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.
Funct Integr Genomics ; 24(5): 152, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39223429

RESUMO

In the complex landscape of cancer biology, the discovery of microproteins has triggered a paradigm shift, thereby, challenging the conventional conceptions of gene regulation. Though overlooked for years, these entities encoded by the small open reading frames (100-150 codons), have a significant impact on various cellular processes. As precision medicine pioneers delve deeper into the genome and proteome, microproteins have come into the limelight. Typically characterized by a single protein domain that directly binds to the target protein complex and regulates their assembly, these microproteins have been shown to play a key role in fundamental biological processes such as RNA processing, DNA repair, and metabolism regulation. Techniques for identification and characterization, such as ribosome profiling and proteogenomic approaches, have unraveled unique mechanisms by which these microproteins regulate cell signaling or pathological processes in most diseases including cancer. However, the functional relevance of these microproteins in cancer remains unclear. In this context, the current review aims to "rethink the essence of these genes" and explore "how these hidden players-microproteins orchestrate the signaling cascades of cancer, both as accelerators and brakes.".


Assuntos
Neoplasias , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Transdução de Sinais , Fases de Leitura Aberta , Animais , Ribossomos/metabolismo , Ribossomos/genética , Micropeptídeos
2.
3 Biotech ; 13(12): 397, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37974928

RESUMO

Glioma coined as a "butterfly" tumor associated with a dismal prognosis. Marine algal compounds with the richest sources of bioactive components act as significant anti-tumor therapeutics. However, there is a paucity of studies conducted on Fucoidan to enhance the anti-glioma efficacy of Temozolomide. Therefore, the present study aimed to evaluate the synergistic anti-proliferative, anti-inflammatory and pro-apoptotic effects of Fucoidan with Temozolomide in in vitro and in silico experimental setup. The anti-proliferative effects of Temozolomide and Fucoidan were evaluated on C6 glioma cells by MTT and migration assay. Modulation of inflammatory markers and apoptosis induction was affirmed at the morphological and transcriptional level by dual staining and gene expression. Molecular docking (MD) and molecular dynamics simulation (MDS) studies were performed against the targets to rationalize the inhibitory effect. The dual-drug combination significantly reduced the cell viability and migration of glioma cells in a synergistic dose-dependent manner. At the molecular level, the dual-drug combination significantly down-regulated inflammatory genes with a concomitant upregulation of pro-apoptotic marker. In consensus with our in vitro findings, molecular docking and simulation studies revealed that the anti-tumor ligands: Temozolomide, Fucoidan with 5-(3-Methy1-trizeno)-imidazole-4-carboxamide (MTIC), and 4-amino-5-imidazole-carboxamide (AIC) had the potency to bind to the inflammatory proteins at their active sites, mediated by H-bonds and other non-covalent interactions. The dual-drug combinatorial treatment synergistically inhibited the proliferation, migration of glioma cells and promoted apoptosis; conversely with the down-regulation of inflammatory genes. However, pre-clinical experimental evidence is warranted for the possible translation of this combination. Supplementary Information: The online version contains supplementary material available at 10.1007/s13205-023-03814-6.

3.
Front Pharmacol ; 14: 1096614, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37025487

RESUMO

Glioma is the most devastating high-grade tumor of the central nervous system, with dismal prognosis. Existing treatment modality does not provide substantial benefit to patients and demands novel strategies. One of the first-line treatments for glioma, temozolomide, provides marginal benefit to glioma patients. Repurposing of existing non-cancer drugs to treat oncology patients is gaining momentum in recent years. In this study, we investigated the therapeutic benefits of combining three repurposed drugs, namely, metformin (anti-diabetic) and epigallocatechin gallate (green tea-derived antioxidant) together with temozolomide in a glioma-induced xenograft rat model. Our triple-drug combination therapy significantly inhibited tumor growth in vivo and increased the survival rate (50%) of rats when compared with individual or dual treatments. Molecular and cellular analyses revealed that our triple-drug cocktail treatment inhibited glioma tumor growth in rat model through ROS-mediated inactivation of PI3K/AKT/mTOR pathway, arrest of the cell cycle at G1 phase and induction of molecular mechanisms of caspases-dependent apoptosis.In addition, the docking analysis and quantum mechanics studies performed here hypothesize that the effect of triple-drug combination could have been attributed by their difference in molecular interactions, that maybe due to varying electrostatic potential. Thus, repurposing metformin and epigallocatechin gallate and concurrent administration with temozolomide would serve as a prospective therapy in glioma patients.

5.
Cell Signal ; 95: 110350, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35525406

RESUMO

Glioblastoma multiforme is one of the calamitous primary glial brain tumors with extensive heterogeneity at cellular and molecular levels. While maximal surgical resection trailed by radio and chemotherapy employing temozolomide remains the gold-standard treatment for malignant glioma patients, the overall prognosis remains dismal and there exists an unmet need for effective therapeutic strategies. In this context, we hypothesize that proper understanding of signaling pathways responsible for glioblastoma multiforme proliferation would be the first trump card while searching for novel targeted therapies. Among the pathways aberrantly activated, PI3K/AKT/mTOR is the most significant pathway, that is clinically implicated in malignancies such as high-grade glioma. Further, the WNT/ß-Catenin cascade is well-implicated in several malignancies, while its role in regulating glioma pathogenesis has only emerged recently. Nevertheless, oncogenic activation of both these pathways is a frequent event in malignant glioma that facilitates tumor proliferation, stemness and chemo-resistance. Recently, it has been reported that the cross-talk of PI3K/AKT/mTOR pathway with multiple signaling pathways could promote glioma progression and reduce the sensitivity of glioma cells to the standard therapy. However, very few studies had focused on the relationship between PI3K/AKT/mTOR and WNT/ß-Catenin pathways in glioblastoma multiforme. Interestingly, in homeostatic and pathologic circumstances, both these pathways depict fine modulation and are connected at multiple levels by upstream and downstream effectors. Thus, gaining deep insights on the collusion between these pathways would help in discovering unique therapeutic targets for glioblastoma multiforme management. Hence, the current review aims to address, "the importance of inter-play between PI3K/AKT/mTOR and WNT/ß-Catenin pathways", and put forward, "the possibility of combinatorially targeting them", for glioblastoma multiforme treatment enhancement.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Serina-Treonina Quinases TOR , Animais , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Terapia Combinada , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
6.
Life Sci ; 301: 120609, 2022 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-35526592

RESUMO

AIMS: Malignant gliomas constitute one of the deadly brain tumors with high degeneration rate. Though temozolomide (TMZ) is the first-line drug for glioma, its efficacy has decreased due to chemo-resistance. Repurposing synthetic and natural compounds have gained increasing interest in glioma. Hence, we combined chloroquine (CHL) a synthetic drug, naringenin (NAR) and phloroglucinol (PGL) (natural derivatives), to investigate whether the apoptotic effect of these drugs both alone and in combination, enhances the anti-tumor effects of TMZ in an in vitro and in vivo orthotopic xenograft glioma model. MAIN METHODS: The cytotoxic effect of the drugs was assessed in C6 (murine) glioma cells, U-87 MG and LN229 (human) glioblastoma cells, primary astrocytes (isolated from rat brain tissues) and HEK-293 T cells. Mitochondrial depolarization and alterations in the cell cycle was determined by confocal imaging and flow cytometry. The expression of angiogenic and apoptotic markers was evaluated using qRT-PCR and ELISA. The efficacy of the combinatorial treatment was assessed in an orthotopic xenograft model using U-87 MG cells. KEY FINDINGS: The combinatorial treatment inhibited cell proliferation, induced apoptosis and contributed to cell cycle arrest in glioma cells. The quadruple combinatorial cocktail down-regulated BCL-2 with a concomitant decrease in VEGF. As observed in vitro, the quadruple combinatorial treatment enhanced the median survival of glioma-induced rats with lower cellularity rate. SIGNIFICANCE: The combination of CHL, NAR and PGL synergistically potentiated the efficacy of TMZ on glioma in vitro and in vivo. Hence, this combination may characterize an advanced strategy for glioma treatment, thereby providing a possible translation to clinical trial.


Assuntos
Neoplasias Encefálicas , Glioma , Animais , Antineoplásicos Alquilantes/farmacologia , Apoptose , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioma/patologia , Células HEK293 , Humanos , Camundongos , Ratos , Temozolomida/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Med Oncol ; 38(5): 53, 2021 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-33811540

RESUMO

Gliomas are one of the most devastating primary brain tumors which impose significant management challenges to the clinicians. The aggressive behaviour of gliomas is mainly attributed to their rapid proliferation, unravelled genomics and the blood-brain barrier which protects the tumor cells from chemotherapeutic regimens. Suspects of brain tumors are usually assessed by magnetic resonance imaging and computed tomography. These images allow surgeons to decide on the tumor grading, intra-operative pathology, feasibility of surgery, and treatment planning. All these data are compiled manually by physicians, wherein it takes time for the validation of results and concluding the treatment modality. In this context, the arrival of artificial intelligence in this era of personalized medicine, has proven promising performance in the diagnosis and management of gliomas. Starting from grading prediction till outcome evaluation, artificial intelligence-based forefronts have revolutionized oncological research. Interestingly, this approach has also been able to precisely differentiate tumor lesion from healthy tissues. However, till date, their utility in neuro-oncological field remains limited due to the issues pertaining to their reliability and transparency. Hence, to shed novel insights on the "clinical utility of this novel approach on glioma management" and to reveal "the black-boxes that have to be solved for fruitful application of artificial intelligence in neuro-oncology research", we provide in this review, a succinct description of the potential gear of artificial intelligence-based avenues in glioma treatment and the barriers that impede their rapid implementation in neuro-oncology.


Assuntos
Inteligência Artificial/tendências , Neoplasias Encefálicas/terapia , Glioma/terapia , Inteligência , Oncologia/tendências , Algoritmos , Neoplasias Encefálicas/diagnóstico , Glioma/diagnóstico , Humanos , Oncologia/métodos , Gradação de Tumores/métodos , Gradação de Tumores/tendências
8.
Mol Biotechnol ; 63(2): 93-108, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33386579

RESUMO

Genome editing employs targeted nucleases as powerful tools to precisely alter the genome of target cells and regulate functional genes. Various strategies have been risen so far as the molecular scissors-mediated genome editing that includes zinc finger nuclease, transcription activator-like effector nucleases, and clustered regularly interspaced short palindromic repeats-CRISPR-related protein 9. These tools allow researchers to understand the basics of manipulating the genome, create animal models to study human diseases, understand host-pathogen interactions and design disease targets. Targeted genome modification utilizing RNA-guided nucleases are of recent curiosity, as it is a fast and effective strategy that enables the researchers to manipulate the gene of interest, carry out functional studies, understand the molecular basis of the disease and design targeted therapies. CRISPR-Cas9, a bacterial defense system employed against viruses, consists of a single-strand RNA-guided Cas9 nuclease connected to the corresponding complementary target sequence. This powerful and versatile tool has gained tremendous attention among the researchers, owing to its ability to correct genetic disorders. To help illustrate the potential of this gene editor in unexplored corners of oncology, we describe the history of CRISPR-Cas9, its rapid progression in cancer research as well as future perspectives.


Assuntos
Sistemas CRISPR-Cas/genética , Neoplasias/terapia , Imunidade Adaptativa , Animais , Pesquisa Biomédica , Modelos Animais de Doenças , Humanos , Neoplasias/diagnóstico , Neoplasias/imunologia , Linfócitos T/imunologia
9.
J Biomed Mater Res B Appl Biomater ; 107(5): 1372-1383, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30265773

RESUMO

The proposed work involves an exclusive study on the synthesis protocol, crystal structure analysis, and imaging contrast features of unique lanthanide phosphates (LnPO4 ). XRD and Raman spectra affirmed the ability of the proposed synthesis technique to achieve unique LnPO4 devoid of impurities. The crystal structure analysis confirms the P121/c1 space setting of NdPO4 , EuPO4 , GdPO4 , and TbPO4 that all uniformly crystallizes in monoclinic unit cell. In a similar manner, the tetragonal crystal setting of DyPO4 , ErPO4 , HoPO4 , and YbPO4 that unvaryingly possess the I41/amd space setting is confirmed. Under the same synthesis conditions, the monoclinic (Eu) and tetragonal (Ho) lanthanide phosphates displayed uniform rod-like morphologies. Absorption and luminescence properties of unique LnPO4 were determined. In vitro biological studies demonstrated low toxicity levels of LnPO4 and clearly distinguished fluorescence of TbPO4 and EuPO4 in Y79, retinoblastoma cell lines. The paramagnetic response of GdPO4 , NdPO4 , DyPO4 , TbPO4 , and HoPO4 facilitated excellent magnetic resonance imaging (MRI) contrast features. Meanwhile, GdPO4 , DyPO4 , HoPO4 , and YbPO4 possessing higher X-ray absorption coefficient than clinical contrast Omnipaque™ exhibited high computed tomography (CT) efficiency. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1372-1383, 2019.


Assuntos
Meios de Contraste , Lantânio , Luminescência , Imageamento por Ressonância Magnética , Sondas Moleculares , Fosfatos , Tomografia Computadorizada por Raios X , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/farmacologia , Humanos , Lantânio/química , Lantânio/farmacologia , Sondas Moleculares/química , Sondas Moleculares/farmacologia , Fosfatos/química , Fosfatos/farmacologia
10.
Protein Expr Purif ; 88(2): 214-20, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23337085

RESUMO

Keratinolytic proteases find extensive applications both in environmental biotechnology and pharmaceutical industries. An extracellular keratinolytic protease was purified and characterized from the fungus, Aspergillus parasiticus, isolated from poultry soil. The enzyme was purified to homogeneity by acetone and ammonium sulfate precipitations followed by CM-Sepharose column chromatography. The molecular mass of the enzyme was 36kDa as judged by SDS-PAGE. The purified keratinase had a pH optimum of 7.0 and temperature optimum of 50(o)C. The enzyme hydrolyzed the substrate azocasein and the Km and Vmax of the purified keratinase were found to be 1.04mg/ml and 3463.34Units/min/mg protein, respectively. The enzyme showed increased activity in the presence of reducing agents. The enzyme was found to be glycosylated. According to the inhibition profiles obtained with the various protease inhibitors, it was confirmed that the purified keratinase belongs to the serine protease type. The purified enzyme activity was enhanced by calcium, magnesium and manganese ions and partially inhibited by cadmium, copper and zinc ions. The purified enzyme showed increased activity with nonionic detergents and urea.


Assuntos
Aspergillus/enzimologia , Peptídeo Hidrolases/química , Peptídeo Hidrolases/metabolismo , Sequência de Aminoácidos , Sulfato de Amônio/química , Aspergillus/química , Aspergillus/isolamento & purificação , Precipitação Química , Detergentes/metabolismo , Eletroforese em Gel de Poliacrilamida , Concentração de Íons de Hidrogênio , Dados de Sequência Molecular , Peptídeo Hidrolases/isolamento & purificação , Substâncias Redutoras/metabolismo , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA