Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Biochem Genet ; 2024 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-38436816

RESUMO

Multiple myeloma is a type of malignant neoplasia whose treatment has changed over the past decade. This study aimed to investigate the effects of combination of Adenovector-carrying interleukin-24 and herpes simplex virus 1 thymidine kinase/ganciclovir on tumor growth, autophagy, and unfolded protein response mechanisms in mouse model of multiple myeloma. Six groups of mice, including Ad-HSV-tk/GCV, Ad-IL-24, Ad-HSV-tk/IL-24, Ad-GFP, and positive and negative controls, were investigated, and each group was injected every 72 h. The tumor size was measured several times. The expression of LC3B evaluated through western blotting and ASK-1, CHOP, Caspase-3, and ATF-6 genes in the UPR and apoptosis pathways were also analyzed by the quantitative polymerase chain reaction (qPCR) method. The present results showed that the injection of Ad-HSV-tk/GCV, Ad-HSV-tk/IL-24, and metformin reduced the tumor size. The expression of LC3B was significantly higher in the treatment groups and positive control groups compared to the negative control group. The expression of CHOP, caspase-3, and ATF-6 genes was significantly higher in the Ad-IL-24 group compared to the other treatment groups. Besides, the ASK-1 expression was significantly lower in the Ad-IL-24 group as compared to the other groups. Overall, the results indicated that the presence of the HSV-tk gene in the adenovectors reduced the size of tumors and induced autophagy by triggering the expression of LC3B protein. The presence of the IL-24 might affect tumor growth but not as much the therapeutic effect of HSV-tk. Furthermore, the results indicated that co-administration of IL-24 and HSV-tk had no synergistic effect on tumor size control.

2.
Adv Pharm Bull ; 13(4): 817-826, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38022809

RESUMO

Purpose: Despite the development of anti-human papillomavirus (HPV) vaccines, cervical cancer is still a common disease in women, especially in developing countries. The presence of a hypoxic microenvironment causes traditional treatments to fail. In this study, we presented a combined treatment method based on the chemotherapeutic agent cisplatin and Clostridium novyi-NT spores to treat normoxic and hypoxic areas of the tumor. Methods: TC-1 Cell line capable of expressing HPV-16 E6/7 oncoproteins was subcutaneously transplanted into female 6-8 week old C57/BL6 mice. The tumor-bearing mice were randomly divided into four groups and treated with different methods after selecting a control group. Group 1: Control without treatment (0.1 mL sterile PBS intratumorally), Group: C. novyi-NT (107 C. novyi-NT). Group 3: Receives cisplatin intraperitoneally (10 mg/kg). Fourth group: Intratumoral administration of C. novyi-NT spores + intraperitoneal cisplatin. Western blot analysis was used to examine the effects of anti-hypoxia treatment and expression of hypoxia-inducible factor 1 (HIF-1) and vascular endothelial growth factor (VEGF) proteins. Results: The results clearly showed that combined treatment based on C. novyi-NT and cisplatin significantly reduced the expression of HIF-1 alpha and VEGF proteins compared to cisplatin alone. At the same time, the amount of necrosis of tumor cells in the combined treatment increased significantly compared to the single treatment and the control. At the same time, the mitotic count decreased significantly. Conclusion: Our research showed that developing a combined treatment method based on C. novyi-NT and cisplatin against HPV-positive cervical cancer could overcome the treatment limitations caused by the existence of hypoxic areas of the tumor.

3.
Curr Cancer Drug Targets ; 23(9): 682-696, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37069721

RESUMO

Resistance to conventional antitumour therapies and Hypoxia in patients with advanced solid tumours are two major reasons for the failure of conventional anti-tumour therapies. Therefore, it is important to find a new therapeutic method that can overcome these problems. An attenuated anaerobic bacterium, Clostridium novyi-NT, could target Hypoxic and Necrotic areas of tumours causing tumour lysis and stimulating a host anti-tumour immune response. To the best of our knowledge, the combination of bacterial anti-tumour therapy, chemotherapy, radiotherapy and immunotherapy may promote tumour regression, inhibit metastasis and develop a new strategy for the treatment of solid tumours. However, the possible molecular mechanisms of the combined therapies are still the biggest challenge. This review provides an overview of the history of bacterial cancer therapy and the development of a non-lethal strain of Clostridium novyi. Below is a precise definition of Hypoxic conditions in solid tumour tissue. To understand the anticancer effect of Clostridium novyi-NT spores, possible cell death mechanisms were summarised by the enzyme phospholipase C (nt01cx0979), which is secreted by Clostridium novyi-NT spores after germination in tumour tissue. The function of Clostridium novyi-NT spores in stimulating the host immune system to elicit anti-tumour responses was reviewed. Then, the results of anti-tumour combination therapies based on Clostridium novyi-NT spores were compiled. Identifying the molecular mechanisms of Clostridium novyi-NT in treating tumours and inducing cell death in invasive cancer cells, ultimately leading to tumour regression, may develop promising clinical strategies in the combined treatment of solid tumours.


Assuntos
Neoplasias , Humanos , Composição de Bases , Filogenia , RNA Ribossômico 16S , Análise de Sequência de DNA , Neoplasias/tratamento farmacológico , Neoplasias/patologia
4.
Bioimpacts ; 12(5): 405-414, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36381634

RESUMO

Introduction: Hypoxia context is highly specific for tumors and represents a unique niche which is not found elsewhere in the body. Clostridium novyi is an obligate anaerobic bacterium. It has a potential to treat tumors. The aim of this study was to produce the C. novyi nontoxic spores and to investigate its oncolytic effect on breast cancer in mice model. Methods: Primarily, the lethal toxin gene in C. novyi type B was removed. Colonies were isolated using PCR testing. To assure the removal of alpha-toxin, plasmid extraction and in vivo assay were conducted. Next, to treat breast cancer model in different sizes of tumors, a single dose of spores of C. novyi nontoxic was tested. Results: The results denoted that C. novyi nontoxic lost lethal toxin and a--ppeared to be safe. For smaller than 1000 mm3 tumors, a single dose of C. novyi nontoxic was able to cure 100% of mice bearing breast tumors. Hence the mice remained free of tumor relapse. Tumors larger than 1000 mm3 were not cured by a single dose- of C. novyi nontoxic treatment. Conclusion: The experiment concluded that the C. novyi nontoxic might be a suitable and safe candidate, a novel therapeutic approach to encounter such hypoxic regions in the center of tumors. Research also showed that bacteriolytic therapy by C. novyi nontoxic could lead to regression in small tumor.

5.
Cancer Cell Int ; 22(1): 164, 2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35477503

RESUMO

BACKGROUND: Novel strategies are required since the hypoxic tumor microenvironment is one of the important impediments for conventional cancer therapy. High mobility group box 1 (HMGB1) protein can block aerobic respiration in cancer cells. We hypothesized that HMGB1could also kill the colorectal cancer cells during hypoxia. METHODS: In this study, we developed oncolytic herpes simplex virus type 1 expressing HMGB1 protein (HSV-HMGB1) and investigated the cytotoxic effect of HSV-HMGB1 and its parental virus (HSV-ble) on three colorectal cancer cells (HCT116, SW480, and HT29) under normoxic (20% oxygen) and hypoxic (1% oxygen) conditions. We further identified potential autophagy- related genes in HT29 cells by retrieving mRNA expression microarray datasets from the Gene Expression Omnibus database. These genes were then detected in HT29 cells infected with HSV-HMGB1 and HSV-ble during normoxia and hypoxia by Real-Time quantitative PCR (qRT-PCR). RESULTS: The cytotoxic effect of HSV-HMGB1 was significantly higher than that of HSV-ble during normoxia; however, during hypoxia, HSV-HMGB1 enhanced the viability of HT29 cells at MOI 0.1. Analyzing the cell death pathway revealed that HSV-HMGB1 induced autophagy in HT29 cells under hypoxic conditions. CONCLUSION: In conclusion, it appears that oncolytic virotherapy is cell context-dependent. Therefore, understanding the cancer cells' characteristics, microenvironment, and cell signaling are essential to improve the therapeutic strategies.

6.
Cancer Treat Res Commun ; 30: 100512, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35026533

RESUMO

The autophagy pathway is the process whereby cells keep cellular homeostasis and respond to stress via recycling their damaged cellular proteins, organelles, and other cellular components. In the context of cancer, autophagy is a dual-edge sword pro- and anti-tumorigenic role depending on the oncogenic context and stage of tumorigenesis. Cancer cells have a higher dependency on autophagy compared with normal cells because of cellular damages and high demands for energy. The carbon, nitrogen, and molecular oxygen are building blocks for highly proliferative cancer cells which extremely depend on glutaminolysis and aerobic glycolysis; when a cancer cell is restricted to glucose and glutamine, it initiates to activate a stress response pathway using autophagy. Oncogenic tyrosine kinases (OncTKs) and receptor tyrosine kinases (RTKs) activation result in autophagy modulation through activation of the PI3K/AKT/mTORC1 and RAS/MAPK signaling pathways. Targeted inhibition of tyrosine kinases (TKs) and RTKs have recently been considered as cancer therapy but drug resistance and cancer relapse continue to be a major limitation of tyrosine kinase inhibitors (TKIs). Manipulation of autophagy pathway along with TKIs may be a promising strategy to circumvent unknown existing drug-resistance mechanisms that may emerge in a treated patient. In this way, clinical trials are ongoing to modulate autophagy to treat cancer. This review aims to summarize the combination therapy of autophagy affecting compounds with anticancer drugs which target cell signaling pathways, metabolism mechanisms, and epigenetics modification to improve therapeutic efficacy against cancers.

7.
J Cancer Res Ther ; 17(1): 225-230, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33723159

RESUMO

BACKGROUND: Telomeres through maintaining chromosomal integrity have key roles in the cell life span. The autophagy is typically a pro-survival process and important for maintaining cellular homeostasis. Conversely, in some conditions, autophagy acts as caspase-independent cell death program. Beclin1 gene plays a principal role in the initiation of autophagy. OBJECTIVE: The aim of this study was to evaluate the effect of autophagy induction via recombinant Beclin1 on telomerase activity and programmed cell death (apoptosis) in MCDK cells. MATERIALS AND METHODS: The recombinant Beclin1-pcDNA3.1(-) was transfected into MDCK cells. Next, the autophagy information was detected by LC3II staining as autophagy marker using flow cytometry. The telomerase activity was measured by telomeric repeat amplification protocol method in MDCK cells. To detection of the cell death in MDCK cells, apoptosis assay was done through Annexin V staining method. RESULTS: The results of flow cytometry analysis indicated that following overexpression of Beclin1 gene, the percentage of the LC3II was 16.08% compared with control group (0.48%). Following induction of autophagy, telomerase activity reduced 10 folds in comparison with the control group. The rate of apoptosis in transfected MDCK cells increased up to 12.74%. CONCLUSION: Crosstalk between telomerase, autophagy, and apoptosis may determine the fate of the cancer cell aging. Hence, manipulation of autophagy may create a novel area to design new compounds and combination therapy to shorten the cancer cell survival.


Assuntos
Apoptose , Autofagia , Proteína Beclina-1/metabolismo , Neoplasias/patologia , Telomerase/antagonistas & inibidores , Animais , Proteína Beclina-1/genética , Cães , Células Madin Darby de Rim Canino , Neoplasias/metabolismo , Transdução de Sinais , Telomerase/metabolismo
8.
Cancer Treat Res Commun ; 27: 100323, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33530025

RESUMO

Human telomerase reverse transcriptase (hTERT) is an enzyme that is critically involved in elongating and maintaining telomeres length to control cell life span and replicative potential. Telomerase activity is continuously expressed in human germ-line cells and most cancer cells, whereas it is suppressed in most somatic cells. In normal cells, by reducing telomerase activity and progressively shortening the telomeres, the cells progress to the senescence or apoptosis process. However, in cancer cells, telomere lengths remain constant due to telomerase's reactivation, and cells continue to proliferate and inhibit apoptosis, and ultimately lead to cancer development and human death due to metastasis. Studies demonstrated that several DNA and RNA oncoviruses could interact with telomerase by integrating their genome sequence within the host cell telomeres specifically. Through the activation of the hTERT promoter and lengthening the telomere, these cells contributes to cancer development. Since oncoviruses can activate telomerase and increase hTERT expression, there are several therapeutic strategies based on targeting the telomerase of cancer cells like telomerase-targeted peptide vaccines, hTERT-targeting dendritic cells (DCs), hTERT-targeting gene therapy, and hTERT-targeting CRISPR/Cas9 system that can overcome tumor-mediated toleration mechanisms and specifically apoptosis in cancer cells. This study reviews available data on the molecular structure of telomerase and the role of oncoviruses and telomerase interaction in cancer development and telomerase-dependent therapeutic approaches to conquest the cancer cells.


Assuntos
Neoplasias/genética , Proteínas Oncogênicas Virais/metabolismo , Retroviridae/patogenicidade , Telomerase/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/genética , Senescência Celular/genética , Modelos Animais de Doenças , Terapia Genética/métodos , Interações entre Hospedeiro e Microrganismos/genética , Humanos , Camundongos , Neoplasias/terapia , Neoplasias/virologia , Proteínas Oncogênicas Virais/genética , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Regiões Promotoras Genéticas , Retroviridae/genética , Telomerase/antagonistas & inibidores , Telômero/metabolismo , Homeostase do Telômero
9.
Autophagy ; 17(1): 1-382, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33634751

RESUMO

In 2008, we published the first set of guidelines for standardizing research in autophagy. Since then, this topic has received increasing attention, and many scientists have entered the field. Our knowledge base and relevant new technologies have also been expanding. Thus, it is important to formulate on a regular basis updated guidelines for monitoring autophagy in different organisms. Despite numerous reviews, there continues to be confusion regarding acceptable methods to evaluate autophagy, especially in multicellular eukaryotes. Here, we present a set of guidelines for investigators to select and interpret methods to examine autophagy and related processes, and for reviewers to provide realistic and reasonable critiques of reports that are focused on these processes. These guidelines are not meant to be a dogmatic set of rules, because the appropriateness of any assay largely depends on the question being asked and the system being used. Moreover, no individual assay is perfect for every situation, calling for the use of multiple techniques to properly monitor autophagy in each experimental setting. Finally, several core components of the autophagy machinery have been implicated in distinct autophagic processes (canonical and noncanonical autophagy), implying that genetic approaches to block autophagy should rely on targeting two or more autophagy-related genes that ideally participate in distinct steps of the pathway. Along similar lines, because multiple proteins involved in autophagy also regulate other cellular pathways including apoptosis, not all of them can be used as a specific marker for bona fide autophagic responses. Here, we critically discuss current methods of assessing autophagy and the information they can, or cannot, provide. Our ultimate goal is to encourage intellectual and technical innovation in the field.


Assuntos
Autofagia , Animais , Autofagossomos , Autofagia/fisiologia , Proteínas Relacionadas à Autofagia/metabolismo , Bioensaio/normas , Biomarcadores , Humanos , Lisossomos
10.
Sci Rep ; 10(1): 5787, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32238821

RESUMO

Cancer is a leading cause of death worldwide. Cervical cancer caused by human papillomavirus (HPV) is a major health problem in women. DNA vaccines are a perfect approach to immunization, but their potency in clinical trials has been insufficient for generating effective immunity, which may be related to the degradation of the DNA via nucleases, poor delivery to antigen-presenting cells (APCs), and insufficient uptake of DNA plasmids by cells upon injection. Archaeosome is a nano-delivery systems based on liposomes with their immunological role have been developed for gene delivery. In this study, human papillomavirus type 16 genes, containing truncated L1, E6, and E7, were simultaneously used in combination therapy with archaeosome and assessed in vivo. Findings supported that archaeosomes promotes immune responses to DNA vaccines and a long-term CTL response was generated with a low antigen dose. Combination therapy with archaeosome/L1/E6/E7 vaccines exhibited a strong cytolytic activity against tumor cells and induced prophylactic and therapeutic effect against the development of tumor in the animal model.


Assuntos
Papillomavirus Humano 16/genética , Proteínas Oncogênicas Virais/genética , Infecções por Papillomavirus/prevenção & controle , Vacinas contra Papillomavirus/uso terapêutico , Neoplasias do Colo do Útero/prevenção & controle , Vacinas de DNA/uso terapêutico , Animais , Feminino , Técnicas de Transferência de Genes , Genes Virais , Células HEK293 , Humanos , Camundongos Endogâmicos C57BL , Proteínas E7 de Papillomavirus/genética , Infecções por Papillomavirus/genética , Vacinas contra Papillomavirus/administração & dosagem , Vacinas contra Papillomavirus/genética , Neoplasias do Colo do Útero/genética , Vacinas de DNA/administração & dosagem , Vacinas de DNA/genética
11.
Cell J ; 22(3): 283-292, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31863653

RESUMO

OBJECTIVE: Currently, application of oncolytic-virus in cancer treatment of clinical trials are growing. Oncolytic-reovirus is an attractive anti-cancer therapeutic agent for clinical testing. Many studies used mesenchymal stem cells (MSCs) as a carrier cell to enhance the delivery and quality of treatment with oncolytic-virotherapy. But, biosynthetic capacity and behavior of cells in response to viral infections are different. The infecting process of reoviruses takes from two-hours to one-week, depends on host cell and the duration of different stages of virus replication cycle. The latter includes the binding of virus particle, entry, uncoating, assembly and release of progeny-viruses. We evaluated the timing and infection cycle of reovirus type-3 strain Dearing (T3D), using one-step replication experiment by molecular and conventional methods in MSCs and L929 cell as control. MATERIALS AND METHODS: In this experimental study, L929 and adipose-derived MSCs were infected with different multiplicities of infection (MOI) of reovirus T3D. At different time points, the quantity of progeny viruses has been measured using virus titration assay and quantitative real-time polymerase chain reaction (qRT-PCR) to investigate the ability of these cells to support the reovirus replication. One-step growth cycle were examined by 50% cell culture infectious dose (CCID50) and qRT-PCR. RESULTS: The growth curve of reovirus in cells shows that MOI: 1 might be optimal for virus production compared to higher and lower MOIs. The maximum quantity of virus production using MOI: 1 was achieved at 48-hours postinfection. The infectious virus titer became stationary at 72-hours post-infection and then gradually decreased. The virus cytopathic effect was obvious in MSCs and this cells were susceptible to reovirus infection and support the virus replication. CONCLUSION: Our data highlights the timing schedule for reovirus replication, kinetics models and burst size. Further investigation is recommended to better understanding of the challenges and opportunities, for using MSCs loaded with reovirus in cancer-therapy.

12.
Arch Virol ; 164(12): 3019-3026, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31598843

RESUMO

Polyethyleneimine (PEI) is a chemical compound that used is as a carrier in gene therapy/delivery. Some studies have investigated the microbicidal potential and antiviral activity (prophylactic or therapeutic) of PEI and its derivatives. The aim of this study was to investigate the effect of branched polyethyleneimine (bPEI) on human immunodeficiency virus (HIV) replication. Infected cells were treated with bPEI for 36 hours, and the concentration of the viral protein P24 (as a virus replication marker) was determined in cell culture supernatants. This study indicated that bPEI increased HIV replication and decreased the viability of infected cells through cytotoxicity. The toxicity of bPEI its association with and cell death (apoptosis, autophagy and necrosis) have been reported in several studies. To investigate bPEI-induced cytotoxicity, we examined apoptosis and autophagy in cells treated with bPEI, and a significant increase in HIV viral load, the P24 antigen level, autophagy, and necrosis observed. Thus, treatment with bPEI leads to cytotoxicity and higher HIV virus yield.


Assuntos
Infecções por HIV/virologia , HIV/efeitos dos fármacos , Polietilenoimina/farmacologia , Replicação Viral/efeitos dos fármacos , Autofagia/efeitos dos fármacos , HIV/genética , HIV/fisiologia , Proteína do Núcleo p24 do HIV/genética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/fisiopatologia , Humanos , Polietilenoimina/química , Carga Viral/efeitos dos fármacos
13.
Viral Immunol ; 32(8): 322-334, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31483214

RESUMO

Autophagy is a finely tuned process in the regulation of innate immunity to avoid excessive inflammatory responses and inflammasome signaling. In contrast, the results of recent studies have shown that autophagy may disease-dependently contribute to the pathogenesis of liver diseases, such as fibrosis, cirrhosis, and hepatocellular carcinoma (HCC) during hepatitis B virus (HBV) infection. HBV has learned to subvert the cell's autophagic machinery to promote its replication. Given the great impact of the autophagy mechanism on the HBV infection and HCC, recognizing these factors may be offered new hope for human intervention and treatment of chronic HBV. This review focuses on recent findings viewing the dual role of autophagy plays in the pathogenesis of HBV infected hepatocytes.


Assuntos
Autofagia , Carcinoma Hepatocelular/patologia , Vírus da Hepatite B/patogenicidade , Hepatite B/patologia , Neoplasias Hepáticas/patologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Autofagia/efeitos dos fármacos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/virologia , Morte Celular/efeitos dos fármacos , Citocinas/metabolismo , Estresse do Retículo Endoplasmático , Hepatite B/tratamento farmacológico , Hepatite B/virologia , Vírus da Hepatite B/fisiologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/virologia , Transdução de Sinais , Replicação Viral
14.
Biomolecules ; 9(10)2019 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-31557936

RESUMO

Autophagy modulation is considered to be a promising programmed cell death mechanism to prevent and cure a great number of disorders and diseases. The crucial step in designing an effective therapeutic approach is to understand the correct and accurate causes of diseases and to understand whether autophagy plays a cytoprotective or cytotoxic/cytostatic role in the progression and prevention of disease. This knowledge will help scientists find approaches to manipulate tumor and pathologic cells in order to enhance cellular sensitivity to therapeutics and treat them. Although some conventional therapeutics suffer from poor solubility, bioavailability and controlled release mechanisms, it appears that novel nanoplatforms overcome these obstacles and have led to the design of a theranostic-controlled drug release system with high solubility and active targeting and stimuli-responsive potentials. In this review, we discuss autophagy modulators-related signaling pathways and some of the drug delivery strategies that have been applied to the field of therapeutic application of autophagy modulators. Moreover, we describe how therapeutics will target various steps of the autophagic machinery. Furthermore, nano drug delivery platforms for autophagy targeting and co-delivery of autophagy modulators with chemotherapeutics/siRNA, are also discussed.


Assuntos
Antineoplásicos/farmacologia , Autofagia/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Animais , Proliferação de Células/efeitos dos fármacos , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Transdução de Sinais/efeitos dos fármacos
15.
IUBMB Life ; 71(1): 41-44, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30290082

RESUMO

Autophagy provides an initial membranous platform for incoming hepatitis C virus (HCV) RNA translation and immune evasion. Once HCV replication is established, this infrastructure will be unnecessary for translation of HCV RNA progeny. So, the autophagy plays key role in the replication and immune pathogenesis of HCV virus. The aim of this study was to study the effect of autophagy induction in Huh7.5 cell on virus titer. The Huh7.5 cell was transfected with recombinant pcDNA-Beclin1. The autophagy induction was evaluated via microtubule associated protein 1 light chain 3 staining as autophagy formation marker using flow cytometry. The HCV (JFH1) was inoculated 12-h post-transfection. Next, to evaluate the viral load, viral RNA was extracted after 24 and 48 h and virus titer was calculated using real-time PCR. The result of the current study shows that the induction of autophagy before virus infection was able to enhance virus yield from 4 × 103 copies/mL to 1 × 104 copies/mL at 24-h post-infection, but reduced viral load after 48 h up to 6 × 103 copies/mL. The study of cross-talk between autophagy and HCV may bring new hope for human intervention and treatment of HCV. Also, it opens new avenue to improve virus cultivation in cell culture and understanding HCV and host cell responses. © 2018 IUBMB Life, 71(1):41-44, 2019.


Assuntos
Autofagia/genética , Hepacivirus/genética , Hepatite C/genética , RNA Viral/genética , Proteína Beclina-1/genética , Linhagem Celular Tumoral , Citometria de Fluxo , Hepacivirus/patogenicidade , Hepatite C/virologia , Hepatócitos/patologia , Hepatócitos/virologia , Humanos , Transfecção , Carga Viral/genética , Replicação Viral/genética
16.
Drug Deliv Transl Res ; 8(1): 12-20, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29063498

RESUMO

Influenza virus causes a highly contagious viral respiratory tract infection with potentially fatal outcomes in humans and animals. There is now widespread influenza virus resistance to commercial drugs due to the genetic diversity of virus. Therefore, new therapeutic formulation needs to be developed. Chitosan/siRNA nanoparticles were generated as a new therapeutic approach against influenza virus infections both in vitro and in vivo. Designed siRNA against influenza nucleoprotein was formulated in chitosan polymer as siRNA/chitosan nanoparticle complex. Particle size and zeta potential of the nanoparticles were measured by dynamic light scattering. The uptake of labeled siRNA into Vero cells was visualized using fluorescence microscopy. Nanoparticle-mediated knockdown of enhanced green fluorescent protein (EGFP) was analyzed and quantified by flow cytometry in Vero cells. Results of the in vitro study showed that chitosan/siRNA nanoparticle was efficiently uptaken by Vero cells, leading to inhibition of influenza virus replication. Furthermore, nasal delivery of siRNA by chitosan nanoparticle complex has antiviral effects and significantly protected BALB/c mice from a lethal influenza challenge. These findings suggest that chitosan nanoparticle equipped with siRNA is a promising system for controlling influenza virus infection.


Assuntos
Quitosana/administração & dosagem , Nanopartículas/administração & dosagem , Nucleoproteínas/genética , Infecções por Orthomyxoviridae/prevenção & controle , RNA Interferente Pequeno/administração & dosagem , Proteínas do Core Viral/genética , Animais , Quitosana/química , Chlorocebus aethiops , Feminino , Proteínas de Fluorescência Verde/genética , Humanos , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/fisiologia , Camundongos Endogâmicos BALB C , Nanopartículas/química , Infecções por Orthomyxoviridae/virologia , RNA Interferente Pequeno/química , Células Vero , Replicação Viral/efeitos dos fármacos
17.
Bioimpacts ; 7(2): 109-114, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28752075

RESUMO

Introduction: Chronic myelogenous leukemia (CML) is a myeloproliferative disorder caused by the Philadelphia chromosome translocation, at (9; 22), which results in BCR-ABL fusion tyrosine kinase oncoprotein. This fusion induces down-regulation of miR-155. Upregulation of miR-155 can influence cell fate via the effect on p27kip1 and apoptosis. The aim of this study was to induce apoptosis in K562 CML cell line by overexpression of miR-155. Methods: The K562 cell line was transfected with pLenti-III-pre mir155-GFP constructs through electroporation. Then, overexpression of miR-155 as well as the expression level of p27kip1 and c-Myc was analyzed by quantitative PCR (qPCR). The level of p27 (Kip1) protein expression was measured by Western blot and the Annexin V method was carried out to investigate apoptosis. Results: Flow cytometric analysis results of K562 cells transfected with pLenti-III-pre mir155-GFP construct showed a significant increase in cell apoptosis. Gene expression and protein level of p27kip1 were upregulated. However, there was no change in c-Myc expression profile. Conclusion: miR-155 could be a promising approach to aid in the treatment of CML. However, further studies are required in this respect.

18.
Pathog Dis ; 75(2)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28175274

RESUMO

HIV1-Tat-based vaccines could elicit broad, durable and neutralizing immune responses and are considered as potential AIDS vaccines. The present study aims to formulate CpG-ODNs adjuvant and Chitosan with Tat protein to enhance the immunogenicity of HIV-1-Tat-based candidate vaccines and to investigate their efficacies in mice. To this end, we added CpG-ODNs, Chitosan and Alum as adjuvants to the Tat-based candidate vaccine formulations. Then, we compared frequency and magnitude of both humoral and cellular immune responses from mice immunized with the adjuvant-formulated Tat candidate vaccines against those obtained from mice immunized with recombinant Tat protein alone. Mice were subcutaneously immunized three times at 2-week intervals with the candidate vaccines. Measurements of anti-Tat immune responses showed that all vaccinated groups had a good immunity compared to the control groups and developed high levels of both humoral and cellular responses. However, immunized mice with CpG-ODNs, and Chitosan-adjuvanted Tat vaccines elicited stronger T-cell responses (both humoral and cellular immunity) compared to the others. These data suggest that co-administration of recombinant Tat protein with CpG-ODNs and Chitosan may serve as a potential formulation for enhancing of the Tat vaccine-induced immunity and might have effects on shaping Th polarization induced by HIV1-Tat protein vaccines.


Assuntos
Adjuvantes Imunológicos , Quitosana , Infecções por HIV/imunologia , HIV-1/imunologia , Oligodesoxirribonucleotídeos , Vacinas contra a AIDS/imunologia , Animais , Quitosana/imunologia , Citotoxicidade Imunológica , Modelos Animais de Doenças , Feminino , Anticorpos Anti-HIV/sangue , Anticorpos Anti-HIV/imunologia , Infecções por HIV/prevenção & controle , Imunização , Interferon gama/biossíntese , Ativação Linfocitária , Linfócitos/imunologia , Linfócitos/metabolismo , Camundongos , Oligodesoxirribonucleotídeos/imunologia , Proteínas Recombinantes/imunologia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/imunologia
19.
Mech Ageing Dev ; 163: 40-45, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28043814

RESUMO

Autophagy is a cellular homeostatic process whereby damaged proteins and organelles are encapsulated into double membrane vesicles, called autophagosomes, for lysosomal digestion. Beclin1 plays a key role in the initial steps of autophagosome formation. In this study, we evaluated the effect of Beclin 1 overexpression in induction of autophagy and the relationship between autophagy induction and telomerase activity in HeLa cells. We found that overexpression of Beclin 1 in HeLa cells leads to autophagosome formation as shown by intracellular autophagosomal marker LC3-II staining. Expression of Beclin1 reduced telomerase activity for about 100 fold compared with the control while it did not affect TERT expression level. The results of cell cycle analysis indicated that the cell cycle and proliferation progressed normally up to 48h post-transfection. Understanding the role of autophagy induction and telomerase in the pathophysiology of aging and human cancer reveal new strategies that hold much promise for intervention and therapeutic uses.


Assuntos
Autofagossomos/metabolismo , Autofagia , Proteína Beclina-1/biossíntese , Proteínas Associadas aos Microtúbulos/metabolismo , Telomerase/metabolismo , Proteína Beclina-1/genética , Células HeLa , Humanos , Proteínas Associadas aos Microtúbulos/genética , Telomerase/genética
20.
AIMS Microbiol ; 3(4): 899-907, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-31294196

RESUMO

Throughout human history, the human-beings have been used different types of plants as antimicrobial agents in fight against infectious diseases. Influenza virus is one of the most common causes of respiratory infection and transmitted through direct contact with flu infected individuals and contaminated substances or droplets. In the current study, both oil-in-water and water-in-oil emulsions with hydroalcoholic extract of eucalyptus leaves (OLHE) were developed and their antiviral efficiency was evaluated. To doing so, Madin-Darbey Canine Kidney (MDCK) cells were treated with effective minimal cytotoxic concentration of the formulated emulsions. The treated cells were then infected with 50% cell culture infectious dose (100 CCID50) of the A/H1N1 virus (the swine flu). The viral titers were measured by hemagglutination (HA) and cell culture infectious dose 50% (CCID50) assays. Also, to check the virus binding inhibition via the formulated extract, the viruses were incubated with the formulated extracts. Our study showed that the oil-in-water emulsions formulated with 2% eucalyptus leaves extract inhibited virus replication completely when the cells were infected by 100 CCID50 and decreased HA titer up to four fold. Therefore, this formulation, may hold promising application to prevent influenza virus transmission through direct contact among children and passengers.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA