Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Med Res Rev ; 44(2): 457-496, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37589457

RESUMO

Neem (Azadirachta indica A. Juss.), a versatile evergreen tree recognized for its ethnopharmacological value, is a rich source of limonoids of the triterpenoid class, endowed with potent medicinal properties. Extracts of neem have been documented to display anticancer effects in diverse malignant cell lines as well as in preclinical animal models that has largely been attributed to the constituent limonoids. Of late, neem limonoids have become the cynosure of research attention as potential candidate agents for cancer prevention and therapy. Among the various limonoids found in neem, azadirachtin, epoxyazadiradione, gedunin, and nimbolide, have been extensively investigated for anticancer activity. Azadirachtin, a potent biodegradable pesticide, exhibits profound antiproliferative effects by preventing mitotic spindle formation and cell division. The antiproliferative activity of gedunin has been demonstrated to be mediated primarily via inhibition of heat shock protein90 and its client proteins. Epoxyazadiradione inhibits pro-inflammatory and kinase-driven signaling pathways to block tumorigenesis. Nimbolide, the most potent cytotoxic neem limonoid, inhibits the growth of cancer cells by regulating the phosphorylation of keystone kinases that drive oncogenic signaling besides modulating the epigenome. There is overwhelming evidence to indicate that neem limonoids exert anticancer effects by preventing the acquisition of hallmark traits of cancer, such as cell proliferation, apoptosis evasion, inflammation, invasion, angiogenesis, and drug resistance. Neem limonoids are value additions to the armamentarium of natural compounds that target aberrant oncogenic signaling to inhibit cancer development and progression.


Assuntos
Antineoplásicos , Azadirachta , Limoninas , Animais , Humanos , Limoninas/farmacologia , Antineoplásicos/farmacologia , Extratos Vegetais
2.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37511200

RESUMO

Patients with comorbidities of obesity and diabetes are recognized to be at high risk of breast cancer development and face worse breast cancer outcomes. Though several reports showed the reinforced link between obesity, diabetes, and prediabetes with breast cancer, the underlying molecular mechanisms are still unknown. The present study aimed to investigate the underlying molecular link between increased risks of breast cancer due to coincident diabetes or obesity using a spontaneous obese rat model with impaired glucose tolerance (WNIN/GR-Ob rat). A single dose of solubilized DMBA suspension (40 mg/kg body weight) was orally administered to the animals at the age of 60 days to induce breast tumors. The tumor incidence, latency period, tumor frequency, and tumor volume were measured. Histology, immunohistochemistry, and immunoblotting were performed to evaluate the tumor morphology and expression levels of signal molecules. The development of mammary tumors in GR-Ob rats was characterized by early onset and shorter latency periods compared to control lean rats. While 62% of obese rats developed breast tumors, tumor development in lean rats was only 21%. Overexpression of ER, PR, Ki67, and p53 markers was observed in tumor tissues of obese rats in comparison with lean rats. The levels of the hallmarks of cell proliferation and angiogenesis involved in IGF-1/PI3K/Akt/GSK3ß/ß-catenin signaling pathway molecules were upregulated in obese rat breast tumors compared to lean rats. Furthermore, obesity with prediabetes is associated with changes in IGF-1 signaling and acts on PI3K/Akt/GSK3ß/ß-catenin signaling, which results in rapid cell proliferation and development of breast tumors in obese rats than the lean rats. These results indicate that tumor onset and development were faster in spontaneous obese rat models with impaired glucose tolerance than in their lean counterparts.


Assuntos
Intolerância à Glucose , Neoplasias , Estado Pré-Diabético , Ratos , Animais , Intolerância à Glucose/complicações , Glicogênio Sintase Quinase 3 beta , Fator de Crescimento Insulin-Like I , beta Catenina , Estado Pré-Diabético/complicações , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Obesidade/metabolismo , Neoplasias/complicações
3.
Biotechnol Appl Biochem ; 69(5): 1929-1941, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34554598

RESUMO

Human papillomavirus (HPV) infection-dependent cervical cancer is one of the most common gynecological cancers and often becomes aggressive, with rapid proliferation, invasion/migration, and drug resistance. Here, 135 fresh human cervical squamous cell carcinoma (CSCC) tissue specimens, comprising 21 adjacent normal (AN), 30 cervical intraepithelial neoplasia (CIN1-3 ), 45 CSCC, and 39 drugs (chemo-radiation)-resistant cervical tumor (DRCT) tissues were included. HPV-positive (HeLa, SiHa), HPV-negative (C33A), and cisplatin-resistant (CisR-HeLa/-SiHa/-C33A) cell lines were used for in vitro studies. HPV16/18 oncoproteins E6/E7, pERK1/2, and glycogen synthase kinase-3 (GSK3) and the matrix metalloproteinases (MMPs) MMP-9/-2 were assessed using immunohistochemistry, WB, and gelatin zymography. HPV16/18 infection was observed in 16.7% of the CIN1-3 , 77.8% of the CSCC, and 89.7% of DRCT samples. Total and inactive GSK3ß expressions were associated with overall CSCC progression (p = 0.039 and p = 0.024, respectively) and chemoresistance (p = 0.004 and p = 0.014, respectively). Positive correlations were observed, between the expression of E6 and pGSK3ß expression (p = 0.013); E6 and CSCC progression (p < 0.0001)/drug resistance (p = 0.0001). CisR-HeLa/-SiHa was more dependent on pGSK3ß, and activation of GSK3 by SMIs (iAkt), treatment with nimbolide, or knockdown of E6/E7 reduced cisplatin resistance and promoted apoptosis. Hence, the activation of GSK3ß with nimbolide and iAkt can be exploited for therapeutic interventions of cervical cancer.


Assuntos
Proteínas Oncogênicas Virais , Infecções por Papillomavirus , Neoplasias do Colo do Útero , Feminino , Humanos , Neoplasias do Colo do Útero/metabolismo , Infecções por Papillomavirus/tratamento farmacológico , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Cisplatino/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Papillomavirus Humano 18/metabolismo , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Oncogênicas Virais/genética , Proteínas Oncogênicas Virais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Resistência a Medicamentos , Linhagem Celular Tumoral
4.
Semin Cancer Biol ; 56: 25-36, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-29309927

RESUMO

Glycogen synthase kinase-3 (GSK-3), a serine/threonine kinase is an archetypal multifunctional moonlighting protein involved in diverse cellular processes including metabolism, insulin signaling, proliferation, differentiation, apoptosis, neuronal function and embryonic development. The two known isoforms, GSK-3α and GSK-3ß that undergo activation/inactivation by post-translational, site-specific phosphorylation incorporate a vast number of substrates in their repertoire. Dysregulation of GSK-3 has been linked to diverse disease entities including cancer. The role of GSK-3 in cancer is paradoxical and enigmatic. The enzyme functions as a tumour promoter or suppressor based on the context, cell type and phosphorylation status. GSK-3 is the central hub that orchestrates signals from the Wnt/ß-catenin, PI3K/PTEN/Akt/mTOR, Ras/Raf/MEK/ERK, hedgehog, Notch and TP53 pathways to elicit regulatory influences on cancer initiation, epithelial-mesenchymal transition, and resistance to therapy. As a direct target of several microRNAs, GSK-3 influences hallmark attributes of cancer, cancer stemness and treatment resistance. There is overwhelming evidence to indicate that GSK-3 is aberrantly regulated in different cancer types. Consequently, GSK-3 has emerged as a potential therapeutic target in cancer. A plethora of natural and synthetic GSK-3 modulators have been discovered and the number of patents published for GSK-3 inhibitors has also been steadily increasing in recent years. This review focuses on the intricate interactions between GSK-3 and oncogenic signalling circuits as well as the feasibility of targeting GSK-3 for the treatment of cancer.


Assuntos
Quinases da Glicogênio Sintase/genética , Quinases da Glicogênio Sintase/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos Fitogênicos , Biomarcadores Tumorais , Suscetibilidade a Doenças , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/química , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Quinases da Glicogênio Sintase/antagonistas & inibidores , Quinases da Glicogênio Sintase/química , Humanos , Isoenzimas , MicroRNAs/genética , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
6.
IUBMB Life ; 71(10): 1595-1610, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31251469

RESUMO

Aberrant activation of the PI3K/Akt signalling pathway, a major driving force of diverse cellular processes has been implicated in tumour development and progression. Here, we report that astaxanthin (AXT), a potent antioxidant ketocarotenoid prevents cancer hallmarks by inhibiting PI3K/Akt and the associated downstream NF-κB and STAT-3 signalling pathways in SCC131 and SCC4 oral cancer cells as well as in the hamster buccal pouch carcinogenesis model. Using small molecule inhibitors of NF-κB, STAT-3 and PI3K and by overexpression of PI3K, we provide evidence to show that AXT inhibits NF-κB and STAT-3 signalling and cancer hallmarks by restraining the kinase activity of PI3K/Akt. Additionally, AXT downregulated the noncoding RNAs (ncRNAs), miR-21 and HOTAIR that influence PI3K/Akt signalling emphasising its modulatory effects on epigenetic regulation. Ethyl cellulose-based AXT nanoparticles showed greater chemotherapeutic efficacy in the hamster oral carcinogenesis model compared to native AXT. We suggest that AXT prevents cell proliferation, apoptosis evasion, invasion and angiogenesis by intercepting the crosstalk between the PI3K/Akt, NF-κB and STAT-3 signalling circuits both in vitro and in vivo. Astaxanthin that abrogates the PI3K/Akt signalling axis, a central hub that orchestrates acquisition of cancer hallmarks is a promising candidate for anticancer drug development.


Assuntos
Carcinoma de Células Escamosas/tratamento farmacológico , Neoplasias Bucais/tratamento farmacológico , Neovascularização Patológica/tratamento farmacológico , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Epigênese Genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Bucais/genética , Neoplasias Bucais/patologia , NF-kappa B/genética , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/genética , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/genética , Transdução de Sinais/efeitos dos fármacos , Fator de Transcrição RelA/genética , Xantofilas/farmacologia
7.
J Toxicol Environ Health A ; 81(24): 1231-1245, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30507362

RESUMO

Substantial evidence has established the negative impact of inhalation exposure to welding fumes on respiratory functions. The aim of the present study was to investigate the effect of welding fume inhalation on expression of molecules that function as sensors, transducers and effectors of DNA damage response (DDR) in the respiratory tract of male Sprague-Dawley rats. Animals were exposed to 50 mg/m3 stainless steel welding fumes for 1 h/d for 4, 8, and 12 weeks, respectively. Histological examination demonstrated preneoplastic changes in trachea and bronchi with focal atelectasis and accumulation of chromium (Cr) in the lungs. This was associated with elevated levels of DNA damage markers (8-oxodG, γH2AX), ATM phosphorylation, cell cycle arrest, apoptosis induction, activation of homologous recombination (HR), non-homologous end joining (NHEJ), and Nrf2 signaling, as well as altered expression of noncoding RNAs (ncRNAs). However, after 12 weeks of exposure, DDR was compromised as reflected by resumption of the cell cycle, repair inhibition, and failure of apoptosis. Data demonstrate that exposure to welding fumes influences two crucial layers of DDR regulation, phosphorylation of key proteins in NHEJ and HR, as well as the ncRNAs that epigenetically modulate DDR. Evidence indicates that marked DNA damage coupled with non-productive DNA repair and apoptosis avoidance may be involved in neoplastic transformation.

8.
Clin Oral Investig ; 21(9): 2721-2731, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28233171

RESUMO

OBJECTIVES: Oral cancer is the sixth most common cancer in the world. Failure of chemoradiation therapy is a major concern for treating oral cancer patients. The objective of this study is to determine the B cell lymphoma-2 (bcl-2) expression and its regulation by nuclear factor κB (NFκB) and activator protein 1 (AP-1) in oral cancer progression and chemoradiation resistance. MATERIALS AND METHODS: In the present study, a total of 123 (n = 123) human samples were included. Briefly, 64 fresh samples were from adjacent normal (AN), primary oral tumors without treatment (PT), and tumors with resistance to chemoradiation therapy with local recurrence (RCRT). Fifty-nine samples were human tongue cancers and normal samples (TMA). Messenger RNA (mRNA) expression levels of bcl-2 and protein levels of bcl-2, NFκB, AP-1, and inactive GSK3α/ß were measured by semiquantitative RT-PCR, immunohistochemistry, Western blot, and ChIP analysis. RESULTS: Increased bcl-2 expression was observed in PT compared to AN. The RCRT tumors showed maximum expression of bcl-2 mRNA and protein over the PT and AN groups. Bcl-2 protein and mRNA expression were positively correlated with NFκB and AP-1 expression. AP-1 expression was strongly correlated with bcl-2 in the RCRT group of tumors. Further, inactive GSK3α/ß showed a positive trend with bcl-2 expression in oral tongue cancer specimens. CONCLUSION: Collectively, our results demonstrated cumulative effect of AP-1 and NFĸB for bcl-2 gene regulation in overall PT progression and chemoradiation resistance. The study provides evidence of increased bcl-2 mRNA/protein fueled by NFĸB in PT and AP-1 in RCRT. These regulations of bcl-2 by NFκB and AP-1 are important in OSCC progression and chemoradiation resistance.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Neoplasias da Língua/metabolismo , Neoplasias da Língua/patologia , Fator de Transcrição AP-1/metabolismo , Adulto , Idoso , Biomarcadores Tumorais/metabolismo , Western Blotting , Carcinoma de Células Escamosas/terapia , Quimiorradioterapia , Imunoprecipitação da Cromatina , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/terapia , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias da Língua/terapia , Regulação para Cima
9.
Tumour Biol ; 37(2): 2083-93, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26342697

RESUMO

Aberrant activation of oncogenic signaling pathways plays a central role in tumor development and progression. The aim of this present study was to investigate the chemopreventive effects of the neem limonoid gedunin in the hamster model of oral cancer based on its ability to modulate aldose reductase (AR), phosphatidyl inositol-3-kinase (PI3K)/Akt, and nuclear factor kappa B (NF-κB) pathways to block angiogenesis. Administration of gedunin suppressed the development of HBP carcinomas by inhibiting PI3K/Akt and NF-κB pathways through the inactivation of Akt and inhibitory kappa B kinase (IKK), respectively. Immunoblot and molecular docking interactions revealed that inhibition of these signaling pathways may be mediated via inactivation of AR by gedunin. Gedunin blocked angiogenesis by downregulating the expression of miR-21 and the pro-angiogenic factors vascular endothelial growth factor and hypoxia inducible factor-1 alpha (HIF-1α). In conclusion, the results of the present study provide compelling evidence that gedunin prevents progression of hamster buccal pouch (HBP) carcinomas via inhibition of the kinases Akt, IKK, and AR, and the oncogenic transcription factors NF-κB and HIF-1α to block angiogenesis.


Assuntos
Antineoplásicos/farmacologia , Limoninas/farmacologia , Neoplasias Bucais/metabolismo , Neovascularização Patológica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Carcinogênese/efeitos dos fármacos , Cricetinae , Modelos Animais de Doenças , Imunofluorescência , Immunoblotting , Masculino , Mesocricetus , Simulação de Acoplamento Molecular , Neoplasias Bucais/patologia , NF-kappa B/metabolismo , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Serina-Treonina Quinases TOR/metabolismo
10.
Tumour Biol ; 37(11): 15253-15264, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27696293

RESUMO

The reversion-inducing cysteine-rich protein with Kazal motifs (RECK) and glycogen synthase kinase (GSK3) are novel tumor suppressors, and emerging evidence has suggested their active role in oral cancer pathogenesis. In the present study, 112 human samples, including 55 fresh samples of 14 adjacent normal tissues, 25 noninvasive oral tumors, and 18 invasive tumors, were included. The messenger RNA (mRNA) expression, protein expression, and promoter methylation of the RECK gene, as well as the expression of GSK3ß, phospho/total ß-catenin, and c-myc, were measured by RT-PCR, bisulphate modification-PCR, immunohistochemistry, and Western blot analysis. Additionally, ectopic expression of in/active GSK3ß was performed in cell culture experiments. This study provided information on the progressive silencing of RECK gene expression at the protein and mRNA levels paralleled with promoter hypermethylation at various stages of oral tumor invasion. RECK expression and the hypermethylation of the RECK gene promoter were negatively and positively correlated with pS9GSK3ß/c-myc expression, respectively. Further, a negative trend of RECK protein expression with nuclear ß-catenin expression was observed. Induced expression of active GSK3ß reversed the RECK silencing in SCC9 cells. Collectively, our results demonstrated that the silencing of the RECK gene, possibly regulated by the GSK3ß pathway, is an important event in oral cancer invasion and this pathway could be exploited for therapeutic interventions.


Assuntos
Carcinoma de Células Escamosas/genética , Metilação de DNA , Proteínas Ligadas por GPI/genética , Regulação Neoplásica da Expressão Gênica , Glicogênio Sintase Quinase 3 beta/metabolismo , Neoplasias Bucais/genética , Adulto , Idoso , Biomarcadores Tumorais , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundário , Estudos de Casos e Controles , Feminino , Proteínas Ligadas por GPI/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Técnicas Imunoenzimáticas , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Estadiamento de Neoplasias , Prognóstico , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
11.
Mol Cancer ; 14: 20, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25645517

RESUMO

BACKGROUND: The study aims to evaluate the expression and activity of glycogen synthase kinase 3 isoforms α/ß (GSK3α/ß) and to assess their oncogenic potential through a correlation with the expression of cyclin D1 and p53 in oral cancer. METHODS: The expression of total and phosphorylated GSK3α/ß as well as cyclin D1 and p53 together with their interaction were assessed in human oral cancer tissue samples, apparently normal adjacent tissues, benign tumor samples, premalignant lesions and healthy normal tissues (total 179) using various methods, such as immunohistochemistry, Western blot assays, immunoprecipitation and RT-PCR analysis. RESULTS: The expression of GSK3ß was significantly higher relative to GSK3α indicating the greater role of the ß isoform in oral cancer. Among various types of oral cancers, OSCC (of the lip and tongue) showed elevated expression of GSK3α/ß, and the expression was correlated with disease progression. The increased expression of pS(21)GSK3α and pS(9)GSK3ß not only correlated positively with cyclin D1 and p53 expression in tongue cancer progression but a gradual shift of their expression from the cytoplasmic to the nuclear compartment and overall disease severity was also observed. The interaction of GSK3ß-cyclin D1 and the positive correlation of pS(9)GSK3ß and the transcription of cyclin D1 were observed. CONCLUSIONS: These results demonstrate that the inactivation of GSK3ß is an important event in OSCC and can be used as a marker for assessing disease severity and may be exploited for therapeutic intervention.


Assuntos
Carcinoma de Células Escamosas/genética , Ciclina D1/genética , Quinase 3 da Glicogênio Sintase/genética , Neoplasias Bucais/genética , Proteína Supressora de Tumor p53/genética , Adulto , Idoso , Carcinoma de Células Escamosas/patologia , Estudos de Casos e Controles , Núcleo Celular/genética , Citoplasma/genética , Progressão da Doença , Feminino , Glicogênio Sintase Quinase 3 beta , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Bucais/diagnóstico
12.
Nutr Cancer ; 67(2): 203-11, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25650669

RESUMO

Chlorophyllin (CHL), a water soluble semisynthetic derivative of the ubiquitous plant pigment chlorophyll used as a food additive, is recognized to confer a wide range of health benefits. CHL has been shown to exhibit potent antigenotoxic, anti-oxidant, and anticancer effects. Numerous experimental and epidemiological studies have demonstrated that dietary supple-mentation of CHL lowers the risk of cancer. CHL inhibits cancer initiation and progression by targeting multiple molecules and pathways involved in the metabolism of carcinogens, cell cycle progression, apoptosis evasion, invasion, and angiogenesis. The modulatory effects of CHL on the hallmark capabilities of cancer appear to be mediated via abrogation of key oncogenic signal transduction pathways such as nuclear factor kappa B, Wnt/ß-catenin, and phosphatidylinositol-3-kinase/Akt signaling. This review provides insights into the molecular mechanisms of the anticancer effects of dietary CHL.


Assuntos
Anticarcinógenos/farmacologia , Antimutagênicos/farmacologia , Clorofilídeos/farmacologia , Suplementos Nutricionais , Neoplasias/prevenção & controle , Animais , Apoptose/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos , Xenobióticos/metabolismo
13.
Tumour Biol ; 35(7): 6725-37, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24715303

RESUMO

Transforming growth factor (TGF) ß signaling pathway plays a central role in the regulation of a wide range of cellular processes involved in the acquisition of the malignant phenotype. The objective of the present study was to examine the effect of chlorophyllin, a semisynthetic derivative of chlorophyll on N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)--induced rat forestomach carcinogenesis based on the modulation of TGFß signaling and the downstream target genes associated with cell proliferation, apoptosis evasion, angiogenesis, invasion, and metastasis. We determined the effect of dietary chlorophyllin on TGFß signaling and the downstream events-cell proliferation, apoptosis evasion, angiogenesis, invasion, and metastasis by semiquantitative and quantitative reverse transcription (RT)-PCR, Western blot, and immunohistochemical analyses. We further validated the inhibition of TGFß signaling by chlorophyllin by performing molecular docking studies. We found that dietary supplementation of chlorophyllin at 4-mg/kg bw inhibits the development of MNNG-induced forestomach carcinomas by downregulating the expression of TGFß RI, TGFß RII, and Smad 2 and 4 and upregulating Smad 7, thereby abrogating canonical TGFß signaling. Docking interactions also confirmed the inhibition of TGFß signaling by chlorophyllin via inactivating TGFß RI. Furthermore, attenuation of TGFß signaling by chlorophyllin also blocked cell proliferation, angiogenesis, invasion, and metastasis, and induced mitochondria-mediated cell death. Dietary chlorophyllin that simultaneously abrogates TGFß signaling pathway and the key hallmark events of cancer appear to be an ideal candidate for cancer chemoprevention.


Assuntos
Carcinogênese/efeitos dos fármacos , Clorofilídeos/administração & dosagem , Neoplasias Gástricas/genética , Fator de Crescimento Transformador beta/genética , Animais , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Fator de Crescimento Transformador beta/metabolismo
14.
Oncol Res ; 32(8): 1287-1308, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39055885

RESUMO

Aldo-keto reductases (AKRs) are a superfamily of enzymes that play crucial roles in various cellular processes, including the metabolism of xenobiotics, steroids, and carbohydrates. A growing body of evidence has unveiled the involvement of AKRs in the development and progression of various cancers. AKRs are aberrantly expressed in a wide range of malignant tumors. Dysregulated expression of AKRs enables the acquisition of hallmark traits of cancer by activating oncogenic signaling pathways and contributing to chemoresistance. AKRs have emerged as promising oncotherapeutic targets given their pivotal role in cancer development and progression. Inhibition of aldose reductase (AR), either alone or in combination with chemotherapeutic drugs, has evolved as a pragmatic therapeutic option for cancer. Several classes of synthetic aldo-keto reductase (AKR) inhibitors have been developed as potential anticancer agents, some of which have shown promise in clinical trials. Many AKR inhibitors from natural sources also exhibit anticancer effects. Small molecule inhibitors targeting specific AKR isoforms have shown promise in preclinical studies. These inhibitors disrupt the activation of oncogenic signaling by modulating transcription factors and kinases and sensitizing cancer cells to chemotherapy. In this review, we discuss the physiological functions of human AKRs, the aberrant expression of AKRs in malignancies, the involvement of AKRs in the acquisition of cancer hallmarks, and the role of AKRs in oncogenic signaling, and drug resistance. Finally, the potential of aldose reductase inhibitors (ARIs) as anticancer drugs is summarized.


Assuntos
Aldo-Ceto Redutases , Antineoplásicos , Neoplasias , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Neoplasias/patologia , Aldo-Ceto Redutases/metabolismo , Aldo-Ceto Redutases/genética , Aldo-Ceto Redutases/antagonistas & inibidores , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia , Animais , Inibidores Enzimáticos/uso terapêutico , Inibidores Enzimáticos/farmacologia , Medicina de Precisão , Transdução de Sinais , Aldeído Redutase/antagonistas & inibidores , Aldeído Redutase/metabolismo
15.
Artigo em Inglês | MEDLINE | ID: mdl-38561609

RESUMO

BACKGROUND: Deregulated DNA damage response (DDR) network is implicated in cancer progression and therapy resistance. OBJECTIVE: The present study was designed to investigate whether nimbolide, an anticancer neem limonoid, targets key components of the DDR signalling pathway in cellular and animal models of oral squamous cell carcinoma (OSCC). METHODS: OSCC cells (SCC-4 and SCC-9), 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinoma model, chemoresistant OSCC patient-derived xenograft (PDX) model established in athymic nude mice, and tissue sections from patients with oral premalignant/malignant disease were used for the study. Key molecules that orchestrate the DDR, including the MRN complex, ATM, DNA-PKcs, H2AX, and p53, were analysed by qRT-PCR, immunoblotting, immunofluorescence, and immunohistochemistry. Cell proliferation and apoptosis indices were evaluated. RESULTS: Nimbolide significantly reduced 8-oxodG levels, expression of MRN, ATMS1891, and γ-H2AX, with an increase in p-p53S15 in OSCC cells as well as in the HBP model. Nimbolide potentiated the effect of KU-55933 in ATM inhibition. In the PDX model, nimbolide suppressed tumor formation, stimulated DDR and apoptosis, inhibited cell proliferation, and enhanced sensitivity to cisplatin. Analysis of p-ATM expression revealed a significant increase during the sequential progression of hamster and human OSCC. CONCLUSIONS: This study provides compelling evidence that nimbolide functions as a DDR inhibitor in cellular and hamster OSCC models and as a DDR activator in the PDX model primarily by targeting ATM. Small molecules like nimbolide that modulate DDR are of immense benefit in cancer therapy. The study has also unveiled p-ATM as a promising biomarker of tumour progression in human OSCCs.

16.
Eur J Nutr ; 52(1): 75-84, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22160170

RESUMO

PURPOSE: Constitutive activation of the Wnt signaling pathway and its downstream effectors plays a key role in neoplastic transformation. The objective of this study was to investigate the effect of ellagic acid, a plant-derived polyphenol on Wnt/ß-catenin signaling and its downstream circuits- NF-κB and mitochondrial apoptosis in the 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster buccal pouch (HBP) carcinogenesis model. METHODS: Hamsters were divided into six groups. The right buccal pouches of animals in groups 1-4 were painted with 0.5% DMBA three times a week for 14 weeks. Animals in groups 2-4 received in addition basal diet containing ellagic acid at a concentration of 0.1, 0.2, and 0.4% in the diet. Group 5 animals were given 0.4% ellagic acid alone. Group 6 animals served as control. The expression of the members of Wnt and NF-κB signaling and intrinsic apoptosis was evaluated by western blot analysis. RESULTS: Dietary supplementation of 0.4% ellagic acid suppressed the development of HBP carcinomas by preventing the constitutive activation of Wnt pathway through the downregulation of Fz, Dvl-2, GSK-3ß and nuclear translocation of ß-catenin. Abrogation of Wnt signaling by ellagic acid was also associated with inactivation of NF-κB and modulation of key components of the mitochondrial apoptotic network. CONCLUSIONS: Our findings suggest a functional crosstalk between Wnt and NF-κB signaling pathways in HBP carcinomas that is blocked by ellagic acid supplementation. Dietary ellagic acid that targets the Wnt/ß-catenin pathway as well as its downstream signaling mediators is a unique candidate for cancer chemoprevention.


Assuntos
Apoptose/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Ácido Elágico/farmacologia , Neoplasias Bucais/tratamento farmacológico , NF-kappa B/metabolismo , Via de Sinalização Wnt , 9,10-Dimetil-1,2-benzantraceno/metabolismo , Animais , Western Blotting , Bochecha/patologia , Cricetinae , Modelos Animais de Doenças , Regulação para Baixo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Masculino , Mucosa Bucal/patologia , Neoplasias Bucais/patologia , NF-kappa B/genética , Extratos Vegetais/farmacologia , beta Catenina/genética , beta Catenina/metabolismo
17.
J Lab Physicians ; 15(2): 237-242, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37323608

RESUMO

Objective The current study was undertaken to investigate the utility of total prostate-specific antigen (tPSA), its isoform [-2] proPSA (p2PSA), and prostate health index (PHI) in the diagnosis of metastatic prostate cancer (PCa). Materials and Methods This study was conducted from March 2016 to May 2019. Eighty-five subjects who were diagnosed with PCa for the first time, following transrectal ultrasound-guided prostate biopsy, were included in the study. The prebiopsy blood samples were analyzed in Beckman Coulter Access-2 Immunoanalyzer for tPSA, p2PSA, and free PSA (fPSA), and the calculated parameters included %p2PSA, %fPSA, and PHI. Mann-Whitney's U test was used as test of significance, and p -value less than 0.05 was considered statistically significant. Results Of the 85 participants, 81.2% ( n = 69) had evidence of metastasis, both clinically and pathologically. The median tPSA (ng/mL), p2PSA (pg/mL), %p2PSA, and PHI were significantly higher in the group with evidence of metastasis (46.5 vs. 13.76; 198.0 vs. 35.72; 3.25 vs. 1.51; 237.58 vs. 59.74, respectively). The sensitivity (%), specificity (%), negative predictive value (%), and positive predictive value (%) to diagnose metastatic PCa of tPSA at a cutoff of 20 ng/mL, PHI at a cutoff of 55, and %p2PSA at a cutoff of 1.66 were 92.7, 98.5, and 94.2; 37.5, 43.7, and 62.5; 54.5, 87.5, and 71.4; and 86.4, 88.3, and 91.5, respectively. Conclusion Using tests such as %p2PSA and PHI in the standard armamentarium for the diagnosis of metastatic PCa in addition to PSA will help in selecting the appropriate treatment strategy, including active surveillance.

18.
Arch Oral Biol ; 150: 105688, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36989865

RESUMO

OBJECTIVE: This study aimed to determine whether glucose transporter-1/3 (GLUT1/3) increased expression could contribute to oral tumor severity. Furthermore, this study detected whether GLUT1/3 mRNA/protein was associated with oncogenic transcription factors (HIF1α, AP1 and NFκB) and whether by blocking GLUT1 along with cisplatin could sensitize drug-resistant OSCC cells. DESIGN: We used 120 post-operated human tissue samples, including 35 primary tumors (PT), 43 invasive tumors (N1-3), 17 recurrent chemoradiation-resistant tumors (RCRT), and 25 PT-adjacent normal tissues (AN). The cisplatin-resistant (CisR-SCC4/9) cells were generated using a drug escalation strategy from parental SCC4/9 cells. The BAY-876 treatment blocked GLUT1 in OSCC cells. Western Blot, Immunohistochemistry, and reverse transcription polymerase chain reaction (RT-PCR) were used to detect various proteins and mRNA. Cell survival was determined by MTT assay. RESULTS: GLUT1/3 expression was observed more in PT over AN tissue (PT > AN), N1-3 > PT, and .RCRT > PT. GLUT1 expression was maximum in the RCRT group and CisR-SCC4/9 cells over their parental counterpart, linked with tumor size (p=0.0037) and loco-regional invasiveness (p=0.0422). GLUT1/3 mRNA/protein was correlated (positively) with oncogenic transcription factors (TFs) like HIF1α, AP1 and NFκB. We found the degree of positive correlation of these TFs with GLUT1/3 was in the order c-Jun > HIF1α > Fra-2 > NFκB > c-Fos. Treatment of BAY-876 and cisplatin-induced cell death in both CisR-SCC4/9 cells, possibly by triggering apoptosis and autophagy. CONCLUSION: Collectively, our results demonstrated increased GLUT1/3 overexpression linked with oral tumor severity like invasion and therapy resistance, and it was powered mainly by c-Jun (AP1). Blocking GLUT1 receptors and cisplatin application can sensitize CisR-OSCC cells.


Assuntos
Cisplatino , Neoplasias Bucais , Humanos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Resistencia a Medicamentos Antineoplásicos , Linhagem Celular Tumoral , Recidiva Local de Neoplasia , Neoplasias Bucais/patologia , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Regulação Neoplásica da Expressão Gênica
19.
Int Immunopharmacol ; 124(Pt A): 110894, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37678027

RESUMO

BACKGROUND: Programmed cell death ligand-1 (PD-L1)is an antitumor immunity molecule and a great target to cure oral cancer; nonetheless, the limited success can be attributed to many complex pathways and tumor-related interferences. METHODS: In the present study, 150 human oral squamous cell carcinoma (OSCC) tissue samples, including 17 adjacent normals, 56 primary tumors, 47 invasive tumors, and 30 therapy-resistant (RT) samples, were included. The parental/cisplatin-resistant (CisR-SCC4/9) cells were utilized for overexpression (Jak1-3 wild type and catalytically inactive), knockdown (PD-L1 siRNA), targeting MAPK/PI3K/Jak-Stat pathways (SMIs) and checking microsomes. The expression of PD-L1, transcription factors (TFs), signaling pathways, survival/apoptosis, therapy resistance, and invasiveness-related molecules/their activity were determined by RT-PCR, Immunohistochemistry, Western blot, Gelatin Zymography, and MTT assay. RESULTS: Advanced OSCC tumors (invasive and drug-resistance), CisR-SCC4/9 cells, and secretory exosomes (CisR-SCC4/9) were found with increased PD-L1 expression. PD-L1 mRNA/protein showed a positive correlation with different TFs (AP1 > Stat3 > c-myc > NFκB) in tumor samples. The PD-L1 expression was more influenced by Jak-Stat/ MAPK-AP1 pathways over PI3K. The ectopic expression of Jak1-3 suggests Jak2 inducted PD-L1 level over Jak1/Jak3. Finally, PD-L1 directly supports survival (Bcl-xL, Bax, cleaved caspase-3), invasion (MMP2/9), and drug-resistance (ALDH-1A1/-3A1) program in OSCC through its link with several molecules. CONCLUSIONS: PD-L1 was regulated mainly by the Jak2-Stat3/ MAPK-AP1 pathway, and besides the routine immunological functions, it supports OSCC survival, invasion, and therapy resistance. PD-L1 can be used as an indicator of severity and can be targeted along with Jak2-Stat3/ MAPK-AP1 for a better outcome OSCC.

20.
Anticancer Agents Med Chem ; 22(14): 2619-2636, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35125086

RESUMO

BACKGROUND & OBJECTIVE: The insulin/IGF-1R/PI3K/Akt signalling cascade is increasingly being linked to breast cancer development, with aldose reductase (AR) playing a key role in mediating the crosstalk between this pathway and angiogenesis. The current study was designed to investigate whether nimbolide, a neem limonoid, targets the oncogenic signaling network to prevent angiogenesis in breast cancer. METHODS: Breast cancer cells (MCF-7, MDA-MB-231), EAhy926 endothelial cells, MDA-MB-231 xenografted nude mice, and tumour tissues from breast cancer patients were used for the study. The expression of AR and key players in IGF-1/PI3K/Akt signaling and angiogenesis was evaluated by qRT-PCR, immunoblotting, and immunohistochemistry. Molecular docking and simulation, overexpression, and knockdown experiments were performed to determine whether nimbolide targets AR and IGF-1R. RESULTS: Nimbolide inhibited AR with consequent blockade of the IGF-1/PI3K/Akt and /HIF-1alpha/VEGF signalling circuit by influencing the phosphorylation and intracellular localisation of key signaling molecules. The downregulation of DNMT-1, HDAC-6, miR-21, HOTAIR, and H19 with the upregulation of miR-148a/miR-152 indicated that nimbolide regulates AR and IGF-1/PI3K/Akt signaling via epigenetic modifications. Coadministration of nimbolide with metformin and the chemotherapeutic drugs tamoxifen/cisplatin displayed higher efficacy than single agents in inhibiting IGF-1/PI3K/Akt/AR signaling. Grade-wise increases in IGF-1R and AR expression in breast cancer tissues underscore their value as biomarkers of progression. CONCLUSION: This study provides evidence for the anticancer effects of nimbolide in cellular and mouse models of breast cancer besides providing leads for new drug combinations. It has also opened up avenues for investigating potential molecules such as AR for therapeutic targeting of cancer.


Assuntos
Azadirachta , Neoplasias da Mama , Limoninas , MicroRNAs , Aldeído Redutase , Animais , Azadirachta/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Células Endoteliais , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Limoninas/farmacologia , Camundongos , Camundongos Nus , MicroRNAs/metabolismo , Simulação de Acoplamento Molecular , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA