Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Invest Ophthalmol Vis Sci ; 65(1): 34, 2024 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-38236186

RESUMO

Purpose: The purpose of this study was to elucidate the involvement of potassium two pore domain channel subfamily K member 5 (KCNK5)-mediated potassium efflux in the pathogenesis of dry eye and to unravel the underlying molecular mechanisms. Methods: To induce experimental dry eye in adult wild-type C57BL/6 mice, scopolamine was administered via subcutaneous injection, and the mice were subjected to desiccating stress. To create an in vitro model of dry eye, desiccation stress was applied to the human corneal epithelial cell line (HCE-T). Intracellular potassium concentration was quantified using inductively coupled plasma mass spectrometry. Cellular death was assessed through lactate dehydrogenase assays. Gene expression profiling was conducted through both RNA sequencing and quantitative real-time PCR. Protein analysis was carried out through Western blotting and immunofluorescence staining. Assessment of the corneal epithelial defect area was conducted through fluorescein sodium staining. Tear secretion was quantified using the phenol red cotton thread method. Results: Potassium efflux was observed to further facilitate corneal epithelial pyroptosis. KCNK5 exhibited upregulation in both in vivo and in vitro models of dry eye. The overexpression of KCNK5 was observed to induce potassium efflux and activate the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated pyroptosis in vitro. Silencing KCNK5 effectively mitigated pyroptosis in dry eye. Additionally, the overexpression of KCNK5 results in the downregulation of TNF superfamily member 10 (TNFSF10) and subsequent impairment of autophagy. TNFSF10 supplementation could promote autophagy and mitigate pyroptosis in dry eye. Conclusions: The upregulation of KCNK5 mediates TNFSF10 to impair autophagy and induce pyroptosis in dry eye. Consequently, targeting KCNK5 may represent a novel and promising approach to therapeutic intervention in the management of dry eye.


Assuntos
Síndromes do Olho Seco , Canais de Potássio de Domínios Poros em Tandem , Ligante Indutor de Apoptose Relacionado a TNF , Animais , Humanos , Camundongos , Autofagia , Síndromes do Olho Seco/metabolismo , Células Epiteliais , Camundongos Endogâmicos C57BL , Canais de Potássio de Domínios Poros em Tandem/metabolismo , Piroptose , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
2.
Int J Mol Sci ; 24(23)2023 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-38068921

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) represents a promising anticancer agent, as it selectively induces apoptosis in transformed cells without altering the cellular machinery of healthy cells. Unfortunately, the presence of TRAIL resistance mechanisms in a variety of cancer types represents a major hurdle, thus limiting the use of TRAIL as a single agent. Accumulating studies have shown that TRAIL-mediated apoptosis can be facilitated in resistant tumors by combined treatment with antitumor agents, ranging from synthetic molecules to natural products. Among the latter, flavonoids, the most prevalent polyphenols in plants, have shown remarkable competence in improving TRAIL-driven apoptosis in resistant cell lines as well as tumor-bearing mice with minimal side effects. Here, we summarize the molecular mechanisms, such as the upregulation of death receptor (DR)4 and DR5 and downregulation of key anti-apoptotic proteins [e.g., cellular FLICE-inhibitory protein (c-FLIP), X-linked inhibitor of apoptosis protein (XIAP), survivin], underlying the TRAIL-sensitizing properties of different classes of flavonoids (e.g., flavones, flavonols, isoflavones, chalcones, prenylflavonoids). Finally, we discuss limitations, mainly related to bioavailability issues, and future perspectives regarding the clinical use of flavonoids as adjuvant agents in TRAIL-based therapies.


Assuntos
Antineoplásicos , Flavonoides , Neoplasias , Animais , Camundongos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose , Linhagem Celular Tumoral , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Ligantes , Neoplasias/tratamento farmacológico , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
3.
Int J Mol Sci ; 24(14)2023 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-37511599

RESUMO

Tubeimoside-1 (TBMS-1), a traditional Chinese medicinal herb, is commonly used as an anti-cancer agent. In this study, we aimed to investigate its effect on the sensitization of cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Our results revealed that even though monotherapy using TBMS-1 or TRAIL at sublethal concentrations did not affect cancer cell death, combination therapy using TBMS-1 and TRAIL increased apoptotic cell death. Mechanistically, TBMS-1 destabilized c-FLIP expression by downregulating STAMBPL1, a deubiquitinase (DUB). Specifically, when STAMBPL1 and c-FLIP bound together, STAMBPL1 deubiquitylated c-FLIP. Moreover, STAMBPL1 knockdown markedly increased sensitivity to TRAIL by destabilizing c-FLIP. These findings were further confirmed in vivo using a xenograft model based on the observation that combined treatment with TBMS-1 and TRAIL decreased tumor volume and downregulated STAMBPL1 and c-FLIP expression levels. Overall, our study revealed that STAMBPL1 is essential for c-FLIP stabilization, and that STAMBPL1 depletion enhances TRAIL-mediated apoptosis via c-FLIP downregulation.


Assuntos
Apoptose , Ligante Indutor de Apoptose Relacionado a TNF , Humanos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Linhagem Celular Tumoral , Regulação para Baixo , Ligantes , Peptídeo Hidrolases/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais
4.
J Oral Pathol Med ; 52(8): 718-726, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37317871

RESUMO

BACKGROUND: Tumor necrosis factor-related apoptosis-inducing ligand activates apoptotic pathways and could potentially be used in anticancer treatments. However, oral squamous cell carcinoma cells are known to be resistant to tumor necrosis factor-related apoptosis-inducing ligand-induced cell death. It has been previously reported that hyperthermia upregulates tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in other cancers. As such, we evaluated whether hyperthermia upregulates tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in a tumor necrosis factor-related apoptosis-inducing ligand-resistant oral squamous cell carcinoma cell line. METHODS: The oral squamous cell carcinoma cell line HSC3 was cultured and divided into hyperthermia and control groups. We investigated the antitumor effects of recombinant human tumor necrosis factor-related apoptosis-inducing ligand using cell proliferation and apoptosis assays. Additionally, we measured death receptor 4 and 5 levels, and determined death receptor ubiquitination status, as well as E3 ubiquitin ligase targeting of death receptor in both hyperthermia and control groups before recombinant human tumor necrosis factor-related apoptosis-inducing ligand administration. RESULTS: Treatment with recombinant human tumor necrosis factor-related apoptosis-inducing ligand produced greater inhibitory effects in the hyperthermia group than in the control group. Moreover, death receptor protein expression in the hyperthermia group was upregulated on the cell surface (and overall), although death receptor mRNA was downregulated. The half-life of death receptor was several hours longer in the hyperthermia group; concomitantly, E3 ubiquitin ligase expression and death receptor ubiquitination were downregulated in this group. CONCLUSION: Our findings suggested that hyperthermia enhances apoptotic signaling by tumor necrosis factor-related apoptosis-inducing ligand via the suppression of death receptor ubiquitination, which upregulates death receptor expression. These data suggest that the combination of hyperthermia and tumor necrosis factor-related apoptosis-inducing ligand has implications in developing a novel treatment strategy for oral squamous cell carcinoma.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Hipertermia Induzida , Neoplasias Bucais , Humanos , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas Reguladoras de Apoptose/farmacologia , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Ligantes , Neoplasias Bucais/terapia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases
5.
Molecules ; 25(22)2020 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-33187225

RESUMO

Oxidative stress plays an important role in cellular processes. Consequently, oxidative stress also affects etiology, progression, and response to therapeutics in various pathological conditions including malignant tumors. Oxidative stress and associated outcomes are often brought about by excessive generation of reactive oxygen species (ROS). Accumulation of ROS occurs due to dysregulation of homeostasis in an otherwise strictly controlled physiological condition. In fact, intracellular ROS levels are closely associated with the pathological status and outcome of numerous diseases. Notably, mitochondria are recognized as the critical regulator and primary source of ROS. Damage to mitochondria increases mitochondrial ROS (mROS) production, which leads to an increased level of total intracellular ROS. However, intracellular ROS level may not always reflect mROS levels, as ROS is not only produced by mitochondria but also by other organelles such as endoplasmic reticulum and peroxisomes. Thus, an evaluation of mROS would help us to recognize the biological and pathological characteristics and predictive markers of malignant tumors and develop efficient treatment strategies. In this review, we describe the pathological significance of mROS in malignant neoplasms. In particular, we show the association of mROS-related signaling in the molecular mechanisms of chemically synthesized and natural chemotherapeutic agents and photodynamic therapy.


Assuntos
Produtos Biológicos/farmacologia , Mitocôndrias/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Alcaloides de Amaryllidaceae/farmacologia , Animais , Antineoplásicos/farmacologia , Antioxidantes/química , Ácido Ascórbico/farmacologia , Linhagem Celular Tumoral , Curcumina/farmacologia , Retículo Endoplasmático/metabolismo , Homeostase/efeitos dos fármacos , Humanos , Isoquinolinas/farmacologia , Estresse Oxidativo , Paclitaxel/farmacologia , Peroxissomos/metabolismo , Fotoquimioterapia , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Taninos/farmacologia , Taxoides/farmacologia , Triterpenos/farmacologia
6.
Int J Mol Med ; 46(1): 280-288, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32319535

RESUMO

Tumor necrosis factor (TNF)­related apoptosis­inducing ligand (TRAIL), a type II transmembrane protein, is a part of the TNF superfamily of cytokines. Cantharidin, a type of terpenoid, is extracted from the blister beetles (Mylabris genus) used in Traditional Chinese Medicine. Cantharidin elicits antibiotic, antiviral and antitumor effects, and can affect the immune response. The present study demonstrated that a cantharidin and TRAIL combination treatment regimen elicited a synergistic outcome in TRAIL­resistant DU145 cells. Notably, it was also identified that cantharidin treatment initiated the downregulation of cellular FLICE­like inhibitory protein (c­FLIP) and upregulation of death receptor 5 (DR­5), and sensitized cells to TRAIL­mediated apoptosis by initiating autophagy flux. In addition, cantharidin treatment increased lipid­modified microtubule­associated proteins 1A/1B light chain 3B expression and significantly attenuated sequestosome 1 expression. Attenuation of autophagy flux by a specific inhibitor such as chloroquine and genetic modification using ATG5 small interfering RNA abrogated the cantharidin­mediated TRAIL­induced apoptosis. Overall, the results of the present study revealed that cantharidin effectively sensitized cells to TRAIL­mediated apoptosis and its effects are likely to be mediated by autophagy, the downregulation of c­FLIP and the upregulation of DR­5. They also suggested that the combination of cantharidin and TRAIL may be a successful therapeutic strategy for TRAIL­resistant prostate cancer.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Cantaridina/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/genética , Autofagia/fisiologia , Western Blotting , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sinergismo Farmacológico , Imunofluorescência , Humanos , Masculino , Microscopia Eletrônica de Transmissão , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteína Sequestossoma-1/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/genética
7.
Cell Death Differ ; 27(7): 2117-2130, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-31959913

RESUMO

Pharmacological targeting via small molecule-based chemical probes has recently acquired an emerging importance as a valuable tool to delineate molecular mechanisms. Induction of apoptosis via CD95/Fas and TRAIL-R1/2 is triggered by the formation of the death-inducing signaling complex (DISC). Caspase-8 activation at the DISC is largely controlled by c-FLIP proteins. However molecular mechanisms of this control have just started to be uncovered. In this study we report the first-in-class chemical probe targeting c-FLIPL in the heterodimer caspase-8/c-FLIPL. This rationally designed small molecule was aimed to imitate the closed conformation of the caspase-8 L2' loop and thereby increase caspase-8 activity after initial processing of the heterodimer. In accordance with in silico predictions, this small molecule enhanced caspase-8 activity at the DISC, CD95L/TRAIL-induced caspase activation, and subsequent apoptosis. The generated computational model provided further evidence for the proposed effects of the small molecule on the heterodimer caspase-8/c-FLIPL. In particular, the model has demonstrated that boosting caspase-8 activity by the small molecule at the early time points after DISC assembly is crucial for promoting apoptosis induction. Taken together, our study allowed to target the heterodimer caspase-8/c-FLIPL and get new insights into molecular mechanisms of its activation.


Assuntos
Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Caspase 8/metabolismo , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Multimerização Proteica , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/química , Caspase 8/química , Linhagem Celular Tumoral , Sobrevivência Celular , Avaliação Pré-Clínica de Medicamentos , Proteína Ligante Fas , Humanos , Modelos Moleculares , Reprodutibilidade dos Testes , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
8.
Biomolecules ; 9(12)2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31817770

RESUMO

Honokiol is a natural biphenolic compound extracted from traditional Chinese medicine Magnolia species, which have been known to display various biological effects including anti-cancer, anti-proliferative, anti-angiogenic, and anti-metastatic activities in cancer cells. Here, we found that honokiol sensitizes cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis through downregulation of anti-apoptotic proteins survivin and c-FLIP. Ectopic expression of survivin and c-FLIP markedly abolished honokiol and TRAIL-induced apoptosis. Mechanistically, honokiol induced protein degradation of c-FLIP and survivin through STAMBPL1, a deubiquitinase. STAMBPL1 interacted with survivin and c-FLIP, resulted in reduction of ubiquitination. Knockdown of STAMBPL1 reduced survivin and c-FLIP protein levels, while overexpression of STAMBPL1 inhibited honokinol-induced survivin and c-FLIP degradation. Our findings provided that honokiol could overcome TRAIL resistance through survivin and c-FLIP degradation induced by inhibition of STAMBPL1 expression.


Assuntos
Compostos de Bifenilo/farmacologia , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/metabolismo , Lignanas/farmacologia , Peptídeo Hidrolases/metabolismo , Survivina/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Proteína Reguladora de Apoptosis Semelhante a CASP8 e FADD/efeitos dos fármacos , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Peptídeo Hidrolases/genética , Extratos Vegetais/farmacologia , Survivina/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Ubiquitinação/genética
9.
Biomaterials ; 219: 119377, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31374478

RESUMO

Natural killer (NK) cells-based therapy has been used widely for cancer treatment in clinic trails. However, the immunotherapeutic efficacy of this method has been greatly hindered by tumor evasion and diminished activities of NK cells. In the present study, a selenium (Se)-bearing ruthenium (Ru) complex (RuSe) was designed that could synergistically potentiate NK cell-mediated killing against prostate cancer cells. As expected, pretreatment of cancer cells with subtoxic doses of RuSe effectively augmented the lysis potency of NK cells, with up to 2.46-fold enhancement than NK cells alone, against PC3 cells. More importantly, low concentrations of RuSe could augment the tumor destroying potency of NK cells derived from 10 clinical patients, with the enhancement range from 0.78- to 11.9-fold against PC3 cells and 0.67- to 3.8-fold against LNCAP cells. Mechanistic studies revealed that the sensitizing effect of RuSe primarily depended on TRAIL/TRAIL-R and Fas/FasL-mediated signaling. Furthermore, the increased expression level of these ligands highly relied on ROS overproduction-triggered DNA damage and the downstream ATM and ATR pathways. Furthermore, RuSe potently activated and synergized with NK cells to restrain tumor growth in vivo without causing toxic side effects on major organs. Taken together, the current study not only provides a strategy for application of metal complexes in chemo-immunotherapy but also sheds light on the potential roles and mechanisms of action on such Se-containing drugs as efficient immune-sensitizing agents for NK cell-based immunotherapy.


Assuntos
Proteína Ligante Fas/metabolismo , Imunoterapia , Células Matadoras Naturais/imunologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/terapia , Rutênio/farmacologia , Selênio/farmacologia , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Degranulação Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Dano ao DNA , Humanos , Células Matadoras Naturais/efeitos dos fármacos , Masculino , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Morte Celular/metabolismo , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Receptor fas/metabolismo
10.
Cancer Chemother Pharmacol ; 84(4): 719-728, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31281953

RESUMO

PURPOSE: Glioblastoma is the most common, malignant and devastating type of primary brain tumor. Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) is characterized by its lethality to precancerous and cancerous cells. However, many kinds of tumor cells, including most glioma cells, tend to evade TRAIL-induced apoptosis. Celastrol is a pleiotropic compound from a traditional Chinese medicine that has proven to be useful as a sensitizer for TRAIL treatment. However, the underlying mechanism and role of celastrol in the sensitization of glioma cells remain to be elucidated. METHODS: The viability of glioma cell lines was examined by the CCK-8 assay. The expression of DR5 was detected by reverse transcriptase quantitative real-time PCR. The protein expression of DR5, cleaved caspase-8, cleaved caspase-3 and PARP were measured by western blot. The apoptosis rates and the sub-G1 population were detected by flow cytometry. The cellular morphological changes were assessed by TUNEL apoptosis and Hoechst 33258 staining assays. The knockdown of DR5 expression was conducted by siRNA. RESULTS: In this study, we observed that celastrol treatment inhibited cell viability in a dose-dependent manner, while glioma and normal human astroglial cell lines were resistant to TRAIL treatment. We also observed that the antiproliferative effects of TRAIL in combination with a noncytotoxic concentration of celastrol were significantly greater than those of celastrol or TRAIL alone. In addition, cell death induced by the combination treatment was apoptotic and occurred through the death receptor pathway via activation of caspase-8, caspase-3, and PARP. Furthermore, celastrol upregulated death receptor 5 (DR5) at the mRNA and protein levels, and siRNA-mediated DR5 knockdown reduced the killing effect of the combination drug treatment on glioma cells and reduced the activation of caspase-3, caspase-8 and PARP. CONCLUSIONS: Taken together, the results of our study demonstrate that celastrol sensitizes glioma cells to TRAIL via the death receptor pathway and that DR5 plays an important role in the effects of this cotreatment. The results indicate that this cotreatment is a promising tumor-killing therapeutic strategy with high efficacy and low toxicity.


Assuntos
Apoptose/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF , Triterpenos/farmacocinética , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Medicamentos de Ervas Chinesas/farmacocinética , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Triterpenos Pentacíclicos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Tripterygium , Regulação para Cima
11.
Phytomedicine ; 62: 152950, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31102888

RESUMO

BACKGROUND: The ability of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to preferentially induce apoptosis in transformed cells while sparing most normal cells is well established. However, the intrinsic and acquired resistance of tumors to TRAIL-induced apoptosis limits its therapeutic applicability. PURPOSE: We investigated the effect of caudatin, a species of C-21 steroidal glycosides isolated from the roots of Cynanchum auriculatum, on TRAIL-induced apoptosis in human breast cancer cells. METHODS: Cell growth inhibition was evaluated by the CCK-8 assay. The cell cycle distribution was assessed by propidium iodide flow cytometry. Apoptosis was determined by TUNEL staining. Protein expression was detected by western blotting analysis. RESULTS: Caudatin enhanced TRAIL-induced apoptosis in human breast cancer cells. This sensitization was achieved by upregulating death receptor 5 (DR5). Knockdown of DR5 abolished the enhancing effect of caudatin on TRAIL responses. The caudatin-induced upregulation of DR5 was accompanied by increased expression of CHOP and phosphorylation of p38 MAPK and JNK. CHOP knockdown blocked caudatin-upregulated DR5 expression. Moreover, cotreatment of breast cancer cells with p38 MAPK and JNK inhibitors significantly counteracted the caudatin-induced expression of DR5. CONCLUSION: Our results showed that caudatin sensitized breast cancer cells to TRAIL-induced apoptosis through activation of CHOP, p38 MAPK and JNK-mediated upregulation of DR5 expression. The combination of TRAIL and caudatin may be a promising therapeutic approach for the treatment of breast cancer.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Glicosídeos/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Esteroides/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Fator de Transcrição CHOP/genética , Fator de Transcrição CHOP/metabolismo , Regulação para Cima/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
12.
Mol Med Rep ; 19(5): 3799-3806, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30896802

RESUMO

Ent­3α­formylabieta­8(14),13(15)­dien­16,12ß­olide (EFLDO) is a compound extracted from Euphorbia lunulata Bge exhibiting anti­proliferative activity in vitro. In the present study, EFLDO was identified to sensitize HepG2 cells to tumor necrosis factor (TNF) superfamily member 10 (TNFSF10)­induced apoptosis. Liver cancer cells were resistant to TNFSF10; however, EFLDO increased TNFSF10­induced cancer cell viability inhibition and cell apoptosis induction as assessed by MTT assay and Annexin V­fluorescein isothiocyanate (FITC)/propidium iodide assay, respectively. The western blotting results suggested that treatment with EFLDO increased TNFSF10­induced upregulation of the protein expression levels of pro­apoptotic proteins, including BCL2 associated agonist of cell death, BCL2 associated X, apoptosis regulator, caspase­3 (CASP3) and CASP8. Furthermore, treatment with EFLDO increased TNFSF10­mediated downregulation of the protein expression level of the anti­apoptotic protein BCL2 apoptosis regulator. Notably, the increase in the activity of CASP3 was consistent with the western blotting results. Treatment with EFLDO sensitized liver cancer cells to TNFSF10, and apoptosis was induced via the upregulation of TNF receptor superfamily member 10a (TNFRSF10A) and TNFRSF10B in a tumor protein p53 (p53)­dependent manner, as detected by reverse transcription­quantitative polymerase chain reaction and western blot analyses. In addition, p53 was identified to be necessary for EFLDO­induced sensitivity to TNFSF10, as assessed by western blotting and Annexin V­FITC assay. Collectively, the present results suggested a novel mechanism underlying EFLDO function in liver cancer. Treatment with EFLDO was able to increase the antitumor effect of TNFSF10 in liver cancer cells in a p53­dependent manner.


Assuntos
Abietanos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Extratos Vegetais/farmacologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Euphorbia/química , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
13.
Am J Chin Med ; 47(1): 237-257, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30612454

RESUMO

The aim of this study was to investigate the effects of bufalin on human nasopharyngeal carcinoma NPC-TW 076 cells in vitro. Bufalin is a cardiotonic steroid and a key active ingredient of the Chinese medicine ChanSu. The extracts of Chansu are used for various cancer treatments in China. In the present study, bufalin induced cell morphological changes, decreased total cell viability and induced G2/M phase arrest of cell cycle in NPC-TW 076 cells. Results also indicated that bufalin induced chromatin condensation (cell apoptosis) and DNA damage by DAPI staining and comet assay, respectively. The induced apoptotic cell death was further confirmed by annexin-V/PI staining assay. In addition, bufalin also increased ROS and Ca 2+ production and decreased the levels of ΔΨm . Furthermore, the alterations of ROS, ER stress and apoptosis associated protein expressions were investigated by Western blotting. Results demonstrated that bufalin increased the expressions of ROS associated proteins, including SOD (Cu/Zn), SOD2 (Mn) and GST but decreased that of catalase. Bufalin increased ER stress associated proteins (GRP78, IRE-1 α , IRE-1 ß , caspase-4, ATF-6 α , Calpain 1, and GADD153). Bufalin increased the pro-apoptotic proteins Bax, and apoptotic associated proteins (cytochrome c, caspase-3, -8 and -9, AIF and Endo G) but reduced anti-apoptotic protein Bcl-2 in NPC-TW 076 cells. Furthermore, bufalin elevated the expressions of TRAIL-pathway associated proteins (TRAIL, DR4, DR5, and FADD). Based on these findings, we suggest bufalin induced apoptotic cell death via caspase-dependent, mitochondria-dependent and TRAIL pathways in human nasopharyngeal carcinoma NPC-TW 076 cells.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/genética , Bufanolídeos/farmacologia , Mitocôndrias/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/patologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Extratos de Tecidos/farmacologia , Bufanolídeos/química , Bufanolídeos/isolamento & purificação , Caspases/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Chaperona BiP do Retículo Endoplasmático , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Humanos , Carcinoma Nasofaríngeo/metabolismo , Superóxido Dismutase/metabolismo , Superóxido Dismutase-1/metabolismo , Extratos de Tecidos/isolamento & purificação , Células Tumorais Cultivadas
14.
Biomed Pharmacother ; 109: 2182-2191, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30551475

RESUMO

This work focused on characterizing hydrophilic fractions of Clematis flammula (CFl). The data here clearly demonstrated that hydrolate fractions act as a free radical scavengers and inhibited proliferation of different cell lines in a time- and concentration-dependent manner, transwell, and with a significant cytotoxic effect. Treating cells with CFl had the effect of suppressing cell growth attenuated by ROS generation in colonic carcinoma. Moreover, CFl in HCT116 cells suppressed survival, proliferation, invasion, angiogenesis and metastasis in vitro by inhibiting gene expression. Following CFl treatment, caspases and PARP cleavage were detected. The up- and down-regulated genes obtained from the WBA of the effect of CFl showed that several biological processes were associated with apoptosis and induction of G1 cell cycle arrest. CFl synergizes the effect of TRAIL by down-regulating the expression of cell survival proteins involved in apoptosis compared to cells treated with CFl or TRAIL alone. Our findings showed that CFl sensitizes apoptosis in TRAIL-resistant cells by activating MAPKs, SP1, and CHOP, that induced DR5 expression. Overall, our data showed that CFl is a promising antitumor agent through kinases and transcription factor induction, both of which are required to activate TRAIL receptors. Colon inflammation induced by LPS was inhibited by CFl hydrolate.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Clematis , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Extratos Vegetais/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Animais , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HL-60 , Humanos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Células MCF-7 , Camundongos , Componentes Aéreos da Planta , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/farmacologia
15.
Int J Mol Med ; 43(2): 701-708, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30483736

RESUMO

Tumor necrosis factor (TNF)­related apoptosis­inducing ligand (TRAIL) is a member of the TNF superfamily and is an antitumor drug that induces apoptosis in tumor cells with minimal or no effects on normal cells. Here, it is demonstrated that 6­shogaol (6­sho), a bioactive component of ginger, exerted anti­inflammatory and anticancer properties, attenuated tumor cell propagation and induced TRAIL­mediated cell death in liver cancer cells. The current study identified a potential pathway by revealing that TRAIL and 6­sho or chloroquine acted together to trigger reactive oxygen species (ROS) production, to upregulate tumor­suppressor protein 53 (p53) expression and to change the mitochondrial transmembrane potential (MTP). Treatment with N­acetyl­L­cysteine reversed these effects, restoring the MTP and attenuated ROS production and p53 expression. Interestingly, treatment with 6­sho increased p62 and microtubule­associated proteins 1A/1B light chain 3B­II levels, indicating an inhibited autophagy flux. In conclusion, attenuation of 6­sho­induced autophagy flux sensitized cells to TRAIL­induced apoptosis via p53 and ROS, suggesting that the administration of TRAIL in combination with 6­sho may be a suitable therapeutic method for the treatment of TRAIL­resistant Huh7 liver cells.


Assuntos
Autofagia/efeitos dos fármacos , Catecóis/farmacologia , Neoplasias Hepáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Proteína Supressora de Tumor p53/biossíntese , Acetilcisteína/farmacologia , Apoptose/efeitos dos fármacos , Caspases Iniciadoras/metabolismo , Catecóis/uso terapêutico , Linhagem Celular Tumoral , Cloroquina/metabolismo , Cloroquina/farmacologia , Cloroquina/uso terapêutico , Sinergismo Farmacológico , Quimioterapia Combinada , Zingiber officinale/química , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mutagênicos/farmacologia , Mutagênicos/uso terapêutico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico
16.
Cell Biol Int ; 42(10): 1377-1385, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29957841

RESUMO

ß-Elemene, an anti-cancer drug extracted from traditional Chinese medicinal herb, showed anti-tumor effects on gastric cancer cells. Our previous studies reported gastric cancer cells are insensitive to TRAIL. However, whether ß-elemene could enhance anti-cancer effects of TRAIL on gastric cancer cells is unknown. In our present study, ß-elemene prevented gastric cancer cell viability in dose-dependent manner, and when combined with TRAIL, obviously inhibited proliferation and promoted apoptosis in gastric cancer cells. Compared to ß-elemene or TRAIL alone, treatment with ß-elemene and TRAIL obviously promoted DR5 clustering as well as translocation of Caspase-8, DR5 and FADD into lipid rafts. This led to cleavage of Caspase-8 and the formation of death-inducing signaling complex (DISC) in lipid rafts. The cholesterol-sequestering agent nystatin partially reversed DR5 clustering and DISC formation, preventing apoptosis triggered by the combination of ß-elemene and TRAIL. Our results suggest that ß-elemene increases the sensitivity of gastric cancer cells to TRAIL partially by promoting the formation of DISC in lipid rafts.


Assuntos
Sesquiterpenos/metabolismo , Sesquiterpenos/farmacologia , Neoplasias Gástricas/metabolismo , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 8/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , China , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/efeitos dos fármacos , Proteínas Adaptadoras de Sinalização de Receptores de Domínio de Morte/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Microdomínios da Membrana , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/patologia , Ligante Indutor de Apoptose Relacionado a TNF/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia
17.
Biomaterials ; 176: 60-70, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29860138

RESUMO

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) can induce apoptosis in cancer cells without toxicity to normal cells. However, the efficiency is greatly limited by its short half-life and wild resistance in various cancer cells. In this study, we reported a micellar hybrid nanoparticle to carry TRAIL ligand (denoted as IPN@TRAIL) for a novel photo-excited TRAIL therapy. These IPN@TRAIL offered increased TRAIL stability, prolonged half-life and enhanced tumor accumulation, monitored by dual mode imaging. Furthermore, IPN@TRAIL nanocomposites enhanced wrapped TRAIL therapeutic efficiency greatly towards resistant cancer cells by TRAIL nanovectorization. More importantly, when upon external laser, these nanocomposites not only triggered tumor photothermal therapy (PTT), but also upregulated the expression of death receptors (DR4 and DR5), resulting in a greater apoptosis mediated by co-delivered TRAIL ligand. Such photo/TRAIL synergistic effect showed its great killing effects in a controllable manner on TRAIL-resistant A549 tumor model bearing mice. Finally, these nanocomposites exhibited rapid clearance without obvious systemic toxicity. All these features rendered our nanocomposites a promising theranostic platform in cancer therapy.


Assuntos
Antineoplásicos/administração & dosagem , Nanocompostos/química , Neoplasias/diagnóstico por imagem , Neoplasias/terapia , Fototerapia/métodos , Ligante Indutor de Apoptose Relacionado a TNF/administração & dosagem , Células A549 , Animais , Sobrevivência Celular/efeitos dos fármacos , Terapia Combinada , Liberação Controlada de Fármacos , Resistencia a Medicamentos Antineoplásicos , Humanos , Nanopartículas de Magnetita/química , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanocápsulas/química , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/química , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Microambiente Tumoral
18.
Biomacromolecules ; 19(6): 2194-2201, 2018 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-29684275

RESUMO

The assembly of low molecular weight polymers into highly efficient and nontoxic nanostructures has broad applicability in gene delivery. In this study, we reported the assembly of coumarin-anchored low generation dendrimers in aqueous solution via hydrophobic interactions. The synthesized material showed significantly improved DNA binding and gene delivery, and minimal toxicity on the transfected cells. Moreover, the coumarin moieties in the assembled nanostructures endow the materials with light-responsive drug delivery behaviors. The coumarin substitutes in the assembled nanostructures were cross-linked with each other upon irradiation at 365 nm, and the cross-linked assemblies were degraded upon further irradiation at 254 nm. As a result, the drug-loaded nanoparticle showed a light-responsive drug release behavior and light-enhanced anticancer activity. The assembled nanoparticle also exhibited a complementary anticancer activity through the codelivery of 5-fluorouracil and a therapeutic gene encoding tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). This study provided a facile strategy to develop light-responsive polymers for the codelivery of therapeutic genes and anticancer drugs.


Assuntos
Antineoplásicos , Cumarínicos , Dendrímeros , Sistemas de Liberação de Medicamentos , Fluoruracila , Técnicas de Transferência de Genes , Nanoestruturas , Neoplasias/terapia , Fototerapia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cumarínicos/química , Cumarínicos/farmacocinética , Cumarínicos/farmacologia , Dendrímeros/química , Dendrímeros/farmacocinética , Dendrímeros/farmacologia , Fluoruracila/química , Fluoruracila/farmacocinética , Fluoruracila/farmacologia , Células HEK293 , Humanos , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
19.
Pharmacol Res ; 128: 366-375, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28951297

RESUMO

Activator protein 1 (AP-1) is a key transcription factor in the control of several cellular processes responsible for cell survival proliferation and differentiation. Dysfunctional AP-1 expression and activity are involved in several severe diseases, especially inflammatory disorders and cancer. Therefore, targeting AP-1 has recently emerged as an attractive therapeutic strategy for cancer prevention and therapy. This review summarizes our current understanding of AP-1 biology and function as well as explores and discusses several natural bioactive compounds modulating AP-1-associated signaling pathways for cancer prevention and intervention. Current limitations, challenges, and future directions of research are also critically discussed.


Assuntos
Neoplasias/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Apoptose/efeitos dos fármacos , Produtos Biológicos/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Compostos Fitoquímicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
20.
Cell Mol Biol (Noisy-le-grand) ; 63(9): 53-58, 2017 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-28980934

RESUMO

Past several years have witnessed dramatic leaps in our understanding of rewiring of gene expression at the translation level during cancer developmentthat provides linchpin support to the transformed phenotype. Most recent and ground-breaking developments in the field of molecular oncology aredriven by an explosion in technological advancements and have started to reveal previously unimagined regulatory mechanisms and how they intricately co-ordinate to modulate cancer progression, loss of apoptosis and development of resistance against different therapeutics. However, the insights gained from work in this natural product research have far-reaching impact because of rapidly increasing repertoire of medicinally and biologically efficient phytochemicals. How Tanshinones mediate targeting of JAK-STAT, ER stress associated signaling cascade,PI3K/AKT/mTOR pathway,autophagy, TRAIL pathway and microRNAs are being discovered and will prove to be helpful in getting a step closer to personalized medicine.


Assuntos
Abietanos/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Abietanos/farmacologia , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinases da Matriz/metabolismo , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA