Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Pharmaceutics ; 15(2)2023 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-36840009

RESUMO

Temozolomide (TMZ), the first-line chemotherapeutic drug against glioblastoma multiforme (GBM), often fails to provide the desired clinical outcomes due to inflammation-induced resistance amid inefficient drug delivery across the blood-brain barrier (BBB). The current study utilized solid lipid nanoparticles (SLNPs) for targeted delivery of TMZ against GBM. After successful formulation and characterization of SLNPs and conjugation with TMZ (SLNP-TMZ), their in-vitro anti-cancer efficacy and effect on the migratory potential of cancer cells were evaluated using temozolomide-sensitive (U87-S) as well as TMZ-resistant (U87-R) glioma cell lines. Elevated cytotoxicity and reduction in cell migration in both cell lines were observed with SLNP-TMZ as compared to the free drug (p < 0.05). Similar results were obtained in-vivo using an orthotopic xenograft mouse model (XM-S and XM-R), where a reduction in tumor size was observed with SLNP-TMZ treatment compared to TMZ. Concomitantly, higher concentrations of the drug were found in brain tissue resections of mice treated with SLNP-TMZ as compared to other vital organs than mice treated with free TMZ. Expression of inflammatory markers (Interleukin-1ß, Interleukin-6 and Tumor Necrosis factor-α) in a resistant cell line (U87-R) and its respective mouse model (XM-R) were also found to be significantly elevated as compared to the sensitive U87-S cell line and its respective mouse model (XM-S). Thus, the in-vitro and in-vivo results of the study strongly support the potential application of SLNP-TMZ for TMZ-sensitive and resistant GBM therapy, indicatively through inflammatory mechanisms, and thus merit further detailed insights.

2.
ACS Omega ; 8(7): 6325-6336, 2023 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-36844542

RESUMO

Gold nanoparticles (AuNPs) have gained importance in the field of biomedical research and diagnostics due to their unique physicochemical properties. This study aimed to synthesize AuNPs using Aloe vera extract, honey, and Gymnema sylvestre leaf extract. Physicochemical parameters for the optimal synthesis of AuNPs were determined using 0.5, 1, 2, and 3 mM of gold salt at varying temperatures from 20 to 50 °C. X-ray diffraction was used to evaluate the crystal structure of AuNPs, which came out to be a face-centered cubic structure. Scanning electron microscopy and energy-dispersive X-ray spectroscopy analysis confirmed the size and shape of AuNPs between 20 and 50 nm from the Aloe vera, honey, and Gymnema sylvestre, as well as large-sized nanocubes in the case of honey, with 21-34 wt % of gold content. Furthermore, Fourier transform infrared spectroscopy confirmed the presence of a broadband of amine (N-H) and alcohol groups (O-H) on the surface of the synthesized AuNPs that prevents them from agglomeration and provides stability. Broad and weak bands of aliphatic ether (C-O), alkane (C-H), and other functional groups were also found on these AuNPs. DPPH antioxidant activity assay showed a high free radical scavenging potential. The most suited source was selected for further conjugation with three anticancer drugs including 4-hydroxy Tamoxifen, HIF1 alpha inhibitor, and the soluble Guanylyl Cyclase Inhibitor 1 H-[1,2,4] oxadiazolo [4,3-alpha]quinoxalin-1-one (ODQ). Evidence of the pegylated drug conjugation with AuNPs was reinforced by ultraviolet/visible spectroscopy. These drug-conjugated nanoparticles were further checked on MCF7 and MDA-MB-231 cells for their cytotoxicity. These AuNP-conjugated drugs can be a good candidate for breast cancer treatment that will lead toward safe, economical, biocompatible, and targeted drug delivery systems.

3.
Front Genet ; 12: 663787, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34262595

RESUMO

Pancreatic cancer (PaCa) is the seventh most fatal malignancy, with more than 90% mortality rate within the first year of diagnosis. Its treatment can be improved the identification of specific therapeutic targets and their relevant pathways. Therefore, the objective of this study is to identify cancer specific biomarkers, therapeutic targets, and their associated pathways involved in the PaCa progression. RNA-seq and microarray datasets were obtained from public repositories such as the European Bioinformatics Institute (EBI) and Gene Expression Omnibus (GEO) databases. Differential gene expression (DE) analysis of data was performed to identify significant differentially expressed genes (DEGs) in PaCa cells in comparison to the normal cells. Gene co-expression network analysis was performed to identify the modules co-expressed genes, which are strongly associated with PaCa and as well as the identification of hub genes in the modules. The key underlaying pathways were obtained from the enrichment analysis of hub genes and studied in the context of PaCa progression. The significant pathways, hub genes, and their expression profile were validated against The Cancer Genome Atlas (TCGA) data, and key biomarkers and therapeutic targets with hub genes were determined. Important hub genes identified included ITGA1, ITGA2, ITGB1, ITGB3, MET, LAMB1, VEGFA, PTK2, and TGFß1. Enrichment analysis characterizes the involvement of hub genes in multiple pathways. Important ones that are determined are ECM-receptor interaction and focal adhesion pathways. The interaction of overexpressed surface proteins of these pathways with extracellular molecules initiates multiple signaling cascades including stress fiber and lamellipodia formation, PI3K-Akt, MAPK, JAK/STAT, and Wnt signaling pathways. Identified biomarkers may have a strong influence on the PaCa early stage development and progression. Further, analysis of these pathways and hub genes can help in the identification of putative therapeutic targets and development of effective therapies for PaCa.

4.
Br J Pharmacol ; 177(10): 2365-2380, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31975484

RESUMO

BACKGROUND AND PURPOSE: Breast cancer is the leading cause of death in women worldwide, with resistance to current therapeutic strategies, including tamoxifen, causing major clinical challenges and leading to more aggressive and metastatic disease. To address this, novel strategies that can inhibit the mechanisms responsible for tamoxifen resistance need to be assessed. EXPERIMENTAL APPROACH: We examined the effect of the novel, clinically-trialled, thiosemicarbazone anti-cancer agent, DpC, and its potential as a combination therapy with the clinically used estrogen receptor (ER) antagonist, tamoxifen, using both tamoxifen-resistant and -sensitive, human breast cancer cells (MDA-MB-453, MDA-MB-231 and MCF-7) in 2D and 3D cell-culture. Synergy was assessed using the Chou-Talalay method. The molecular and anti-proliferative effects of these agents and their combination was examined via Western blot, immunofluorescence and colony formation assays. KEY RESULTS: Combinations of tamoxifen with DpC were highly synergistic, leading to potent inhibition of cell proliferation, colony formation, and ER-α transcriptional activity. The combination also more efficiently reduced major molecular drivers of proliferation of tamoxifen-resistant cells, including c-Myc, cyclin D1, and p-AKT, while up-regulating the cell cycle inhibitor, p27, and inhibiting oncogenic phosphorylation of ER-α at Ser167. Assessing these effects using 3D cell culture further confirmed the greater effects of DpC combined with tamoxifen in reducing ER-α expression, and that of the proliferation marker, Ki-67, in both tamoxifen-sensitive and -resistant MCF-7 spheroids. CONCLUSIONS AND IMPLICATIONS: These studies demonstrate that the synergistic combination of DpC with tamoxifen could be a promising new therapeutic strategy to overcome tamoxifen resistance in ER-positive breast cancer.


Assuntos
Neoplasias da Mama , Tiossemicarbazonas , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Feminino , Humanos , Células MCF-7 , Receptores de Estrogênio , Tamoxifeno/farmacologia , Tiossemicarbazonas/farmacologia
5.
Gene ; 679: 44-56, 2018 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-30118891

RESUMO

Breast Cancer is the most common cancer among women with several genes involved in disease susceptibility. As majority of genome-wide significant variants fall outside the coding region, it is likely that some of them alter specific gene functions. GWAS database was used to interpret the regulatory functions of these genetic variants. A total of 320 SNPs for breast cancer were selected via GWAS, which were entered into the SNAP web portal tool, to determine the one's found to be in Linkage Disequilibrium (r2 < 0.80). The resulting 2024 proxy SNP's were processed in RegulomeDB to predict their regulatory role. Of these, 1440 produced a score ranging from 1-6, whereas the remaining produced no data. Only the variants under score 4 (cut-off value) in RegulomeDB has been studied further. From these variants, 221 had scores of less than 4, indicating a high degree of potential regulatory role associated with them. Further study revealed that 61 of the 221 SNPs were reported to be genome-wide significant for breast cancer, 52 to be associated with other diseases, 99 as unconfirmed for association with breast cancer, leaving only 9 to be novel proxy SNPs linked to breast cancer. Therefore, the study further confirmed postulation of non-coding variants being linked to disease risk thereby, requiring additional validation through genome-wide association studies to substantiate their underlying mechanism.


Assuntos
Neoplasias da Mama/genética , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único , Biologia Computacional/métodos , Bases de Dados Genéticas , Feminino , Redes Reguladoras de Genes , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Humanos
6.
PLoS One ; 13(5): e0196312, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29787591

RESUMO

Insulin-like growth factor 1 receptor (IGF-1R) is an important therapeutic target for breast cancer treatment. The alteration in the IGF-1R associated signaling network due to various genetic and environmental factors leads the system towards metastasis. The pharmacophore modeling and logical approaches have been applied to analyze the behaviour of complex regulatory network involved in breast cancer. A total of 23 inhibitors were selected to generate ligand based pharmacophore using the tool, Molecular Operating Environment (MOE). The best model consisted of three pharmacophore features: aromatic hydrophobic (HyD/Aro), hydrophobic (HyD) and hydrogen bond acceptor (HBA). This model was validated against World drug bank (WDB) database screening to identify 189 hits with the required pharmacophore features and was further screened by using Lipinski positive compounds. Finally, the most effective drug, fulvestrant, was selected. Fulvestrant is a selective estrogen receptor down regulator (SERD). This inhibitor was further studied by using both in-silico and in-vitro approaches that showed the targeted effect of fulvestrant in ER+ MCF-7 cells. Results suggested that fulvestrant has selective cytotoxic effect and a dose dependent response on IRS-1, IGF-1R, PDZK1 and ER-α in MCF-7 cells. PDZK1 can be an important inhibitory target using fulvestrant because it directly regulates IGF-1R.


Assuntos
Antineoplásicos/farmacologia , Estradiol/análogos & derivados , Receptores de Somatomedina/antagonistas & inibidores , Antineoplásicos/química , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Bases de Dados de Produtos Farmacêuticos , Avaliação Pré-Clínica de Medicamentos , Estradiol/química , Estradiol/farmacologia , Antagonistas do Receptor de Estrogênio/química , Antagonistas do Receptor de Estrogênio/farmacologia , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Fulvestranto , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Ligantes , Células MCF-7 , Proteínas de Membrana , Modelos Químicos , Modelos Moleculares , Receptor IGF Tipo 1 , Receptores de Somatomedina/química , Receptores de Somatomedina/genética , Transdução de Sinais/efeitos dos fármacos , Interface Usuário-Computador
7.
PeerJ ; 5: e3822, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28929029

RESUMO

BACKGROUND: C-X-C chemokine ligand 12 (CXCL12) has important implications in breast cancer (BC) pathogenesis. It is selectively expressed on B and T lymphocytes and is involved in hematopoiesis, thymocyte trafficking, stem cell motility, neovascularization, and tumorigenesis. The single nucleotide polymorphism (SNP) rs1801157 of CXCL12 gene has been found to be associated with higher risk of BC. METHODS: Our study focuses on the genotypic and allelic distribution of SNP (rs1801157; G/A) in Pakistani population as well as its association with the clinico-pathological features. The association between rs1801157 genotypes (G/A) and BC risks was assessed by a multivariate logistic regression (MLR) analysis. Genotyping was performed in both healthy individuals and patients of BC using PCR-restriction fragment length polymorphism (PCR-RFLP) method. Furthermore, in-silico approaches were adapted to investigate the association of CXCL12 and its receptor CXCR4 with genes/proteins involved in BC signalling. RESULTS: Significant differences in allelic and genotypic distribution between BC patients and healthy individuals of genotype (G/G) and (A/G) (p < 0.05) were observed. The frequency of the allele G in the BC group (77%) was significantly higher as compared to control group (61%) (p = 0.01). The association of genotype GG with clinico-pathological features including age, stages of cancer and organ (lung, liver, bones and brain) metastasis (p > 0.05) was assessed. In a MLR analysis, a number of variables including age, weight of an individual, affected lymph nodes, hormonal status (estrogen and progesterone receptor), alcohol consumption and family history associated with the GG genotype (GG:AA, odds ratio (OR) = 1.30, 95% CI [1.06-1.60]) were found to be independent risk factors for BC. Our in-vitro results suggest that genotype GG is possibly increasing the risk of BC in Pakistani cohorts. in-silico analysis finds that CXCL12-CXCR4 is associated with an increased expression of PDZK1, PI3k and Akt which lead the breast tumor towards metastasis. CONCLUSION: Multiple targets such as CXCL12, CXCR4, PDZK1, PI3k and Akt can be inhibited in combined strategies to treat BC metastasis.

8.
PeerJ ; 4: e2542, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27781158

RESUMO

BACKGROUND: Breast cancer (BC) is one of the leading cause of death among females worldwide. The increasing incidence of BC is due to various genetic and environmental changes which lead to the disruption of cellular signaling network(s). It is a complex disease in which several interlinking signaling cascades play a crucial role in establishing a complex regulatory network. The logical modeling approach of René Thomas has been applied to analyze the behavior of estrogen receptor-alpha (ER-α) associated Biological Regulatory Network (BRN) for a small part of complex events that leads to BC metastasis. METHODS: A discrete model was constructed using the kinetic logic formalism and its set of logical parameters were obtained using the model checking technique implemented in the SMBioNet software which is consistent with biological observations. The discrete model was further enriched with continuous dynamics by converting it into an equivalent Petri Net (PN) to analyze the logical parameters of the involved entities. RESULTS: In-silico based discrete and continuous modeling of ER-α associated signaling network involved in BC provides information about behaviors and gene-gene interaction in detail. The dynamics of discrete model revealed, imperative behaviors represented as cyclic paths and trajectories leading to pathogenic states such as metastasis. Results suggest that the increased expressions of receptors ER-α, IGF-1R and EGFR slow down the activity of tumor suppressor genes (TSGs) such as BRCA1, p53 and Mdm2 which can lead to metastasis. Therefore, IGF-1R and EGFR are considered as important inhibitory targets to control the metastasis in BC. CONCLUSION: The in-silico approaches allow us to increase our understanding of the functional properties of living organisms. It opens new avenues of investigations of multiple inhibitory targets (ER-α, IGF-1R and EGFR) for wet lab experiments as well as provided valuable insights in the treatment of cancers such as BC.

9.
Haematologica ; 97(5): 661-9, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22180422

RESUMO

BACKGROUND: Iron metabolism during pregnancy maintains fetal iron levels at the expense of the mother. The mechanism behind this regulation is still not clear despite recent advances. Here we examine the role of maternal and fetal Hfe, its downstream signaling molecule, hepcidin and dietary iron in the regulation of placental iron transfer. DESIGN AND METHODS: Hfe wild-type, knockout and heterozygote dams were fed iron deficient (12.5 ppm), adequate (50 ppm) and replete (150 ppm) iron diets and mated with heterozygote males to produce pups of all genotypes. Dams and pups were sacrificed at Day 18 of gestation; serum, placenta, body and liver iron parameters were measured. Protein and mRNA levels of various iron transporter genes were determined in duodenum, liver and placenta by Western blotting and real time PCR. RESULTS: Maternal liver iron levels were dependent on both dietary iron intake and Hfe genotype. Increasing iron levels in the maternal diet resulted in increased total iron in the fetus, primarily in the liver. However, fetuses of Hfe-knockout mothers showed further elevation of liver iron levels, concomitant with elevated expression of Tfr1, Dmt1 and Fpn in the placenta. Hfe-knockout fetuses that express low levels of liver hepcidin accumulated more iron in their liver than wild-type fetuses due to increased ferroportin levels in the placenta. CONCLUSIONS: Maternal and fetal status, as well as dietary iron, is important in regulating iron transfer across placenta. Maternal Hfe regulates iron transfer by altering gene expression in the placenta. Fetal Hfe is important in regulating placental iron transfer by modulating fetal liver hepcidin expression.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/genética , Feto/metabolismo , Antígenos de Histocompatibilidade Classe I/fisiologia , Ferro da Dieta/administração & dosagem , Ferro/metabolismo , Fígado/metabolismo , Proteínas de Membrana/fisiologia , Placenta/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/metabolismo , Western Blotting , Duodeno/efeitos dos fármacos , Duodeno/metabolismo , Feminino , Sangue Fetal/metabolismo , Feto/efeitos dos fármacos , Feto/embriologia , Proteína da Hemocromatose , Hepcidinas , Fígado/efeitos dos fármacos , Fígado/embriologia , Masculino , Troca Materno-Fetal , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Placenta/efeitos dos fármacos , Gravidez , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Kidney Int ; 78(7): 660-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20631677

RESUMO

Erythropoietin is produced by the kidney and stimulates erythropoiesis; however, in chronic renal disease its levels are reduced and patients develop anemia that is treatable with iron and recombinant hormone. The mechanism by which erythropoietin improves iron homeostasis is still unclear, but it may involve suppression of the iron regulatory peptide hepcidin and/or a direct effect on intestinal iron absorption. To investigate these possibilities, we used the well-established 5/6th nephrectomy rat model of chronic renal failure with or without human recombinant erythropoietin treatment. Monolayers of human intestinal Caco-2 cells were also treated with erythropoietin to measure any direct effects of this hormone on intestinal iron transport. Nephrectomy increased hepatic hepcidin expression and decreased intestinal iron absorption; these effects were restored to levels found in sham-operated rats on erythropoietin treatment of the rats with renal failure. In Caco-2 cells, the addition of erythropoietin significantly increased the expression of apical divalent metal transporter 1 (DMT1) and basolateral ferroportin and, consequently, iron transport across the monolayer. Taken together, our results show that erythropoietin not only exerts a powerful inhibitory action on the expression of hepcidin, thus permitting the release of iron from reticuloendothelial macrophages and intestinal enterocytes, but also acts directly on enterocytes to increase iron absorption.


Assuntos
Eritropoetina/farmacologia , Absorção Intestinal/efeitos dos fármacos , Ferro/metabolismo , Falência Renal Crônica/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Células CACO-2 , Proteínas de Transporte de Cátions/genética , Modelos Animais de Doenças , Duodeno/metabolismo , Hepcidinas , Humanos , Masculino , Nefrectomia , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Receptores da Eritropoetina/análise , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA