Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Phytother Res ; 37(4): 1539-1573, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36788644

RESUMO

Emerging evidence on molecular biology related to tumors, inflammation, and immunity, highlights their architectural commonality shifting cancer treatment paradigms toward more economical prevention than treatment. Statistical surveys reveal exponentially growing herbal drug supplementation in cancer worldwide as vast pre-clinical and clinical data unravel their multi-mechanistic pharmacology. The integrative oncological approach calls for more "holistic" principles to be amalgamated into cancer care. New cancer drug development from herbs need not be limited by the archetypal 'RCT-Standardization' bottlenecks. Based on comprehensive literature scoping as per Prisma-ScR guidelines, we herein concurrently reviewed evidence-based research reports of selected Indian Traditional Medicine (ITM) herbs of anticancer repute in parallel with their holistic therapeutics; a rationalistic exploration of ITM's scientific genre. Their synergy effect on cancer revisited using a trans-pharmacological approach validates ITM's seemingly simplistic health/disease equation model, showing a fresh new avenue for re-purposing whole herbal drug complexes in cancer management. Herbal drugs as per ITM are natural matrices whose dynamics of interaction in the etiopathology of cancer are conceptually and mechanistically integrative. Lateral perspective to the same as laid out in this review holds the key to their effectual development as more tangible cancer chemopreventives/new drug targets/leads if not as new pharmacological tools.


Assuntos
Antineoplásicos , Produtos Biológicos , Medicamentos de Ervas Chinesas , Neoplasias , Humanos , Produtos Biológicos/uso terapêutico , Neoplasias/tratamento farmacológico , Fitoterapia , Medicina Tradicional , Medicamentos de Ervas Chinesas/farmacologia , Antineoplásicos/uso terapêutico
2.
Cell Signal ; 101: 110496, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36252791

RESUMO

Nitric Oxide (NO) is a highly diffusible, ubiquitous signaling molecule and a free radical that is naturally synthesized by our body. The pleiotropic effects of NO in biological systems are due to its reactivity with different molecules, such as molecular oxygen (O2), superoxide anion, DNA, lipids, and proteins. There are several contradictory findings in the literature pertaining to its role in oncology. NO is a Janus-faced molecule shown to have both tumor promoting and tumoricidal effects, which depend on its concentration, duration of exposure, and location. A high concentration is shown to have cytotoxic effects by triggering apoptosis, and at a low concentration, NO promotes angiogenesis, metastasis, and tumor progression. Upregulated NO synthesis has been implicated as a causal factor in several pathophysiological conditions including cancer. This dichotomous effect makes it highly challenging to discover its true potential in cancer biology. Understanding the mechanisms by which NO acts in different cancers helps to develop NO based therapeutic strategies for cancer treatment. This review addresses the physiological role of this molecule, with a focus on its bimodal action in various types of cancers.


Assuntos
Neoplasias , Óxido Nítrico , Humanos , Óxido Nítrico/metabolismo , Neoplasias/patologia , Transdução de Sinais , Apoptose , Superóxidos/metabolismo
3.
Adv Protein Chem Struct Biol ; 125: 149-191, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33931138

RESUMO

The development of cancer is a complex phenomenon driven by various extrinsic as well as intrinsic risk factors including epigenetic modifications. These post-translational modifications are encountered in diverse cancer cells and appear for a relatively short span of time. These changes can significantly affect various oncogenic genes and proteins involved in cancer initiation and progression. Histone lysine acetylation and deacetylation processes are controlled by two opposing classes of enzymes that modulate gene regulation either by adding an acetyl moiety on a histone lysine residue by histone lysine acetyltransferases (KATs) or via removing it by histone deacetylases (KDACs). Deregulated KAT activity has been implicated in the development of several diseases including cancer and can be targeted for the development of anti-neoplastic drugs. Here, we describe the predominant epigenetic changes that can affect key KAT superfamily members during carcinogenesis and briefly highlight the pharmacological potential of employing lysine acetyltransferase inhibitors (KATi) for cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores Enzimáticos/uso terapêutico , Histona Acetiltransferases , Proteínas de Neoplasias , Neoplasias , Histona Acetiltransferases/antagonistas & inibidores , Histona Acetiltransferases/metabolismo , Humanos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia
4.
Int J Biochem Cell Biol ; 108: 17-20, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30630112

RESUMO

Long non-coding RNAs (lncRNAs) are a novel class of gene regulators playing multifaceted roles in physiological processes as well as pathological conditions such as cancer. Cancer stem cells (CSCs) are a small subset of tumor cells that constitute the origin and development of various malignant tumors. CSCs have been identified in a wide spectrum of human tumors and could act as a critical link underlying the processes of tumor metastasis and recurrence. Mounting evidence indicates that lncRNAs are aberrantly expressed in diverse CSCs and regulate CSC properties at different molecular levels. Here, we very briefly summarize the recent findings on the potential roles of lncRNAs in regulating various functions of CSCs, and elaborate on how can lncRNAs impact CSC properties via interacting with other macromolecules at the epigenetic, transcriptional, and post-transcriptional levels. This mini-review also highlights the understanding of the modular regulatory principles of lncRNA interactions in CSCs.


Assuntos
Neoplasias/genética , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , RNA Longo não Codificante/genética , Epigênese Genética/genética , Humanos , Transcrição Gênica/genética
5.
Int J Mol Sci ; 19(10)2018 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-30274259

RESUMO

The tumor necrosis factor-α-induced protein 8-like (TIPE/TNFAIP8) family is a recently identified family of proteins that is strongly associated with the regulation of immunity and tumorigenesis. This family is comprised of four members, namely, tumor necrosis factor-α-induced protein 8 (TIPE/TNFAIP8), tumor necrosis factor-α-induced protein 8-like 1 (TIPE1/TNFAIP8L1), tumor necrosis factor-α-induced protein 8-like 2 (TIPE2/TNFAIP8L2), and tumor necrosis factor-α-induced protein 8-like 3 (TIPE3/TNFAIP8L3). Although the proteins of this family were initially described as regulators of tumorigenesis, inflammation, and cell death, they are also found to be involved in the regulation of autophagy and the transfer of lipid secondary messengers, besides contributing to immune function and homeostasis. Interestingly, despite the existence of a significant sequence homology among the four members of this family, they are involved in different biological activities and also exhibit remarkable variability of expression. Furthermore, this family of proteins is highly deregulated in different human cancers and various chronic diseases. This review summarizes the vivid role of the TIPE family of proteins and its association with various signaling cascades in diverse chronic diseases.


Assuntos
Doença Crônica , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias/metabolismo
6.
Reproduction ; 155(1): 61-71, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29066531

RESUMO

Recent studies showed that genetic aberrations in the MED12 gene, probably through the canonical WNT/ß-catenin pathway, lead to the pathogenesis of uterine fibroids. However, a comprehensive analysis of the WNT pathway in MED12-mutated and MED12-wild-type fibroids has not been performed. The objective of this study was to determine the status of the WNT pathway in human fibroids. We performed Sanger sequencing to define the MED12 mutational status of fibroids and normal myometrium samples. qPCR arrays were carried out to determine the status of the WNT signaling pathway in MED12-mutated and MED12-wild-type fibroids. Liquid chromatography-mass spectrometry (LC-MS), Western blotting and immunohistochemistry were used to monitor the expression of ß-catenin. We showed that ß-catenin expression was increased in fibroids compared to the adjacent myometrium samples. However, ß-catenin expression showed no correlation with MED12 mutation status. Of all the WNT signaling components, WNT inhibitors showed the greatest differences in expression between fibroids and controls. WIF1, a WNT inhibitor, was identified as the most significantly upregulated gene in fibroids. We cultured primary fibroid cells on hydrogels of known stiffness to decipher the influence of biomechanical cues on ß-catenin expression and revealed increased levels of ß-catenin when cells were cultured on a stiffer surface. In conclusion, our data showed that ß-catenin expression in fibroids occurs independently of MED12 mutations. Biomechanical changes upregulate ß-catenin expression in fibroids, providing an attractive avenue for developing new treatments for this disease.


Assuntos
Matriz Extracelular/metabolismo , Leiomioma/patologia , Mutação , Miométrio/patologia , Neoplasias Uterinas/patologia , beta Catenina/metabolismo , Células Cultivadas , Feminino , Humanos , Leiomioma/genética , Leiomioma/metabolismo , Complexo Mediador/genética , Miométrio/metabolismo , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo , Via de Sinalização Wnt , beta Catenina/genética
7.
Oncotarget ; 8(8): 12831-12842, 2017 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-28086233

RESUMO

A prior screening programme carried out using MTT assay by our group identified a series of novel benzimidazole derivatives, among which Methyl 2-(5-fluoro-2-hydroxyphenyl)-1H- benzo[d]imidazole-5-carboxylate (MBIC) showed highest anticancer efficacy compared to that of chemotherapeutic agent, cisplatin. In the present study, we found that MBIC inhibited cell viability in different hepatocellular carcinoma (HCC) cell lines without exerting significant cytotoxic effects on normal liver cells. Annexin V-FITC/PI flow cytometry analysis and Western blotting results indicated that MBIC can induce apoptosis in HCC cells, which was found to be mediated through mitochondria associated proteins ultimately leading to the activation of caspase-3. The exposure to MBIC also resulted in remarkable impairment of HCC cell migration and invasion. In addition, treatment with MBIC led to a rapid generation of reactive oxygen species (ROS) and substantial activation of c-Jun-N-terminal kinase (JNK). The depletion of ROS by N-Acetyl cysteine (NAC) partially blocked MBIC-induced apoptosis and JNK activation in HCC cells. Finally, MBIC significantly inhibited tumor growth at a dose of 25 mg/kg in an orthotopic HCC mouse model. Taken together, these results demonstrate that MBIC may inhibit cell proliferation via ROS-mediated activation of the JNK signaling cascade in HCC cells.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Carcinoma Hepatocelular/metabolismo , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/metabolismo , Camundongos , Camundongos Nus , Espécies Reativas de Oxigênio , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Target Oncol ; 12(1): 1-10, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27510230

RESUMO

Hepatocellular carcinoma (HCC) is one of the most common forms of liver cancer diagnosed worldwide. HCC occurs due to chronic liver disease and is often diagnosed at advanced stages. Chemotherapeutic agents such as doxorubicin are currently used as first-line agents for HCC therapy, but these are non-selective cytotoxic molecules with significant side effects. Sorafenib, a multi-targeted tyrosine kinase inhibitor, is the only approved targeted drug for HCC patients. However, due to adverse side effects and limited efficacy, there is a need for the identification of novel pharmacological drugs beyond sorafenib. Several agents that target and inhibit various signaling pathways involved in HCC are currently being assessed for HCC treatment. In the present review article, we summarize the diverse signal transduction pathways responsible for initiation as well as progression of HCC and also the potential anticancer effects of selected targeted therapies that can be employed for HCC therapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Carcinogênese , Carcinoma Hepatocelular/patologia , Humanos , Neoplasias Hepáticas/patologia , Transdução de Sinais
9.
Semin Cancer Biol ; 40-41: 35-47, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27038646

RESUMO

The association between chronic inflammation and cancer development has been well documented. One of the major obstacles in cancer treatment is the persistent autocrine and paracrine activation of pro-inflammatory transcription factors such as nuclear factor-κB, signal transducer and activator of transcription 3, activator protein 1, fork head box protein M1, and hypoxia-inducible factor 1α in a wide variety of tumor cell lines and patient specimens. This, in turn, leads to an accelerated production of cellular adhesion molecules, inflammatory cytokines, chemokines, anti-apoptotic molecules, and inducible nitric oxide synthase. Numerous medicinal plant-derived compounds have made a tremendous impact in drug discovery research endeavors, and have been reported to modulate the activation of diverse oncogenic transcription factors in various tumor models. Moreover, novel therapeutic combinations of standard chemotherapeutic drugs with these agents have significantly improved patient survival by making cancer cells more susceptible to chemotherapy and radiotherapy. In this review, we critically analyze the existing literature on the modulation of diverse transcription factors by various natural compounds and provide views on new directions for accelerating the discovery of novel drug candidates derived from Mother Nature.


Assuntos
Anticarcinógenos/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Neoplasias/prevenção & controle , Fatores de Transcrição/metabolismo , Animais , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Carcinogênese/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Terapia de Alvo Molecular , Neoplasias/tratamento farmacológico , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
10.
Pharmacol Ther ; 162: 86-97, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26478441

RESUMO

Signal Transducers and Activators of Transcription (STATs) comprise an important class of transcription factors that have been implicated in a wide variety of essential cellular functions related to proliferation, survival, and angiogenesis. Among various STAT members, STAT3 is frequently overexpressed in tumor cells as well as tissue samples, and regulates the expression of numerous oncogenic genes controlling the growth and metastasis of tumor cells. The current review briefly discusses the importance of STAT3 as a potential target for cancer therapy and also provides novel insights into various classes of existing pharmacological inhibitors of this transcription factor that can be potentially developed as anti-cancer drugs.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/prevenção & controle , Fator de Transcrição STAT3/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinogênese/metabolismo , Humanos , Neoplasias/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
11.
World J Gastroenterol ; 21(43): 12261-73, 2015 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-26604635

RESUMO

Frequent activation of phosphatidylinositol-3 kinases (PI3K)/Akt/mTOR signaling pathway in gastric cancer (GC) is gaining immense popularity with identification of mutations and/or amplifications of PIK3CA gene or loss of function of PTEN, a tumor suppressor protein, to name a few; both playing a crucial role in regulating this pathway. These aberrations result in dysregulation of this pathway eventually leading to gastric oncogenesis, hence, there is a need for targeted therapy for more effective anticancer treatment. Several inhibitors are currently in either preclinical or clinical stages for treatment of solid tumors like GC. With so many inhibitors under development, further studies on predictive biomarkers are needed to measure the specificity of any therapeutic intervention. Herein, we review the common dysregulation of PI3K/Akt/mTOR pathway in GC and the various types of single or dual pathway inhibitors under development that might have a superior role in GC treatment. We also summarize the recent developments in identification of predictive biomarkers and propose use of predictive biomarkers to facilitate more personalized cancer therapy with effective PI3K/Akt/mTOR pathway inhibition.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/antagonistas & inibidores , Carcinoma/tratamento farmacológico , Descoberta de Drogas/métodos , Terapia de Alvo Molecular , Inibidores de Fosfoinositídeo-3 Quinase , Medicina de Precisão , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Neoplasias Gástricas/tratamento farmacológico , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma/enzimologia , Carcinoma/genética , Carcinoma/patologia , Humanos , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Resultado do Tratamento
12.
Oncotarget ; 6(41): 43806-18, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26517526

RESUMO

Chromatin acetylation is attributed with distinct functional relevance with respect to gene expression in normal and diseased conditions thereby leading to a topical interest in the concept of epigenetic modulators and therapy. We report here the identification and characterization of the acetylation inhibitory potential of an important dietary flavonoid, luteolin. Luteolin was found to inhibit p300 acetyltransferase with competitive binding to the acetyl CoA binding site. Luteolin treatment in a xenografted tumor model of head and neck squamous cell carcinoma (HNSCC), led to a dramatic reduction in tumor growth within 4 weeks corresponding to a decrease in histone acetylation. Cells treated with luteolin exhibit cell cycle arrest and decreased cell migration. Luteolin treatment led to an alteration in gene expression and miRNA profile including up-regulation of p53 induced miR-195/215, let7C; potentially translating into a tumor suppressor function. It also led to down-regulation of oncomiRNAs such as miR-135a, thereby reflecting global changes in the microRNA network. Furthermore, a direct correlation between the inhibition of histone acetylation and gene expression was established using chromatin immunoprecipitation on promoters of differentially expressed genes. A network of dysregulated genes and miRNAs was mapped along with the gene ontology categories, and the effects of luteolin were observed to be potentially at multiple levels: at the level of gene expression, miRNA expression and miRNA processing.


Assuntos
Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/patologia , Neoplasias de Cabeça e Pescoço/patologia , Luteolina/farmacologia , Fatores de Transcrição de p300-CBP/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Citometria de Fluxo , Humanos , Immunoblotting , Imuno-Histoquímica , Lisina/metabolismo , Camundongos , Simulação de Acoplamento Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Transcriptoma/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Mol Cell Endocrinol ; 401: 98-104, 2015 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-25462587

RESUMO

Pigment epithelium-derived factor (PEDF) is an anti-angiogenic, immunomodulatory, and neurotrophic serine protease inhibitor protein. PEDF is evolving as a novel metabolic regulatory protein that plays a causal role in insulin resistance. Insulin resistance is the central pathogenesis of metabolic disorders such as obesity, type 2 diabetes mellitus, polycystic ovarian disease, and metabolic syndrome, and PEDF is associated with them. The current evidence suggests that PEDF administration to animals induces insulin resistance, whereas neutralisation improves insulin sensitivity. Inflammation, lipolytic free fatty acid mobilisation, and mitochondrial dysfunction are the proposed mechanism of PEDF-mediated insulin resistance. This review summarises the probable mechanisms adopted by PEDF to induce insulin resistance, and identifies PEDF as a potential therapeutic target in ameliorating insulin resistance.


Assuntos
Proteínas do Olho/metabolismo , Resistência à Insulina , Mitocôndrias/metabolismo , Fatores de Crescimento Neural/metabolismo , Serpinas/metabolismo , Animais , Humanos , Metabolismo dos Lipídeos , Síndrome Metabólica/etiologia , Mitocôndrias/patologia
14.
Oncol Res ; 21(2): 93-102, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24406045

RESUMO

Malignant gliomas have a highly tumorigenic subpopulation, termed cancer stem cells (CSCs), that drives tumor formation and proliferation. CSCs possess inherent resistance mechanisms against radiation- and chemotherapy-induced cancer cell death, enabling them to survive and initiate tumor recurrence. We examined the effect of secreted frizzled-related protein 4 (sFRP4), a Wnt signaling antagonist, in chemosensitizing the glioma cell line U138MG and glioma stem cells (GSCs) enriched from U138MG to chemotherapeutics. We found that sFRP4 alone and in combination with either doxorubicin or cisplatin induced apoptosis. Proliferation decreased substantially in GSC-enriched population as measured by MTT and BrdU assays. JC-1 and caspase-3 assays demonstrated that cell death was through the apoptotic pathway. sFRP4 treatment also decreased neurosphere formation and induced neuronal differentiation. Inhibition by sFRP4 was abolished by Wnt3a, indicating that sFRP4 acts through the frizzled receptor. Further indication that sFRP4 acts through the Wnt ß-catenin pathway was provided by decrease in the ß-catenin protein and decrease in the ß-catenin-stimulated gene cyclin D1 upon sFRP4 induction. By real-time PCR, an increase in apoptotic markers Bax and p21, a decrease in pro-proliferative marker CycD1, and a decrease in the GSC marker CD133 were observed. These observations indicate that sFRP4 is able to sensitize glioma cells and stem cells to chemotherapeutics. We thus identified for the first time that sFRP4 could help to destroy cancer stem cells of glioma cell line, which would lead to effective treatment regimen to combat brain tumors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cisplatino/farmacologia , Doxorrubicina/farmacologia , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Doxorrubicina/administração & dosagem , Sinergismo Farmacológico , Glioblastoma/patologia , Humanos , Células-Tronco Neoplásicas/patologia , Proteínas Proto-Oncogênicas/administração & dosagem , Esferoides Celulares , Proteína Wnt3A/farmacologia
15.
Oncol Res ; 20(1): 1-6, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23035359

RESUMO

Secreted frizzled-related protein 4 (sFRP4) is a Wnt signaling antagonist. Classically, sFRP4 antagonizes the canonical Wnt signaling pathway, resulting in decreased cellular proliferation and increased apoptosis. Recent research from our laboratory has established that sFRP4 inhibits angiogenesis by decreasing proliferation, migration, and tube formation of endothelial cells. The objective of this study was to examine the role of sFRP4's cysteine-rich domain (CRD) and netrin-like domain (NLD) in angiogenesis inhibition. Experiments were carried out to examine cell death and tube formation of endothelial cells after treatment with the CRD and the NLD. The CRD was seen to inhibit tube formation of endothelial cells, which suggests that this domain is important to sFRP4's antiangiogenesis property. In addition, the NLD promoted endothelial cell death and may also inhibit angiogenesis. Furthermore, treatment with the CRD and the NLD increased endothelial intracellular calcium levels. Our findings implicate a role for both the CRD and NLD in angiogenesis inhibition by sFRP4. It is suggestive of alternative antiangiogenic downstream targets of canonical Wnt signaling and a possible importance of the noncanonical Ca2+ Wnt signaling pathway in sFRP4-mediated angiogenesis inhibition.


Assuntos
Inibidores da Angiogênese/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos , Motivos de Aminoácidos/fisiologia , Apoptose/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cisteína/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Estrutura Terciária de Proteína/fisiologia
16.
BMC Cell Biol ; 13: 25, 2012 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-23039795

RESUMO

BACKGROUND: Ovarian cancer is one of the most lethal malignancies in women, as it is frequently detected at an advanced stage, and cancers often become refractory to chemotherapy. Evidence suggests that dysregulation of pro-apoptotic genes plays a key role in the onset of chemoresistance. The secreted Frizzled-Related Protein (sFRP) family is pro-apoptotic and also a negative modulator of the Wnt signalling cascade. Studies have demonstrated that the re-expression of sFRPs, in particular sFRP4, is associated with a better prognosis, and that experimentally induced expression results in cell death. RESULTS: In vitro experimental models determined that sFRP4 was differentially expressed in chemosensitive (A2780) and chemoresistant (A2780 ADR and A2780 Cis) ovarian cell lines, with chemosensitive cells expressing significantly higher levels of sFRP4. Transfection of the chemoresistant cell lines with sFRP4 significantly increased their sensitivity to chemotherapy. Conversely, silencing of sFRP4 expression in the chemosensitive cell line resulted in a corresponding increase in chemoresistance. Comparison of sFRP4 expression in tumour biopsies revealed a positive trend between sFRP4 expression and tumour grade, with mucinous cyst adenocarcinomas exhibiting significantly decreased sFRP4 levels compared to mucinous borderline tumours. CONCLUSIONS: This study indicates a role for sFRP4 as a predictive marker of chemosensitivity in ovarian cancer and suggests that this pathway may be worth exploiting for novel therapies.


Assuntos
Antineoplásicos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/toxicidade , Proteínas Proto-Oncogênicas/metabolismo , Adenocarcinoma Mucinoso/metabolismo , Adenocarcinoma Mucinoso/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Estadiamento de Neoplasias , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Transfecção , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
17.
Mol Cell Endocrinol ; 362(1-2): 48-59, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22652149

RESUMO

Sphingolipid mediators such as ceramide are pleiotropic regulators of cellular growth, differentiation and apoptosis. We investigated the role of ceramide biosynthesis, metabolism and actions in term human cytotrophoblasts syncytialized over 7 days in culture. Intracellular C16 ceramide levels increased modestly after 3 days in culture, then declined. Ceramidase was present at particularly high levels in syncytialized trophoblasts; inhibition of ceramidase reduced the degree of cell fusion. Exposure to short chain C8 ceramide or aSMase enhanced secretion of the differentiation marker hCG without affecting fusion or cell viability. In contrast, pharmacological inhibition of ceramidase reduced the extent of fusion. Inhibition of the ceramide-responsive JNK and PP2A pathways did not abolish the effects of ceramide, and JNK phosphorylation was unresponsive to ceramide; however, ceramide significantly inhibited phosphorylation of Akt. This study suggests that changes in ceramide biosynthesis and metabolism play a differential role in the biochemical and morphological features of trophoblast differentiation.


Assuntos
Diferenciação Celular , Ceramidas/biossíntese , Células Gigantes/fisiologia , Trofoblastos/fisiologia , Antracenos/farmacologia , Antígenos de Diferenciação/metabolismo , Caspase 8/metabolismo , Fusão Celular , Células Cultivadas , Ceramidases/metabolismo , Ceramidas/metabolismo , Ceramidas/fisiologia , Gonadotropina Coriônica/metabolismo , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Feminino , Expressão Gênica , Células Gigantes/enzimologia , Células Gigantes/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ácido Okadáico/farmacologia , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Piranos/farmacologia , Esfingomielina Fosfodiesterase/metabolismo , Compostos de Espiro/farmacologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Trofoblastos/enzimologia , Trofoblastos/metabolismo
18.
Reprod Biomed Online ; 24(2): 224-34, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22197131

RESUMO

Sphingosine and sphingosine-1-phosphate (S1P) are involved in regulating cell differentiation. This study postulated that changes in sphingolipid biosynthesis and metabolism are important in trophoblast syncytialization and therefore examined the production, metabolism and actions of sphingosine and S1P during spontaneous trophoblast differentiation and fusion in vitro. Significant declines in intracellular sphingosine concentration (P≤0.05) and sphingosine kinase 1 (SPHK1) expression (P≤0.01) were observed during trophoblast syncytialization. Secreted S1P concentrations dropped steeply after 72h, before rising to basal concentrations with syncytialization. Intracellular S1P concentrations were undetectable throughout. Treating cells with exogenous sphingosine (P≤0.01), S1P (P≤0.001) or a specific SPHK1 inhibitor (P≤0.05) for up to 72h in culture significantly inhibited trophoblast differentiation (measured as reduced human chorionic gonadotrophin production); effects on other biochemical and morphological markers of differentiation were absent or inconsistent. Phosphorylation of Akt, an established down-stream target of S1P that spontaneously declines with trophoblast differentiation, was markedly reduced by S1P (P≤0.05). In conclusion, changes in the sphingosine-S1P pathway are involved in the regulation of trophoblast differentiation in term human placenta. Dysregulation of sphingolipid homeostasis could, therefore, disrupt placental formation and function with deleterious consequences for pregnancy outcome.


Assuntos
Lisofosfolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Trofoblastos/fisiologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Fosfotransferases (Aceptor do Grupo Álcool)/biossíntese , Gravidez , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esfingosina/farmacologia , Trofoblastos/efeitos dos fármacos , Trofoblastos/metabolismo
19.
Toxicol Sci ; 111(1): 49-63, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19520675

RESUMO

2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a highly toxic pollutant ubiquitously present in the environment. Most of the toxic effects of TCDD are believed to be mediated by high-affinity binding to the aryl hydrocarbon receptor (AhR) and subsequent effects on gene transcription. TCDD causes cancer in multiple tissues in different animal species and is classified as a class 1 human carcinogen. In initiation-promotion studies TCDD was shown to be a potent liver tumor promotor. Among other theories it has been hypothesized that TCDD acts as a tumor promotor by preventing initiated cells from undergoing apoptosis. We examined the effects of TCDD on ultraviolet C (UV-C) light-induced apoptosis in primary rat hepatocytes and Huh-7 human hepatoma cells. TCDD inhibits UV-C light-induced apoptosis in both cell types. This effect is seen with chromatin condensation and fragmentation and appears to be mediated by the AhR in rat hepatocytes. Apoptosis induced by UV-C light in these cells is caspase-dependent and is accompanied by alterations in apoptosis-related gene expression such as up-regulation of proapoptotic bcl-2 family genes like bak and bax, and a marked down regulation of the expression of the antiapoptotic bcl-2. TCDD treatment of irradiated hepatocytes induces the expression of some apoptosis-related genes (birc3, dad1, pycard, tnf). Upstream apoptotic events, namely caspase activation and caspase substrate cleavage are not inhibited by TCDD treatment. We hypothesize that TCDD inhibits late-stage apoptotic events that lead to internucleosomal DNA fragmentation, maintaining chromosomal integrity probably in order to sustain metabolic capacity and hepatic elimination of substrates despite of an initiation of apoptosis.


Assuntos
Apoptose/efeitos da radiação , Poluentes Ambientais/farmacologia , Fígado/citologia , Dibenzodioxinas Policloradas/farmacologia , Raios Ultravioleta , Animais , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cromatina/efeitos dos fármacos , Cromatina/efeitos da radiação , Corantes , Citocromo P-450 CYP1A1/biossíntese , Fragmentação do DNA , Desoxirribonucleases/metabolismo , Endodesoxirribonucleases/metabolismo , Hepatócitos/efeitos dos fármacos , Hepatócitos/efeitos da radiação , Neoplasias Hepáticas/patologia , Masculino , Poli(ADP-Ribose) Polimerases/metabolismo , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Biochem Biophys Res Commun ; 377(2): 606-611, 2008 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-18938133

RESUMO

The skin provides vital protection from infection and dehydration. Maintenance of the skin is through a constant program of proliferation, differentiation and apoptosis of epidermal cells, whereby proliferating cells in the basal layer differentiating to form the keratinized, anucleated stratum corneum. The WNT signalling pathway is known to be important in the skin. WNT signalling has been shown to be important both in epidermal development and in the maintenance and cycling of hair follicles and epidermal stem cells. However, the precise role for this pathway in epidermal differentiation remains unknown. We investigated the role of the WNT signalling inhibitor sFRP4 in epidermal differentiation. sFRP4 is expressed in both normal skin and keratinocytes in culture. Expression of sFRP4 mRNA and protein increases with keratinocyte differentiation and apoptosis, whilst exposure of keratinocytes to exogenous sFRP4 promotes apoptosis and expression of the terminal differentiation marker Involucrin. These data suggest sFRP4 promotes epidermal differentiation.


Assuntos
Apoptose , Diferenciação Celular , Células Epidérmicas , Proteínas Proto-Oncogênicas/fisiologia , Apoptose/genética , Diferenciação Celular/genética , Células Cultivadas , Fragmentação do DNA , Epiderme/metabolismo , Humanos , Queratinócitos/citologia , Queratinócitos/metabolismo , Precursores de Proteínas/biossíntese , Proteínas Proto-Oncogênicas/genética , RNA Mensageiro/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...