Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Gen Subj ; 1866(5): 130113, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35202768

RESUMO

Breast cancer (BCa) has become the leading cause of death in women worldwide. Irrespective of advancement in cancer treatments, e.g., surgery, radiation, chemotherapy, hormonal therapy, immunotherapy, and targeted therapy, recurrence leading to metastasis poses the greatest threat in BCa management. BCa receptors estrogen (ER), progesterone (PR), and human epidermal growth factor receptor-2 (HER2) hold significant reputations as prognostic and predictive biomarkers in therapeutic decision-making. Under normal physiological conditions, these receptors modulate critical biological functions, e.g., cell migration, proliferation, and apoptosis events, etc. However, aberrant expression causes deviations, triggering signaling course to adapt permanent switching "ON" mode. The later events induce rapid and unrestrained proliferation leading to cancer. As conventional ways of cancer management ultimately lead to resistance; therefore, recently targeted therapies have been extensively studied to conquer resistance. Targeting various small molecules in downstream signaling has become an area of interest in scientific society. The severity of cancer converts many folds soon after it takes on a migratory approach that eventually commences metastasis. Cancer migration comprises protrusion of cytoplasm at the leading edge of the migration forward-facing, establishing adhesions with the basic cell-matrix, disassembly of the adhesions at the back end of the cell, and actin-myosin fiber contractions to pull the bulk of the cytoplasm forward. On the other hand, metastatic progression comprises a cascade of events, including invasion, migration, and establishment of tumor microenvironment. The progression of BCa from early stage to metastatic development causes remarkable heterogeneity. Interference at any explicit level could hamper the process, and it has thus become an area of interest for scientists. Metastasis is the ultimate cause of spreading tumor cells to invade distant organs. Recently small molecule inhibitors of protein tyrosine kinases, which can cross the blood-brain barrier, have become a center point of research for investigators in developing novel treatment strategies against BCa management.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/patologia , Movimento Celular , Estrogênios/uso terapêutico , Feminino , Humanos , Progesterona , Microambiente Tumoral
2.
Arch Pharm Res ; 44(11): 1025-1036, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34751932

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common tumor types globally. Despite the progress made in surgical procedures and therapeutic options, HCC remains a considerable cause of cancer-related mortality. In this study, we investigated the antitumor effects of sanguinarine (Sang) on HCC and its potential mechanisms. Our findings showed that Sang impairs the acidic environment of lysosomes by inhibiting cathepsin D maturation. In addition, Sang inhibited the formation of autolysosomes in RFP-GFP-LC3 transfected cells, subsequently suppressing late mitophagy. Sang also induced reactive oxygen species (ROS)-dependent autophagy and apoptosis in HCC cells, which was significantly attenuated following treatment with a ROS scavenger. Further investigation using autophagy inhibitors revealed that sanguinarine-induced mitochondrial dysfunction and mitophagy led to mitochondrial apoptosis in HCC cells. Immunohistochemical staining of sanguinarine-treated xenograft samples revealed that it initiated and blocked autophagy. In summary, our findings suggest that in HCC cells, Sang impairs lysosomal function and induces ROS-dependent mitophagy and apoptosis.


Assuntos
Benzofenantridinas/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Isoquinolinas/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Benzofenantridinas/uso terapêutico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Isoquinolinas/uso terapêutico , Neoplasias Hepáticas/patologia , Lisossomos/efeitos dos fármacos , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/patologia , Mitofagia/efeitos dos fármacos , Espécies Reativas de Oxigênio/agonistas , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Cell Death Dis ; 12(5): 490, 2021 05 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990544

RESUMO

Lung cancer (LC) is one of the leading causes of cancer-related death. As one of the key features of tumor microenvironment, hypoxia conditions are associated with poor prognosis in LC patients. Upregulation of hypoxic-induced factor-1α (HIF-1α) leads to the activation of various factors that contribute to the increased drug resistance, proliferation, and migration of tumor cells. Apurinic/apyrimidinic endonuclease-1 (APEX1) is a multi-functional protein that regulates several transcription factors, including HIF-1α, that contribute to tumor growth, oxidative stress responses, and DNA damage. In this study, we explored the mechanisms underlying cell responses to hypoxia and modulation of APEX1, which regulate HIF-1α and downstream pathways. We found that hypoxia-induced APEX1/HIF-1α pathways regulate several key cellular functions, including reactive oxygen species (ROS) production, carbonic anhydrase 9 (CA9)-mediated intracellular pH, migration, and angiogenesis. Cephalomannine (CPM), a natural compound, exerted inhibitory effects in hypoxic LC cells via the inhibition of APEX1/HIF-1α interaction in vitro and in vivo. CPM can significantly inhibit cell viability, ROS production, intracellular pH, and migration in hypoxic LC cells as well as angiogenesis of HUVECs under hypoxia through the inhibition of APEX1/HIF-1α interaction. Taken together, CPM could be considered as a promising compound for LC treatment.


Assuntos
DNA Liase (Sítios Apurínicos ou Apirimidínicos)/metabolismo , Neoplasias Pulmonares/genética , Espécies Reativas de Oxigênio/metabolismo , Taxoides/metabolismo , Animais , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Microambiente Tumoral
4.
Phytomedicine ; 84: 153500, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33626427

RESUMO

BACKGROUND: A large number of breast cancer patients perishes due to metastasis instead of primary tumor, but molecular mechanisms contributing towards cancer metastasis remain poorly understood. Therefore, prompting development of novel treatment is inevitable. A vast variety of plant derived natural substance possesses several therapeutically active constituents, e.g. alkaloids, flavonoids, tannins, resins, terpenoids etc. that exhibit various pharmacological properties e.g. anti-inflammatory, anti-microbial and anti-cancer properties. Sanguinarine (SAN) alkaloid found its place among such naturally occurring substances that exerts several pharmacological activities, including anti-cancer effects. PURPOSE: Until now, role of SAN not only against epithelial-mesenchymal transition (EMT) but also against metastasis progression in breast cancer remains indistinct. Thus, aim of the present study was to investigate effects of SAN on EMT process and cancer metastasis in animal model. METHODS: MTT assay was performed to assess SAN effects on proliferation in breast cancer. Scratch assay was performed to evaluate effects of SAN on migration in breast cancer. Colony formation assay was performed to determine effects of SAN on colonization characteristics of breast cancer. Western blotting was performed to measure EMT regulating protein expression as well as major pathway protein expression induced against TGF-ß treatment in breast cancer. Tail vein method of injecting breast cancer cells in bulb/c mice was conducted to study metastasis progression and thereafter assessing effects of SAN against metastasis in mice. RESULTS: In vivo results: MTT assay performed, demonstrated dose dependent inhibition of cell proliferation in breast cancer. Scratch assay results showed, SAN played a major role as migration inhibitor in estrogen receptor positive (ER+) breast cancer. Colony forming assay results demonstrated that SAN constrains ability of breast cancer to develop into well-defined colonies. Western blotting results for EMT regulating protein expression, after TGF-ß treatment showed, SAN inhibited cadherin switch in ER+ breast cancer. Moreover, expression of pathway proteins involved in EMT process after TGF-ß treatment i.e. Smad, PI3K/Akt and MAP kinase were significantly masked against SAN treatment. IN VIVO RESULTS: The appearance of metastatic nodules in lung tissues of mice model, helps to study the effects of SAN against metastasis in bulb/c mice. The obtained results have confirmed that SAN impeded lung metastasis. The macroscopic examination has confirmed metastasis inhibitory role of SAN in breast cancer. The Hematoxylin and eosin (H&E) staining results further advocate anti-metastatic characteristics of SAN, presented by fewer metastatic nodule and lesions appearance in SAN treated mice compared to untreated metastasis mice. CONCLUSION: In summary, SAN displayed prominent anti-metastatic effects in animal model and anti-proliferation effects together with significant inhibitory potential on EMT regulating protein expression against TGF-ß treatment in ER+ breast cancer. So, overall findings of our study highlighted the pre-clinical significance of SAN in animal model therefore, further studies in humans as a part of clinical trial will be needed to establish pharmacokinetics and other effects of SAN, so that it can be a potential candidate for future treatment of metastatic breast cancer (MBC).


Assuntos
Benzofenantridinas/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Isoquinolinas/farmacologia , Metástase Neoplásica/prevenção & controle , Animais , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Fosfatidilinositol 3-Quinases , Fator de Crescimento Transformador beta/farmacologia
5.
Phytomedicine ; 84: 153503, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33636580

RESUMO

BACKGROUND: Breast cancer is the most common female cancer worldwide. Large hypoxic area is one of the features of tumor microenvironment. Highly activated hypoxia-induced pathways positively correlate with poor clinical response to chemo- and radiotherapy and high mortality in breast cancer patients. PURPOSE: We explore the effect of sanguinarine on hypoxia-induced activation of Ephrin type-B receptor 4 (EphB4) and hypoxia inducible factor-1α (HIF-1α) pathways in breast cancer. RESULTS: Hypoxia-induced expression of a receptor tyrosine kinase EphB4 was observed in hypoxic breast cancer cell models. Sanguinarine, a natural alkaloid, could effectively combat hypoxia-induced EphB4 and HIF-1α expression. Sanguinarine inhibited the activation of downstream protein signal transducer and activator of transcription-3 (STAT3), thereby blocking hypoxia-induced HIF-1α/STAT3 interaction and downregulating the mRNA levels of their target genes. Mechanically, sanguinarine attenuated HIF-1α protein levels via inhibition of MAPK/ERK pathways and promotion of HIF-1α proteasome degradation. Sanguinarine inhibited STAT3 activation through targeting its upstream EphB4 and accelerating STAT3 dephosphorylation. Correspondingly, xenograft models confirmed that sanguinarine treatment disrupted hypoxia-induced pathways and inhibited tumor growth in vivo. CONCLUSIONS: Our results may bring insights to the hypoxia-induced pathways in breast cancers, and suggest sanguinarine as a promising candidate for EphB4 and HIF-1α-targeted inhibition.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Benzofenantridinas/farmacologia , Neoplasias da Mama/tratamento farmacológico , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isoquinolinas/farmacologia , Receptor EphB4/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Camundongos Endogâmicos BALB C , Receptor EphB4/genética , Fator de Transcrição STAT3/metabolismo , Hipóxia Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Cell Mol Med ; 24(6): 3756-3761, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32065498

RESUMO

Breast cancer is one leading cause of death in females, especially triple-negative breast cancer (TNBC). Hypoxia is a key feature leading to tumour progression driven by hypoxia-inducible factor (HIF)-1α. The aim is to investigate the mechanism of HIF-1α and signal transducer and activator of transcription-3 (STAT3) interaction and discover a compound to disrupt the interaction in breast cancer cells. The regulation pattern of HIF-1α and STAT3 was analysed in hypoxic TNBC cells and patient samples. The effects of a natural alkaloid, sanguinarine, on HIF-1α and STAT3 colocalization and interaction were evaluated in vitro and mouse xenograft models. We observed strong colocalization of HIF-1α, p-STAT3-Tyr and p-STAT3-Ser in TNBC patient samples. Sanguinarine could inhibit the nuclear colocalization and interaction of HIF-1α with p-STAT3-Tyr and p-STAT3-Ser in vivo and in vitro. Our results may bring insights to the HIF-1α/STAT3 interaction in breast cancers and suggest sanguinarine as a promising candidate for HIF-α/STAT3 inhibition.


Assuntos
Benzofenantridinas/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isoquinolinas/farmacologia , Fator de Transcrição STAT3/metabolismo , Serina/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Tirosina/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos
7.
Cell Death Dis ; 10(12): 939, 2019 12 09.
Artigo em Inglês | MEDLINE | ID: mdl-31819036

RESUMO

Epithelial-mesenchymal transition (EMT) plays a crucial role in hepatocellular carcinoma (HCC) progression. Hypoxia and excessive transforming growth factor-ß (TGF-ß) have been identified as inducers and target for EMT in HCC. Here, we show hypoxia inducible factor-1α (HIF-1α) and TGF-ß form a feed-forward loop to induce EMT in HCC cells. Further mechanistic study indicates under both hypoxia and TGF-ß stimulation, Smad and PI3K-AKT pathways are activated. We show sanguinarine, a natural benzophenanthridine alkaloid, impairs the proliferation of nine kinds of HCC cell lines and the colony formation of HCC cells. In hypoxic and TGF-ß cell models, sanguinarine inhibits HIF-1α signaling and the expression of EMT markers, translocation of Snail and activation of both Smad and PI3K-AKT pathways. Sanguinarine could also inhibit TGF-ß-induced cell migration in HCC cells. In vivo studies reveal that the administration of sanguinarine inhibits tumor growth and HIF-1α signaling, inhibits the expression changes of EMT markers as well as Smad and PI3K-AKT pathway proteins. Our findings suggest that sanguinarine is a promising candidate targeting HIF-1α/TGF-ß signaling to improve the treatment for HCC patients.


Assuntos
Benzofenantridinas/farmacologia , Benzofenantridinas/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Isoquinolinas/farmacologia , Isoquinolinas/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Hipóxia Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...