is a gene called EZH2 that makes B cells proliferate, either because of mutations that increase its activity or because of a net increase in its concentration in lymphoma cells. Tazemetostat is a drug that was designed to inhibit EZH2 protein and thus lymphoma cell growth. Phase I and II studies have been completed for this drug showing a good safety profile. In Phase II, reponses were seen in 69% of patients who have the EZH2 mutations and 35% of the other patients. The US FDA has approved tazemetostat for patients with FL who have had at least two previous treatments and harbor the EZH2 mutations, or for patients with FL who have no other therapeutic options. However, the drug has not yet been approved in Europe. Randomized trials and long-term follow-up will be of interest to make sure this drug is efficient and safe enough to be given to patients in earlier lines of treatment or in combination with other active agents used to treat patients with FL.


Assuntos
Benzamidas/uso terapêutico , Compostos de Bifenilo/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Linfoma Folicular/tratamento farmacológico , Morfolinas/uso terapêutico , Mutação , Recidiva Local de Neoplasia/tratamento farmacológico , Piridonas/uso terapêutico , Terapia de Salvação , Ensaios Clínicos Fase I como Assunto , Ensaios Clínicos Fase II como Assunto , Avaliação Pré-Clínica de Medicamentos , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Linfoma Folicular/genética , Linfoma Folicular/patologia , Dose Máxima Tolerável , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Prognóstico , Taxa de Sobrevida
20.
J Ethnopharmacol ; 269: 113691, 2021 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-33321190

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Ilex cornuta Lindl. et Paxt. (Aquifoliaceae family) belongs to the Ilex genus. The leaves of this plant are used for the popular herbal tea "Ku-Ding-Cha" in China due to their health benefits for sore throat, obesity and hypertension. Our previous studies have shown that the extract of Ilex cornuta root exerts cardioprotective effects in rat models of myocardial ischaemic injury, and several new kinds of triterpenoid saponins from Ilex cornuta (TSIC) have protective effects against hydrogen peroxide (H2O2)-induced cardiomyocyte injury. AIM OF THE STUDY: The aim of this study was to clarify the underlying mechanisms by which TSIC protect against H2O2-induced cardiomyocyte injury. MATERIALS AND METHODS: An H2O2-treated H9c2 cardiomyocyte line was used as an in vitro model of oxidation-damaged cardiomyocytes to evaluate the effects of TSIC. Apoptosis was detected with CCK-8 and annexin V assays and via analysis of the levels of apoptosis-associated proteins or genes. The underlying mechanisms related to Akt signalling, Ezh2 expression and activity, and ROS were clarified by Western blotting, quantitative PCR, flow cytometry and rescue experiments. RESULTS: TSIC protected H9c2 cells from H2O2-induced apoptosis. This effect of TSIC was attributable to inhibition of Ezh2 activity, as exhibited by attenuation of H2O2-induced Akt signalling-dependent phosphorylation of Ezh2 at serine 21 (pEzh2S21) upon TSIC pretreatment. In addition, feedback pathway between Akt-dependent Ezh2 phosphorylation and ROS was involved in TSIC-mediated protection of H9c2 cells from apoptosis. CONCLUSIONS: Our findings indicate a pivotal role of the pEzh2S21 network in TSIC-mediated protection against cardiomyocyte apoptosis, potentially providing evidence of the mechanism of TSIC in the treatment and prevention of cardiovascular diseases.


Assuntos
Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Peróxido de Hidrogênio/toxicidade , Ilex , Miócitos Cardíacos/metabolismo , Saponinas/farmacologia , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Cardiotônicos/isolamento & purificação , Cardiotônicos/farmacologia , Linhagem Celular , Miócitos Cardíacos/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Ratos , Saponinas/isolamento & purificação , Triterpenos/isolamento & purificação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA