Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Anticancer Drugs ; 33(1): 30-47, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34261915

RESUMO

Atractylodes is the dry root of atractylodes macrocephala koidz and has been commonly used as a traditional Chinese medicine (TCM). Atractylenolide III, a main component of atractylodes, has displayed significant effects on anti-inflammation and anticancer. However, the effects of atractylenolide III on growth inhibition and apoptosis induction in colon cancer remain unclear. The results showed that atractylenolide III significantly inhibited the cell growth and induce cellular apoptosis in HCT-116 cells in a concentration dependence manner in vitro. Mechanistic studies further showed that atractylenolide III could regulate the Bax/Bcl-2 apoptotic signaling pathway through promoting the expression of proapoptotic related gene/proteins Bax, caspase-9 and caspase-3 but inhibiting the expression of antiapoptotic related gene/protein Bcl-2 in HCT-116 cells. Furthermore, atractylenolide III also significantly inhibited the tumor growth of HCT-116 tumor xenografts bearing in nude mice through inducing apoptosis by upregulation of the expressions of Bax, cleaved caspase-3 and p53 but downregulation of the expressions of Bcl-2 in HCT-116 tumor tissues in vivo. The studies may provide the scientific rationale for the understanding of the anticancer effect of atractylenolide III. Therefore, atractylenolide III may have the potential to be developed as a promising novel anticancer agent for the treatment of colorectal cancer clinically.


Assuntos
Neoplasias do Colo/patologia , Lactonas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Sesquiterpenos/farmacologia , Proteína X Associada a bcl-2/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Caspase 3/efeitos dos fármacos , Caspase 9/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Ratos , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Phytother Res ; 36(1): 525-542, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34847624

RESUMO

Alnustone, a diarylheptane compound, exhibits potent growth inhibition against hepatocellular carcinoma (HCC) BEL-7402 cells. However, the underlying mechanisms associated with its anticancer activity remain unknown. In the present study, we evaluated the anticancer effect of alnustone against several human cancers focused on HCC and the possible associated mechanisms. The results showed that alnustone significantly inhibited the growth of several cancer cells by CCK-8 assay. Alnustone markedly induced apoptosis and decreased mitochondrial membrane potential in BEL-7402 and HepG2 cells. Alnustone inhibited the expression of proteins related to apoptosis and PI3K/Akt/mTOR/p70S6K pathways and generated ROS production in BEL-7402 and HepG2 cells. Moreover, N-acetyl-L-cysteine (NAC, a ROS inhibitor) could significantly reverse the effects of alnustone on the growth inhibition of BEL-7402 and HepG2 cells and the expression of proteins related to apoptosis and PI3K/Akt/mTOR signaling pathway in HepG2 cells. Furthermore, alnustone significantly inhibited tumor growth of HepG2 xenografts, obviously induced apoptosis in the tumor tissues and improved the pathological condition of liver tissues of mice in vivo. The study provides evidence that alnustone is effective against HCC via ROS-mediated PI3K/Akt/mTOR/p70S6K pathway and the compound has the potential to be developed as a novel anticancer agent for the treatment of HCC clinically.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Espécies Reativas de Oxigênio , Proteínas Quinases S6 Ribossômicas 70-kDa , Serina-Treonina Quinases TOR
3.
J Proteome Res ; 20(1): 950-959, 2021 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33105993

RESUMO

Drug addiction is a chronic relapsing brain disease. Alterations of glucose uptake and metabolism are found in the brain of drug addicts. Insulin mediates brain glucose metabolism and its abnormality could induce brain injury and cognitive impairment. Here, we established a rat model of phenobarbital addiction by 90 days of dose escalation and evaluated addiction-related symptoms. We also performed 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) to detect glucose uptake in the brain and proteomic analysis of the function of the differentially expressed (DE) proteins via bioinformatics in brain tissues by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) on days 60 and 90 of phenobarbital or 0.5% carboxymethyl cellulose sodium (CMC-Na) (vehicle) administration. The results showed that phenobarbital-addictive rats developed severe withdrawal symptoms after abstinence and glucose uptake was significantly increased in the brain. Proteomics analysis showed that numerous DE proteins were enriched after phenobarbital administration, among which CALM1, ARAF, and Cbl proteins (related to the insulin signaling pathway) were significantly downregulated on day 60 but not day 90. However, SLC27A3 and NF-κB1 proteins (related to insulin resistance) were significantly upregulated on day 90 (data are available via ProteomeXchange with identifier PXD021101). Our data indicate that the insulin signaling pathway and insulin resistance may play a role in the development of phenobarbital addiction and brain injury, so the findings may have important clinical implications.


Assuntos
Fluordesoxiglucose F18 , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Animais , Encéfalo/diagnóstico por imagem , Cromatografia Líquida , Glucose , Insulina , Fenobarbital/toxicidade , Proteômica , Ratos , Transdução de Sinais , Espectrometria de Massas em Tandem
4.
Neoplasma ; 68(6): 1181-1189, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34585588

RESUMO

Mere15, an anticancer polypeptide with a molecular weight of 15 kDa, is extracted from the marine species Meretrix meretrix. A previous study in our laboratory has confirmed that Mere15 displays a potent antitumor activity. However, the underlying mechanism of Mere15 still remains unclear. The effect of Mere15 on the growth of a variety of tumor cells was measured by the CCK-8 assay. Hoechst33342/PI double staining and flow cytometry assays were used to detect the apoptosis status of cancer cells. Western blotting was used to detect the expression of apoptosis-related proteins, migration and invasion-related protein, and the changes in the PI3K/Akt/mTOR signaling pathway-related proteins. Treatment with Mere15 inhibited cancer cell growth significantly. Scratch wound-healing assay, as well as Transwell experiments, revealed that the polypeptide was able to inhibit the invasion and migration of NSCLC cells significantly. Western blotting analysis confirmed that treatment with Mere15 inhibited the phosphorylation of PI3K, Akt, and mTOR significantly. The effects of Mere15 were also evaluated in the presence of an activator or inhibitor of the PI3K/Akt/mTOR pathway. Downregulated expression of MMP-2, MMP-9, and Snail, and increased expression of E-cadherin were also found in cells treated with Mere15. In vivo study revealed that Mere15 inhibited tumor growth significantly in xenograft nude mice bearing NCI-H460 cancer cells. The study provides evidence that Mere15 has the potential to be developed as a novel antimetastatic agent for the treatment of NSCLC patients. The work also provides further evidence that targeting PI3K/Akt/mTOR pathway is an important strategy for overcoming cancer metastasis.


Assuntos
Bivalves/química , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Peptídeos/farmacologia , Animais , Apoptose , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
5.
J Cell Biochem ; 120(1): 56-70, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30246452

RESUMO

Endotoxin tolerance is defined as a reduced capacity of a cell to respond endotoxin (lipopolysaccharide, LPS) challenge after an initial encounter with endotoxin in advance. The body becomes tolerant to subsequent challenge with a lethal dose of endotoxin and cytokines release and cell/tissue damage induced by inflammatory reaction are significantly reduced in the state of endotoxin tolerance. The main characteristics of endotoxin tolerance are downregulation of inflammatory mediators such as tumor necrosis factor α (TNF-α), interleukin-1ß (IL-1ß), and C-X-C motif chemokine 10 (CXCL10) and upregulation of anti-inflammatory cytokines such as IL-10 and transforming growth factor ß (TGF-ß). Therefore, endotoxin tolerance is often regarded as the regulatory mechanism of the host against excessive inflammation. Endotoxin tolerance is a complex pathophysiological process and involved in multiple cellular signal pathways, receptor alterations, and biological molecules. However, the exact mechanism remains elusive up to date. To better understand the underlying cellular and molecular mechanisms of endotoxin tolerance, it is crucial to investigate the comprehensive cellular signal pathways, signaling proteins, cell surface molecules, proinflammatory and anti-inflammatory cytokines, and other mediators. Endotoxin tolerance plays an important role in reducing the mortality of sepsis, endotoxin shock, and other endotoxin-related diseases. Recent reports indicated that endotoxin tolerance is also related to other diseases such as cystic fibrosis, acute coronary syndrome, liver ischemia-reperfusion injury, and cancer. The aim of this review is to discuss the recent advances in endotoxin tolerance mainly based on the cellular and molecular mechanisms by outline the current state of the knowledge of the involvement of the toll-like receptor 4 (TLR4) signaling pathways, negative regulate factor, microRNAs, apoptosis, chromatin modification, and gene reprogramming of immune cells in endotoxin tolerance. We hope to provide a new idea and scientific basis for the rational treatment of endotoxin-related diseases such as endotoxemia, sepsis, and endotoxin shock clinically.


Assuntos
Apoptose/imunologia , Lipopolissacarídeos , Choque Séptico , Transdução de Sinais , Animais , Humanos , Inflamação/imunologia , Inflamação/patologia , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/toxicidade , Choque Séptico/imunologia , Choque Séptico/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
6.
BMC Cancer ; 16: 114, 2016 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-26884234

RESUMO

BACKGROUND: Brainstem dose limitations influence radiation dose reaching to tumor in the patients with locally-advanced nasopharyngeal cancer (NPC). METHODS: A retrospective analysis of the prognostic value of the distance between the primary tumor and brainstem (Dbs) in 358 patients with locally-advanced NPC after intensity-modulated radiation therapy (IMRT). Receiver operating characteristic (ROC) curves were used to identify the cut-off value to analyze the impact of Dbs on tumor dose coverage and prognosis. RESULTS: The three-year overall survival (OS), local relapse-free survival (LRFS), distant metastasis-free survival (DMFS), and disease-free survival (DFS) were 88.8 vs. 78.4% (P = 0.007), 96.5 vs. 91.1% (P = 0.018), 87.8 vs. 79.3% (P = 0.067), and 84.1 vs. 69.6% (P = 0.002) for the patients with the Dbs > 4.7 vs. ≤ 4.7 mm, respectively. ROC curves revealed Dbs (4.7 mm) combined with American Joint Committee on Cancer (AJCC) T classification had a significantly better prognostic value for OS (P < 0.05). CONCLUSIONS: Dbs (≤ 4.7 mm) is an independent negative prognostic factor for OS/LRFS/DFS and enhances the prognostic value of T classification in the patients with locally-advanced NPC.


Assuntos
Tronco Encefálico/patologia , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/patologia , Carcinoma , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/epidemiologia , Neoplasias Nasofaríngeas/radioterapia , Valor Preditivo dos Testes , Prognóstico , Curva ROC , Radioterapia de Intensidade Modulada , Estudos Retrospectivos , Risco
7.
Molecules ; 21(3): 260, 2016 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-26927040

RESUMO

Eucommia ulmoides Oliver (E. ulmoides) is a traditional Chinese medicine with many beneficial effects, used as a tonic medicine in China and other countries. Chlorogenic acid (CGA) is an important compound in E. ulmoides with neuroprotective, cognition improvement and other pharmacological effects. However, it is unknown whether chlorogenic acid-enriched Eucommia ulmoides Oliver bark has antidepressant potential through neuron protection, serotonin release promotion and penetration of blood-cerebrospinal fluid barrier. In the present study, we demonstrated that CGA could stimulate axon and dendrite growth and promote serotonin release through enhancing synapsin I expression in the cells of fetal rat raphe neurons in vitro. More importantly, CGA-enriched extract of E. ulmoides (EUWE) at 200 and 400 mg/kg/day orally administered for 7 days showed antidepressant-like effects in the tail suspension test of KM mice. Furthermore, we also found CGA could be detected in the the cerebrospinal fluid of the rats orally treated with EUWE and reach the level of pharmacological effect for neuroprotection by UHPLC-ESI-MS/MS. The findings indicate CGA is able to cross the blood-cerebrospinal fluid barrier to exhibit its neuron protection and promotion of serotonin release through enhancing synapsin I expression. This is the first report of the effect of CGA on promoting 5-HT release through enhancing synapsin I expression and CGA-enriched EUWE has antidepressant-like effect in vivo. EUWE may be developed as the natural drugs for the treatment of depression.


Assuntos
Antidepressivos/farmacologia , Ácido Clorogênico/farmacologia , Depressão/tratamento farmacológico , Eucommiaceae/química , Nootrópicos/farmacologia , Núcleos da Rafe/efeitos dos fármacos , Sinapsinas/genética , Administração Oral , Animais , Antidepressivos/isolamento & purificação , Barreira Hematoencefálica/metabolismo , Ácido Clorogênico/isolamento & purificação , Depressão/genética , Depressão/metabolismo , Depressão/fisiopatologia , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas , Feto , Expressão Gênica , Elevação dos Membros Posteriores , Masculino , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Nootrópicos/isolamento & purificação , Permeabilidade , Casca de Planta/química , Extratos Vegetais/química , Núcleos da Rafe/metabolismo , Núcleos da Rafe/fisiopatologia , Ratos , Serotonina/biossíntese , Serotonina/metabolismo , Sinapsinas/agonistas , Sinapsinas/metabolismo
8.
Tumour Biol ; 36(12): 9667-76, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26150336

RESUMO

Glioblastoma stem cells (GSCs) are the initiating cells in glioblastoma multiforme (GBM) and contribute to the resistance of GBM to chemotherapy and radiation. In the present study, we investigated the effects of cardamonin (3,4,2,4-tetrahydroxychalcone) on the self-renewal and apoptosis of GSCs, and if its action is associated with signal transducer and activator of transcription 3 (STAT3) pathway. CD133(+) GSCs, a kind of GSCs line, was established from human glioblastoma tissues. Cardamonin inhibited the proliferation and induced apoptosis in CD133+ GSCs. The proapoptotic effects of temozolomide (TMZ) were further enhanced by cardamonin in CD133+ GSCs and U87 cells in vitro. For in vivo study, injection of 5 × 10(5) cells of CD133+ GSCs subcutaneously (s.c.) into nude mice, 100 % of large tumors were developed within 8 weeks in all mice; in contrast, only one out of five mice developed a small tumor when 5 × 10(5) cells of CD133(-) GMBs cells were injected. Cardamonin also inhibited STAT3 activation by luciferase assay and suppressed the expression of the downstream genes of STAT3, such as Bcl-XL, Bcl-2, Mcl-1, survivin, and VEGF. Furthermore, cardamonin locked nuclear translocation and dimerization of STAT3 in CD133(+) GSCs. Docking analysis confirmed that cardamonin molecule was successfully docked into the active sites of STAT3 with a highly favorable binding energy of -10.78 kcal/mol. The study provides evidence that cardamonin is a novel inhibitor of STAT3 and has the potential to be developed as a new anticancer agent targeting GSCs. This study also reveals that targeting STAT3 signal pathway is an important strategy for the treatment of human GBM.


Assuntos
Chalconas/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética , Glioblastoma/tratamento farmacológico , Fator de Transcrição STAT3/biossíntese , Animais , Apoptose/efeitos dos fármacos , Domínio Catalítico , Linhagem Celular Tumoral , Chalconas/química , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/biossíntese , Células-Tronco Neoplásicas/química , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Conformação Proteica/efeitos dos fármacos , Fator de Transcrição STAT3/química , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Mol Pharm ; 11(2): 457-67, 2014 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-24329001

RESUMO

We recently reported the identification and characterization of a novel small chemical molecule designated FL118. FL118 selectively inhibits multiple cancer survival and proliferation-associated antiapoptotic proteins (survivin, Mcl-1, XIAP, cIAP2) and eliminates small and large human tumor xenografts in animal models (Ling et al., PLoS One 2012, 7, e45571). Here, we report a follow-up study on the structure-activity relationship (SAR) of the hydroxyl group in the lactone ring of FL118. We found that the superior antitumor efficacy of FL118 heavily depends on its steric configuration through comparing the antitumor activity of FL118 with FL113 (the racemic mixture of FL118). Consistently, FL118 proved much more effective in inhibiting the expression of survivin, Mcl-1, and cIAP2, both in vitro and in vivo, compared to FL113. Additionally, Tet-on controlled induction of survivin or forced expression of Mcl-1 protects cancer cells from FL118-mediated growth inhibition and cell death. To further explore the SAR, we synthesized seven position 20-esterifiable FL118 and FL113 derivatives. Studies on these seven new compounds revealed that keeping a free hydroxyl group of FL118 is also important for high antitumor efficacy. Together, these studies confirm the superior anticancer activity of FL118 and narrow the window for further SAR studies to generate novel analogues based on FL118 core structure on its other potential chemical positions.


Assuntos
Antineoplásicos/farmacologia , Benzodioxóis/química , Benzodioxóis/farmacologia , Indolizinas/química , Indolizinas/farmacologia , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Humanos , Estrutura Molecular , Ligação Proteica/efeitos dos fármacos , Relação Estrutura-Atividade
10.
J Ethnopharmacol ; 301: 115805, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36216195

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Shenkang injection (SKI), a Chinese patent medicine injection, has been approved for the treatment of chronic kidney disease (CKD) due to its definite clinical therapeutic efficacy. However, the effect and associated underlying mechanism of Shenkang injection against cisplatin (CDDP)-induced acute kidney injury (AKI) has not yet been well elucidated. AIM OF THE STUDY: This study aims to investigate the therapeutic effect and associated underlying mechanism of Shenkang injection against CDDP-induced AKI. MATERIALS AND METHODS: We established a CDDP-induced AKI mouse model to evaluate renal function by biochemical markers measurement and to observe histopathological alterations by haemotoxylin and eosin (HE)-staining sections of renal. In addition, the distribution of representative components of SKI in the kidneys of mice was evaluated by liquid chromatography tandem mass spectrometry (LC-MS/MS). Furthermore, the degree of oxidative stress and inflammation were assessed by detecting the levels of inflammatory cytokines and oxidants, while the related mechanisms were elucidated by network pharmacology. RESULTS: CDDP could induce excessive inflammation and severe injury to the kidneys of mice. However, SKI significantly ameliorated the kidney damages and improved the renal function by reducing the levels of renal function markers (SCr, BUN and urine protein), and inhibiting the production of inflammatory cytokines IL-34, IL-6 and TNF-α. SKI repaired oxidative balance through up-regulation of antioxidants SOD and GSH and down-regulated oxidants MDA. Moreover, 4 components from SKI were detected in the kidney by LC-MS/MS quantification. In addition, pharmacology network indicated the PI3K/AKT, TNF, MAPK, and p53 were the possible signaling pathways for the therapeutic effect of SKI against CDDP-induced AKI, which were related to inflammation, oxidative stress and apoptosis. CONCLUSION: In the present study, we for the first time demonstrated that SKI alleviates CDDP-induced nephrotoxicity by antioxidant and anti-inflammation via regulating PI3K/AKT, MAPK, TNF, and p53 signaling pathways. The study may provide a scientific rationale for the clinical indication of SKI.


Assuntos
Injúria Renal Aguda , Cisplatino , Camundongos , Animais , Cisplatino/toxicidade , Cromatografia Líquida , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Espectrometria de Massas em Tandem , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/metabolismo , Rim , Estresse Oxidativo , Apoptose , Inflamação/patologia , Antioxidantes/farmacologia , Oxidantes/metabolismo , Citocinas/metabolismo
11.
BMC Cancer ; 12: 293, 2012 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-22804960

RESUMO

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) accounts for more than 80% of the cases of renal cell carcinoma. In ccRCC deactivation of Von-Hippel-Lindau (VHL) gene contributes to the constitutive expression of hypoxia inducible factors 1 and 2 alpha (HIF-α), transcriptional regulators of several genes involved in tumor angiogenesis, glycolysis and drug resistance. We have demonstrated inhibition of HIF-1α by Se-Methylselenocysteine (MSC) via stabilization of prolyl hydroxylases 2 and 3 (PHDs) and a significant therapeutic synergy when combined with chemotherapy. This study was initiated to investigate the expression of PHDs, HIF-α, and VEGF-A in selected solid cancers, the mechanism of HIF-α inhibition by MSC, and to document antitumor activity of MSC against human ccRCC xenografts. METHODS: Tissue microarrays of primary human cancer specimens (ccRCC, head & neck and colon) were utilized to determine the incidence of PHD2/3, HIF-α, and VEGF-A by immunohistochemical methods. To investigate the mechanism(s) of HIF-α inhibition by MSC, VHL mutated ccRCC cells RC2 (HIF-1α positive), 786-0 (HIF-2α positive) and VHL wild type head & neck cancer cells FaDu (HIF-1α) were utilized. PHD2 and VHL gene specific siRNA knockdown and inhibitors of PHD2 and proteasome were used to determine their role in the degradation of HIF-1α by MSC. RESULTS: We have demonstrated that ccRCC cells express low incidence of PHD2 (32%), undetectable PHD3, high incidence of HIF-α (92%), and low incidence of VEGF-A compared to head & neck and colon cancers. This laboratory was the first to identify MSC as a highly effective inhibitor of constitutively expressed HIF-α in ccRCC tumors. MSC did not inhibit HIF-1α protein synthesis, but facilitated its degradation. The use of gene knockdown and specific inhibitors confirmed that the inhibition of HIF-1α was PHD2 and proteasome dependent and VHL independent. The effects of MSC treatment on HIF-α were associated with significant antitumor activity against ccRCC xenograft. CONCLUSIONS: Our results show the role of PHD2/3 in stable expression of HIF-α in human ccRCC. Furthermore, HIF-1α degradation by MSC is achieved through PHD2 dependent and VHL independent pathway which is unique for HIF-α regulation. These data provide the basis for combining MSC with currently used agents for ccRCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Renais/metabolismo , Compostos Organosselênicos/farmacologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo , Animais , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dioxigenases/genética , Dioxigenases/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia , Neoplasias Renais/genética , Camundongos , Camundongos Nus , Pró-Colágeno-Prolina Dioxigenase/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteólise/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Eur J Nutr ; 51(1): 107-17, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21516492

RESUMO

BACKGROUND: Colorectal cancer (CRC) is one of the most common and preventable cancers. Regular consumption of apples is conducive to reduction in CRC risk. AIM OF THE STUDY: To evaluate effects of modified apple polysaccharide (MAP) on tumorigenesis in a mouse model of colitis-associated colon cancer. METHODS: One hundred male ICR mice were administered with 1, 2-dimethyl-hydrazine (DMH) and dextran sodium sulfate (DSS). Forty mice were given no further treatment, the rest were fed basal diet blended with three different doses of MAP; 2.5, 5, and 10% (20 mice in each group). RESULTS: MAP significantly protected ICR mice against DMH/DSS-induced tumorigenesis. The incidence of tumor development was 90% (18/20) in the mice treated with DMH/DSS, but that was reduced to 25% (5/20), 15% (3/20), and 5% (1/20), respectively, in the mice treated with basal diets plus 2.5, 5, and 10% of MAP. Study of apoptosis of colonic epithelial cells revealed that MAP moderately increased apoptosis, suggesting that the anti-tumor potency of MAP was probably attributed to its ability to induce apoptosis. Western blot analysis demonstrated that carbohydrate-binding protein galectin-3 changed in both the nucleus and the cytoplasm during the process from colitis to colon cancer in the model. And MAP could inhibit the binding of galectin-3 to its ligand: this is, at least in part, the possible mechanism of MAP by enhancing apoptosis and preventing tumorigenesis. CONCLUSIONS: These data suggest that MAP has a potential role in clinical prevention and treatment for colon cancer.


Assuntos
Anticarcinógenos/uso terapêutico , Apoptose , Colite/fisiopatologia , Neoplasias do Colo/prevenção & controle , Galectina 3/metabolismo , Malus/química , Polissacarídeos/uso terapêutico , Animais , Anticarcinógenos/química , Anticarcinógenos/isolamento & purificação , Anticarcinógenos/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Transformação Celular Neoplásica , Neoplasias do Colo/etiologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Citoplasma/metabolismo , Suplementos Nutricionais/análise , Modelos Animais de Doenças , Enterócitos/imunologia , Enterócitos/metabolismo , Enterócitos/patologia , Frutas/química , Galectina 3/sangue , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Proteínas de Neoplasias/metabolismo , Polissacarídeos/química , Polissacarídeos/isolamento & purificação , Polissacarídeos/metabolismo , Distribuição Aleatória
13.
Oncol Rep ; 48(2)2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35730618

RESUMO

Alpinia katsumadai Hayata (AKH), a widely used traditional Chinese medicine, exerts various biological functions, including anti­inflammatory, antioxidant, anti­microbial and anti­asthmatic effects. However, studies on its anticancer activity and associated mechanisms are limited. The present study investigated the effects of ethanol extract from AKH on the viability of various human cancer and normal liver LX­2 cells using Cell Counting Kit­8 assay. Apoptosis was detected by Hoechst 33342/PI staining and Annexin­V­FITC/PI double staining. Autophagy was examined by Ad­GFP­LC3B transfection. The association between AKH­induced autophagy and apoptosis was investigated by pre­treatment of the cells with the autophagy inhibitors, 3­methyladenine (3MA) and bafilomycin A1 (Baf­A1), followed by treatment with AKH. The expression levels of cleaved poly(ADP­ribose) polymerase (PARP), caspase­8, caspase­3, caspase­9, phosphorylated (p­)AMP­activated protein kinase (AMPK), Akt, mTOR and p70S6K were examined using western blot analysis. The in vivo antitumor activity of AKH was investigated in nude mice bearing A549 lung cancer xenografts. The components of AKH were detected by liquid chromatography mass spectrometry­ion trap­time­of­flight mass spectrometry. The results revealed that AKH significantly inhibited the proliferation of various cancer cells with the half maximal inhibitory concentration (IC50) values of 203­284 µg/ml; however, its inhibitory effect was much less prominent against normal liver LX­2 cells with an IC50 value of 395 µg/ml. AKH markedly induced apoptosis and autophagy, and upregulated the protein expression of cleaved­caspase­3, caspase­8, caspase­9 and cleaved PARP in a concentration­dependent manner. Of note, the autophagy inhibitors (3MA and Baf­A1) significantly attenuated its pro­apoptotic effects on human pancreatic cancer Panc­28 and lung cancer A549 cells. Furthermore, AKH significantly increased the levels of p­AMPK, and decreased those of p­Akt, p­mTOR and p­p70S6K in Panc­28 and A549 cells. AKH markedly inhibited the growth of A549 tumor xenografts in vivo. In addition, a total of nine compounds were detected from AKH. The present study demonstrates that AKH markedly inhibits the growth and induces autophagy­related apoptosis in cancer cells by regulating the AMPK and Akt/mTOR/p70S6K signaling pathways. AKH and/or its active fractions may thus have potential to be developed as novel anticancer agents for clinical use.


Assuntos
Alpinia , Neoplasias Pulmonares , Proteínas Quinases Ativadas por AMP/metabolismo , Alpinia/metabolismo , Animais , Apoptose , Autofagia , Caspase 3/metabolismo , Caspase 8/metabolismo , Caspase 9/metabolismo , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
14.
Biochim Biophys Acta Gen Subj ; 1865(1): 129759, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33038451

RESUMO

Protein kinases play a fundamental role in the intracellular transduction because of their ability to phosphorylate plethora of proteins. Over the past three decades, numerous protein kinase inhibitors have been identified and are being used clinically successfully. The biodiversity of marine organisms provides a rich source for the discovery and development of novel anticancer agents in the treatment of human malignancies and a lot of bioactive ingredients from marine organisms display anticancer effects by affecting the protein kinases-mediated pathways. In the present mini-review, anticancer compounds from marine source were reviewed and discussed in context of their targeted pathways associated with protein kinases and the progress of these compounds as anticancer agents in recent five years were emphasized. The molecular entities and their modes of actions were presented. We focused on protein kinases-mediated signaling pathways including PI3K/Akt/mTOR, p38 MAPK, and EGFR. The marine compounds targeting special pathways of protein kinases were highlighted. We have also discussed the existing challenges and prospects related to design and development of novel protein kinase inhibitors from marine sources.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Animais , Antineoplásicos/uso terapêutico , Organismos Aquáticos , Produtos Biológicos/uso terapêutico , Descoberta de Drogas , Receptores ErbB/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Front Biosci (Landmark Ed) ; 26(11): 1349-1361, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34856772

RESUMO

Backgroud: Protein kinases play an important role in cell proliferation, differentiation, mobility and cell cycle arrest etc. These enzymes act as important targets in developing anticancer agents. Over the years, a large number of protein kinase inhibitors have been discovered and developed as anticancer agents for the treatment of cancers clinically. However, the drug-resiatance and off-targeting limit their effeciancy for the treatment of human cancer. Materials and methods: Alkaloids are an important class of natural products with broad spectrum biological activities. In the past decades, numerus alkaloids with significant anticancer activity by inhibiting protein kinases were identified. In the present mini-review, we will present the key enzymes including mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and janus-activated kinases/signal transducer and activator of transcription (JAK/STAT) targeted by alkaloids and highlight the special sites targeted by alkaloids on protein kinases and/or reversing drug resistance. Additionally, the challenge and prospect of developing alkaloids as new anticancer agents are also discussed. Conclusion: Alkaloids suppressed tumor growth through targeting different signaling pathways mediated by protein kinases of cancer cells. It is conceivable that novel alkaloids anticancer agents with promising clinical value will be developed in the future.


Assuntos
Alcaloides , Antineoplásicos , Alcaloides/farmacologia , Antineoplásicos/farmacologia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
16.
Mol Med Rep ; 23(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33576443

RESUMO

Human umbilical vein endothelial cells (HUVECs) serve a critical role in maintaining normal vascular function. Lipopolysaccharide (LPS), which is released from pathogenic bacteria in the blood, induces HUVEC apoptosis and injury to cause vascular dysfunction and infectious vascular diseases. Procyanidin B2 (PB2) possesses numerous functions, including antioxidant, antitumor, anti­inflammatory and antiapoptosis effects, but the molecular mechanism is not completely understood. The present study investigated the effects of PB2 on LPS­induced cytotoxicity and apoptosis in HUVECs, as well as the underlying mechanisms. The effects of PB2 on LPS­mediated alterations to cytotoxicity, mitochondrial membrane potential, apoptosis were assessed by performing Cell Counting Kit­8, JC­1 fluorescence, Hoechst 33258 staining assays, respectively. IL­1ß, IL­6 and TNF­α mRNA expression and protein levels were measured by performing reverse transcription­quantitative PCR and ELISAs, respectively. Bcl­2, Bax, cleaved caspase­3, cleaved caspase­7, cleaved caspase­9, phosphorylated (p)­IκB­α, p­IκB­ß, p­NF­κB­p65 and total NF­κB p65 protein expression levels were determined via western blotting. NF­κB p65 nuclear translocation was assessed via immunofluorescence. PB2 pretreatment markedly attenuated LPS­induced cytotoxicity and apoptosis in HUVECs. PB2 also significantly downregulated the expression levels of IL­1ß, IL­6, TNF­α, Bax, cleaved caspase­3, cleaved caspase­7, cleaved caspase­9 and p­NF­κB­p65, but upregulated the expression levels of Bcl­2, p­IκB­α and p­IκB­ß in LPS­induced HUVECs. Moreover, PB2 markedly inhibited LPS­induced NF­κB p65 nuclear translocation in HUVECs. The results suggested that the potential molecular mechanism underlying PB2 was associated with the Bax/Bcl­2 and NF­κB signalling pathways. Therefore, PB2 may serve as a useful therapeutic for infectious vascular diseases.


Assuntos
Apoptose/efeitos dos fármacos , Biflavonoides/farmacologia , Catequina/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , NF-kappa B/metabolismo , Proantocianidinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Expressão Gênica/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Modelos Biológicos , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
17.
Chemotherapy ; 56(3): 223-33, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20551639

RESUMO

BACKGROUND: The purpose of this study was: (1) to document the critical requirement of cystine for growth of human tumor cells in vitro, and (2) to determine the effect of the anticancer agent irinotecan on the cystine transporter x(c)(-) in head and neck FaDu xenografts. METHODS: Cell growth was measured by sulforhodamine B assay. xCT protein, glutathione (GSH) and DNA damage were determined using Western blot, spectrophotometry, and immunohistochemistry, respectively. RESULTS: Depletion of cystine from the medium inhibited tumor cell growth. Treatment of FaDu tumor with a therapeutic dose of irinotecan resulted in depression of xCT protein levels, leading to tumor growth retardation and downregulation of GSH with increased reactive oxygen species (ROS). The accumulation of ROS correlated with increased DNA damage as evidenced by increased H2AX. CONCLUSION: Depression of xCT protein by irinotecan resulted in downregulation of GSH and increase in ROS, which could be the other possible mechanisms of DNA damage by irinotecan.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Camptotecina/análogos & derivados , Cistina/fisiologia , Regulação para Baixo/fisiologia , Neoplasias de Cabeça e Pescoço/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto , Sistemas de Transporte de Aminoácidos/antagonistas & inibidores , Sistemas de Transporte de Aminoácidos/biossíntese , Animais , Camptotecina/farmacologia , Linhagem Celular Tumoral , Cistina/antagonistas & inibidores , Cistina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Irinotecano , Camundongos , Camundongos Nus , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
18.
Drug Des Devel Ther ; 14: 3495-3507, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32921986

RESUMO

PURPOSE: MiR-34a, which acts as an important tumor suppressor gene, plays an important role in pancreatic cancer. However, the therapeutic application of miR-34a is limited by the lack of an effective delivery system. In the present study, we synthesize exosomes-coated miR-34a (exomiR-34a), and the anticancer effect of exomiR-34a was evaluated in pancreatic cancer. MATERIALS AND METHODS: An ultrasound approach was used to synthesize exomiR-34a, and its transfection efficiency was examined by confocal microscopy and flow cytometry. The level of miR-34a and its targeted gene Bcl-2 was detected by real-time quantitative PCR (qRT-PCR). MTT analysis was performed to determine the effect of exomiR-34a on the growth of pancreatic cancer cells. Annexin-V/PI double staining and Western blot analysis were carried out to determine the apoptosis of the pancreatic cancer cells. The xenograft nude mice model bearing human pancreatic cancer Panc28 cells was used to determine the antitumor effect of exomiR-34a in vivo. RESULTS: The exomiR-34a could cross the cell membrane efficiently, and downregulated the expression of the targeted gene Bcl-2. Treatment with exomiR-34a inhibited the growth of the pancreatic cancer cells significantly and the nanoparticles also induced apoptosis in cancer cells via affecting the expression of apoptotic-related genes. In vivo study using xenograft nude mice bearing Panc28 cancer cells revealed that exomiR-34a suppressed the growth of tumors significantly. CONCLUSION: ExomiR-34a can inhibit the growth of pancreatic cancer both in vitro and in vivo. Targeting miR-34a is a promising strategy for the treatment of pancreatic cancer. ExomiR-34a has the potential to be developed as a novel anticancer agent for the treatment of human pancreatic malignancy.


Assuntos
Exossomos/metabolismo , MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , Antineoplásicos , Apoptose , Proliferação de Células , Células Cultivadas , Exossomos/química , Humanos , MicroRNAs/síntese química , MicroRNAs/química , Neoplasias Pancreáticas/patologia
19.
Biomed Pharmacother ; 126: 110073, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32179201

RESUMO

PURPOSE: Myocardial fibrosis (MF) is an unavoidable complication in patients with hypertensive heart disease. Valsartan, a widely used antihypertensive drug, was reported to inhibit MF. Deficiency in the 5-hydroxytryptamine (5-HT, serotonin) transporter gene has been proven to cause MF. Long-term sympathetic nerve excitability activates renin angiotensin aldosterone system leading to MF. Tandospirone, a partial agonist of the 5-HT1A receptor, has been commonly used to relieve psychiatric symptoms. However, there is limited evidence on the combination of valsartan and tandospirone for the treatment of MF. Therefore, we investigated the synergistic effect of tandospirone on the anti-MF activity of valsartan in spontaneously hypertensive rats (SHRs). METHODS: Systolic blood pressure (SBP) of SHRs (12-week-old) was measured weekly using the tail-cuff method for eight weeks; the left ventricular was collected and weighted for calculation of the left ventricular mass index (LVMI). The myocardial histopathology of left ventricle was evaluated in rats by hematoxylin and eosin (H&E) and Mason's trichrome staining assays. The mRNA and protein expressions of transforming growth factor ß (TGF-ß1), Sma- and Mad-related protein 3 (Smad3), and fibronectin (Fn) were investigated by real time PCR, immunohistochemistry, and Western blotting analysis, respectively. RESULTS: Tandospirone (40 mg/kg) could significantly improve the effect of valsartan (30 mg/kg) in decreasing the SBP of SHRs and lower the ratio of the LVMI in SHRs, compared to that of rats treated with valsartan or tandospirone alone. Tandospirone could also enhance the valsartan-induced reduction in collagen deposition in the myocardial tissues of SHRs. Furthermore, tandospirone could enhance the effect of valsartan on downregulating the expression levels of TGF-ß1, Smad3, and Fn at both mRNA and protein levels. CONCLUSION: We report for the first time that tandospirone could improve the anti-MF efficacy of valsartan via the TGF-ß1/Smad3 signaling pathway in SHRs. Our findings may provide valuable insight into the scientific rationale for combining tandospirone and valsartan in the treatment of MF clinically.


Assuntos
Anti-Hipertensivos/farmacologia , Isoindóis/farmacologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Agonistas do Receptor de Serotonina/farmacologia , Valsartana/farmacologia , Animais , Biomarcadores , Pressão Sanguínea , Cardiomiopatias/tratamento farmacológico , Cardiomiopatias/etiologia , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Sinergismo Farmacológico , Fibrose , Expressão Gênica , Hipertensão/complicações , Imuno-Histoquímica , Masculino , Modelos Biológicos , Miocárdio/metabolismo , Ratos , Ratos Endogâmicos SHR , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo
20.
Biochem Pharmacol ; 171: 113716, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751535

RESUMO

Melanoma is one of the most aggressive malignancies. Drug resistance and toxicity limits the clinical efficacy of melanoma chemotherapeutic drugs such as dacarbazine. Therefore, the development of chemotherapeutic agents for melanoma treatment is urgently needed. RJT-101 significantly inhibits the proliferation of melanoma cells, however has low cytotoxicity to non-malignant cells. RJT-101 induces apoptosis, DNA damage, and G2/M phase arrest, as a consequent, attenuates tumor growth, lung metastasis in vivo as well as prolongs survival of tumor bearing mice. RJT-101 could block topoisomerase I (Top1) activity as well as induce its degradation through proteasome system. Interestingly, Top1 is over-expressed in melanoma cells, compared to non-malignant cells. Knock down of Top1 suppresses melanoma cells growth and induces apoptosis and DNA damage in melanoma cells. RJT-101 effectively inhibits melanoma cells (including vemurafenib-resistant melanoma cells) proliferation in vitro and in vivo through the induction of DNA damage and apoptosis by inhibiting of Top1, indicating RJT-101 warrants further clinical evaluation.


Assuntos
Antineoplásicos/farmacologia , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Dano ao DNA/efeitos dos fármacos , Melanoma/tratamento farmacológico , Neoplasias Cutâneas/tratamento farmacológico , Inibidores da Topoisomerase I/farmacologia , Animais , Antineoplásicos/química , Camptotecina/química , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA Topoisomerases Tipo I/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Humanos , Melanoma/genética , Melanoma/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Nus , Estrutura Molecular , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Inibidores da Topoisomerase I/química , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA