Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Braz. arch. biol. technol ; 65: e22210032, 2022. tab, graf
Article in English | LILACS | ID: biblio-1364475

ABSTRACT

Abstract Zika fever is a viral infection of great relevance in public health, especially in tropic regions, in which there is a predominance of mosquitoes of the genus Aedes, vectors of the disease. Microcephaly in neonatal children and Guillain-Barré syndrome in adults can be caused by the action of the Zika virus (ZIKV). Non-structural proteins, such as NS2B, NS3 and NS5, are important pharmacological targets, due to their action in the life cycle. The absence of anti-Zika drugs raises new research, including prospecting for natural products. This work investigated the in silico antiviral activity of bixin and six other derived molecules against the Zika viral proteins NS2B-NS3 and NS5. The optimized structure was subjected to molecular docking to characterize the interaction between bixinoids and ZIKV non-structural proteins, where significant interactions were observed with amino acid residues in the catalytic site in each enzyme. These results suggest that bixin and ethyl bixin has the potential to interfere with the enzymatic activity of NS2B, NS3 and NS5, thus being an indication of being a promising anti-Zika agent.


Subject(s)
Antiviral Agents/therapeutic use , Plant Extracts/therapeutic use , Bixa orellana/therapeutic use , Zika Virus Infection/drug therapy , Phytotherapy , Virus Replication/drug effects
2.
Protein & Cell ; (12): 717-733, 2021.
Article in English | WPRIM | ID: wpr-888715

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic is caused by infection with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is spread primary via respiratory droplets and infects the lungs. Currently widely used cell lines and animals are unable to accurately mimic human physiological conditions because of the abnormal status of cell lines (transformed or cancer cells) and species differences between animals and humans. Organoids are stem cell-derived self-organized three-dimensional culture in vitro and model the physiological conditions of natural organs. Here we showed that SARS-CoV-2 infected and extensively replicated in human embryonic stem cells (hESCs)-derived lung organoids, including airway and alveolar organoids which covered the complete infection and spread route for SARS-CoV-2 within lungs. The infected cells were ciliated, club, and alveolar type 2 (AT2) cells, which were sequentially located from the proximal to the distal airway and terminal alveoli, respectively. Additionally, RNA-seq revealed early cell response to virus infection including an unexpected downregulation of the metabolic processes, especially lipid metabolism, in addition to the well-known upregulation of immune response. Further, Remdesivir and a human neutralizing antibody potently inhibited SARS-CoV-2 replication in lung organoids. Therefore, human lung organoids can serve as a pathophysiological model to investigate the underlying mechanism of SARS-CoV-2 infection and to discover and test therapeutic drugs for COVID-19.


Subject(s)
Humans , Adenosine Monophosphate/therapeutic use , Alanine/therapeutic use , Alveolar Epithelial Cells/virology , Antibodies, Neutralizing/therapeutic use , COVID-19/virology , Down-Regulation , Drug Discovery , Human Embryonic Stem Cells/metabolism , Immunity , Lipid Metabolism , Lung/virology , RNA, Viral/metabolism , SARS-CoV-2/physiology , Virus Replication/drug effects
3.
Gac. méd. Méx ; 156(6): 580-585, nov.-dic. 2020. graf
Article in Spanish | LILACS | ID: biblio-1249970

ABSTRACT

Resumen El virus SARS-CoV-2 ha sido identificado como el agente patológico causante de la pandemia de COVID-19. Aun cuando no se cuenta con un tratamiento estándar, se han probado antivirales como remdesivir y otros fármacos como cloroquina e ivermectina, que interfieren con la replicación del virus. También se han intentado algunas estrategias encaminadas a disminuir los mecanismos inmunitarios, como el uso de tocilizumab y antioxidantes naturales. Los fármacos relacionados con el sistema renina-angiotensina han resultado controversiales. Aún se debe estudiar con detalle los mecanismos de patogenicidad, así como los tratamientos controlados para proponer alguna opción terapéutica viable que evite la entrada y replicación del virus o que aumente los sistemas inmunitarios del huésped.


Abstract SARS-CoV-2 virus has been identified as the causative agent of the COVID-19 pandemic. Even when no standard treatment is available, antivirals such as remdesivir and other drugs such as chloroquine and ivermectin, which interfere with viral replication, have been assayed. Some strategies aimed to reduce immune mechanisms, such as the use of tocilizumab and natural antioxidants, have also been tested. The use of drugs related to the renin-angiotensin system has been controversial. Pathogenicity mechanisms, as well as controlled treatments, still have to be studied in detail in order to propose a viable therapeutic option that prevents the entry and replication of the virus or enhances the host immune system.


Subject(s)
Humans , Animals , Antiviral Agents/administration & dosage , COVID-19/drug therapy , Antiviral Agents/pharmacology , Virus Replication/drug effects , Virus Internalization/drug effects , SARS-CoV-2/isolation & purification , SARS-CoV-2/drug effects , COVID-19/virology
4.
Rev. Soc. Bras. Med. Trop ; 52: e20180441, 2019.
Article in English | LILACS | ID: biblio-1041501

ABSTRACT

Abstract Hepatitis B is a major public health problem worldwide and associated with significant mortality. To prevent or delay the deleterious effects of chronic infection by the hepatitis B virus, patients should be carefully followed, and antiviral therapy indicated according to specific recommendations. Currently, available drugs inhibit viral replication and slow or stop the progression of inflammation and fibrosis of the liver. However, the drugs for oral use in the treatment of hepatitis B, jointly referred to as nucleoside/nucleotide analogs, are indicated for prolonged use and have potential side effects. The reduction in bone mineral density was associated with the use of tenofovir, already evaluated in patients infected with HIV because the drug is also part of the therapeutic arsenal for this viral infection. There are few studies on the effects of tenofovir in patients with mono hepatitis B. Therefore, this literature review proposes to examine how hepatitis B acts in the body and the mechanisms by which antiretroviral drugs (especially tenofovir) can affect bone metabolism.


Subject(s)
Humans , Bone Density/drug effects , Bone Remodeling/drug effects , Hepatitis B, Chronic/drug therapy , Anti-Retroviral Agents/adverse effects , Virus Replication/drug effects , Anti-Retroviral Agents/administration & dosage
5.
Braz. j. microbiol ; 49(4): 785-789, Oct.-Dec. 2018. tab, graf
Article in English | LILACS | ID: biblio-974292

ABSTRACT

ABSTRACT Bovine viral diarrhea virus can cause acute disease in livestock, leading to economic losses. We show that Prostaglandin A1 inhibits bovine viral diarrhea virus replication in Madin-Darby bovine kidney cells (94% inhibition using 5 µg/mL). Light and electron microscopy of infected cells shows that Prostaglandin A1 also prevents virus-induced vacuolization, but at higher concentrations (10 µg/mL).


Subject(s)
Animals , Cattle , Antiviral Agents/pharmacology , Prostaglandins A/pharmacology , Bovine Virus Diarrhea-Mucosal Disease/virology , Diarrhea Viruses, Bovine Viral/drug effects , Antiviral Agents/analysis , Prostaglandins A/analysis , Virus Replication/drug effects , Bovine Virus Diarrhea-Mucosal Disease/drug therapy , Cell Line , Diarrhea Viruses, Bovine Viral/physiology , Diarrhea Viruses, Bovine Viral/genetics , Diarrhea
6.
Braz. j. infect. dis ; 22(6): 477-486, Nov.-Dec. 2018. tab, graf
Article in English | LILACS | ID: biblio-984016

ABSTRACT

ABSTRACT Antiviral drug resistance is the most important factor contributing to treatment failure using nucleos(t)ide analogs such as lamivudine for chronic infection with hepatitis B virus (HBV). Development of a system supporting efficient replication of clinically resistant HBV strains is imperative, and new antiviral drugs are needed urgently to prevent selection of drug-resistant HBV mutants. A novel fluorinated cytidine analog, NCC (N-cyclopropyl-4′-azido-2′-deoxy-2′-fluoro-β-d-cytidine), was recently shown to strongly inhibit human HBV in vitro and in vivo. This study was designed to evaluate the antiviral activity of NCC against lamivudine-resistant HBV. We generated a stable cell line encoding the major pattern of lamivudine-resistant mutations rtL180M/M204V and designated it "HepG2.RL1". Immuno-transmission electron microscopic examination and enzyme-linked immunosorbent assay were used to detect secretion of HBV-specific particles and antigens. Quantification of extracellular DNA and intracellular DNA of HepG2.RL1 cells by quantitative real-time polymerase chain reaction revealed >625-fold and >5556-fold increases in the 50% inhibitory concentration of lamivudine, respectively, compared with that for the wild-type virus. The results showed that NCC inhibited DNA replication and HBeAg production in wild-type or lamivudine-resistant HBV in a dose-dependent manner. In conclusion, screening for antiviral compounds active against lamivudine-resistant HBV can be carried out with relative ease using hepG2.RL1 cells. NCC is a potential antiviral agent against wild-type HBV and clinical lamivudine-resistant HBV and deserves evaluation for the treatment of HBV infection.


Subject(s)
Humans , Female , Middle Aged , Antiviral Agents/pharmacology , Virus Replication/drug effects , Hepatitis B virus/drug effects , Lamivudine/pharmacology , Cytidine/analogs & derivatives , DNA, Viral/chemistry , Microbial Sensitivity Tests , Cell Line , Hepatitis B virus/isolation & purification , Hepatitis B virus/physiology , Hepatocytes/virology , Drug Resistance, Viral/drug effects , Mutation
7.
Rev. Soc. Bras. Med. Trop ; 51(5): 584-590, Sept.-Oct. 2018. tab, graf
Article in English | LILACS | ID: biblio-957459

ABSTRACT

Abstract INTRODUCTION: The Mayaro virus (MAYV), which is an arbovirus closely related to the Chikungunya virus, causes a dengue-like acute illness that is endemic to Central and South America. We investigated the anti-MAYV activity of prostaglandin A1 (PGA1), a hormone which exhibits antiviral activity against both ribonucleic acid (RNA) and deoxyribonucleic acid (DNA) viruses. Further, we examined the effects of inducting the stress protein HSP70 following PGA1 treatment. METHODS: Hep-2 cells infected with MAYV were treated with PGA1 (0.1-6μg/ml) 12h before infection and for different periods post-infection. Inhibition of viral replication inhibition was analyzed via viral titer determination, whereas the effect of PGA1 on viral morphogenesis was examined via transmission electron microscopy (TEM). Autoradiography (with 35S methionine labeling) and western blotting were used to assess the effect of PGA1 treatment on viral and cellular protein synthesis, and on HSP70 induction, respectively. RESULTS: PGA1 strongly reduced viral replication in Hep-2 cells, particularly when added during the early stages of viral replication. Although PGA1 treatment inhibited viral replication by 95% at 24 hours post-infection (hpi), viral structural protein synthesis was inhibited only by 15%. TEM analysis suggested that PGA1 inhibited replication before viral morphogenesis. Western blot and densitometry analyses showed that PGA1 treatment increased HSP70 protein levels, although this was not detectable via autoradiography. CONCLUSIONS: PGA1 inhibits MAYV replication in Hep-2 cells at early stages of viral replication, prior to production of viral structural proteins, possibly via HSP70 induction.


Subject(s)
Humans , Animals , Cattle , Prostaglandins A/pharmacology , Virus Replication/drug effects , Alphavirus/drug effects , HSP70 Heat-Shock Proteins/pharmacology , Epithelial Cells/virology , Antiviral Agents/pharmacology , Cell Line , Blotting, Western , Alphavirus/ultrastructure , Microscopy, Electron, Transmission , Electrophoresis, Polyacrylamide Gel , Epithelial Cells/ultrastructure
8.
Braz. j. med. biol. res ; 51(7): e7220, 2018. tab, graf
Article in English | LILACS | ID: biblio-889115

ABSTRACT

An abnormality in the Lin28/let-7a axis is relevant to the progression of hepatitis B virus (HBV)-positive hepatocellular carcinoma (HCC), which could be a novel therapeutic target for this malignant tumor. The present study aimed to investigate the antiproliferative and anti-invasive effects of urolithin A in a stable full-length HBV gene integrated cell line HepG2.2.15 using CCK-8 and transwell assays. The RNA and protein expressions of targets were assessed by quantitative PCR and western blot, respectively. Results revealed that urolithin A induced cytotoxicity in HepG2.2.15 cells, which was accompanied by the cleavage of caspase-3 protein and down-regulation of Bcl-2/Bax ratio. Moreover, urolithin A suppressed the protein expressions of Sp-1, Lin28a, and Zcchc11, and elevated the expression of microRNA let-7a. Importantly, urolithin A also regulated the Lin28a/let-7a axis in transient HBx-transfected HCC HepG2 cells. Furthermore, urolithin A decelerated the HepG2.2.15 cell invasion, which was involved in suppressing the let-7a downstream factors HMGA2 and K-ras. These findings indicated that urolithin A exerted the antiproliferative effect by regulating the Lin28a/let-7a axis and may be a potential supplement for HBV-infected HCC therapy.


Subject(s)
Humans , RNA-Binding Proteins/drug effects , Carcinoma, Hepatocellular/drug therapy , Coumarins/pharmacology , MicroRNAs/drug effects , Liver Neoplasms/drug therapy , Reference Values , Sincalide/analysis , Time Factors , Virus Replication/drug effects , Cell Survival/drug effects , Blotting, Western , Reproducibility of Results , Analysis of Variance , RNA-Binding Proteins/analysis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , MicroRNAs/analysis , Cell Proliferation/drug effects , Hep G2 Cells , Real-Time Polymerase Chain Reaction , Liver Neoplasms/genetics , Liver Neoplasms/virology
9.
Braz. j. med. biol. res ; 50(10): e6586, 2017. tab, graf
Article in English | LILACS | ID: biblio-888930

ABSTRACT

Human enterovirus 71 (EV71) is the major causative agent of hand, foot, and mouth disease (HFMD), particularly in infants and children below 4 years of age. Shikonin is a bioactive compound with anti-inflammatory, antiviral, and antibacterial activities derived from the roots of the Chinese medicinal herb Lithospermum erythrorhizon. This study aimed to examine the antiviral activity of PMM-034, a shikonin ester derivative, against EV71 in rhabdomyosarcoma (RD) cells. Cytotoxicity of PMM-034 on RD cells was determined using WST-1 assay. Dose- and time-dependent effects of PMM-034 on EV71 replication in RD cells were determined using plaque reduction assay. mRNA expression levels of EV71/VP1 and pro-inflammatory cytokines (IL-1β, IL-6, IL-8, and TNF-α) were determined by real-time RT-PCR, and EV71/VP1 and phospho-p65 protein expressions were determined by western blot analysis. PMM-034 exhibited only weak cytotoxicity against RD cells. However, PMM-034 exhibited significant antiviral activity against EV71 in RD cells with 50% inhibitory concentration of 2.31 μg/mL. The VP1 mRNA and protein levels were significantly reduced in cells treated with PMM-034. Furthermore, relative mRNA expression levels of IL-1β, IL-6, IL-8, and TNF-α significantly decreased in the cells treated with PMM-034, while the phospho-p65 protein expression was also significantly lower in the treated cells. These results indicated that PMM-034 suppressed the expressions of pro-inflammatory cytokines in RD cells, exhibiting antiviral activity against EV71, as evidenced by the reduced VP1 mRNA and protein levels in PMM-034-treated cells. Thus, PMM-034 is a promising candidate for further development as an EV71 inhibitor.


Subject(s)
Humans , Antiviral Agents/pharmacology , Enterovirus A, Human/drug effects , Naphthoquinones/pharmacology , Rhabdomyosarcoma/virology , Blotting, Western , Cell Line, Tumor , Cytokines/analysis , Dose-Response Relationship, Drug , Real-Time Polymerase Chain Reaction , Toxicity Tests , Viral Plaque Assay , Virus Replication/drug effects
10.
Rev. bras. epidemiol ; 18(supl.2): 3-16, Out.-Dez. 2015. tab
Article in English | LILACS | ID: lil-776698

ABSTRACT

RESUMO: Objetivo: Descrever as principais doenças crônicas não transmissíveis (DCNT) no país segundo as informações coletadas em indivíduos de 18 anos ou mais de idade. Métodos: Foram utilizados dados da Pesquisa Nacional de Saúde (PNS), 2013, estudo transversal de base populacional. As proporções de cada DCNT foram calculadas e apresentadas segundo sexo, com intervalo de confiança de 95% (IC95%), com os valores absolutos. Resultados: Do total de entrevistados, 45,1% referiram ter pelo menos uma DCNT. A Região com maior prevalência de DCNT foi a Sul (52,1%). A hipertensão arterial apresentou a maior prevalência dentre as DCNT, com 21,4%, seguida por problema crônico de coluna (18,5%), depressão (7,6%), artrite (6,4%) e diabetes (6,2%). O grau de limitação intenso/muito intenso apresentou maiores prevalências para outra doença mental (37,6%) e acidente vascular cerebral (AVC) (25,5%). Conclusão: A melhoria dos serviços de saúde é indispensável para uma resposta efetiva à dupla carga de adoecimento de países de média e baixa renda.


ABSTRACT: Objective: To describe the major noncommunicable diseases (NCDs) in Brazil, according to the information collected from individuals aged 18 years or older. Methods: Data from the National Health Survey (PNS), 2013, a transversal population-based study, were used. The proportions of each NCD were calculated and presented according to sex, with a 95% confidence interval (95%CI), with the absolute values. Results: Of the total respondents, 45.1% reported presenting at least one NCD. The region with the highest prevalence of NCDs was the South (52.1%). Hypertension showed the highest prevalence among NCDs, with 21.4%, followed by chronic back problem (18.5%), depression (7.6%), arthritis (6.4%), and diabetes (6.2%). The intense/very intense degree of limitation showed a higher prevalence of other mental illnesses (37.6%) and cerebrovascular accident (25.5%). Conclusion: The improvement of health services is essential for an effective response to the double burden of illness in the middle- and low-income countries.


Subject(s)
Humans , Antiviral Agents/pharmacology , /metabolism , Enterovirus A, Human/drug effects , Enterovirus Infections/genetics , Isoflavones/pharmacology , Prostaglandins E/metabolism , Virus Replication/drug effects , /genetics , Enterovirus A, Human/physiology , Enterovirus Infections/enzymology , Enterovirus Infections/metabolism , Enterovirus Infections/virology
11.
Braz. j. infect. dis ; 19(5): 453-458, tab, graf
Article in English | LILACS | ID: lil-764500

ABSTRACT

ABSTRACTWe have evaluated the efficacy of short-interfering RNAs targeting the nucleoprotein gene and also the brain immune response in treated and non-treated infected mice. Mice were inoculated with wild-type virus, classified as dog (hv2) or vampire bat (hv3) variants and both groups were treated or leaved as controls. No difference was observed in the lethality rate between treated and non-treated groups, although clinical evaluation of hv2 infected mice showed differences in the severity of clinical disease (p = 0.0006). Evaluation of brain immune response 5 days post-inoculation in treated hv2 group showed no difference among the analyzed genes, whereas after 10 days post-inoculation there was increased expression of 2',5'-oligoadenylate synthetase 1, tumor necrosis factor alpha, interleukin 12, interferon gamma, and C-X-C motif chemokine 10 associated with higher expression of Ngene in the same period (p < 0.0001). In hv2 non-treated group only higher interferon beta expression was found at day 5. The observed differences in results of the immune response genes between treated and non-treated groups is not promising as they had neither impact on mortality nor even a reduction in the expression of N gene in siRNA treated animals. This finding suggests that the use of pre-designed siRNA alone may not be useful in rabies treatment.


Subject(s)
Animals , Dogs , Female , Humans , Mice , Antiviral Agents/administration & dosage , Chiroptera/virology , RNA, Small Interfering/administration & dosage , Rabies virus/drug effects , Rabies/therapy , Brain/immunology , Cell Line , Disease Models, Animal , Reverse Transcriptase Polymerase Chain Reaction , Rabies virus/immunology , Rabies/virology , Virus Replication/drug effects , Virus Replication/genetics
12.
Braz. j. microbiol ; 45(3): 1089-1094, July-Sept. 2014. ilus, tab
Article in English | LILACS | ID: lil-727042

ABSTRACT

P34 is an antimicrobial peptide produced by a Bacillus sp. strain isolated from the intestinal contents of a fish in the Brazilian Amazon basin with reported antibacterial activity. The aim of this work was to evaluate the peptide P34 for its in vitro antiviral properties against canine adenovirus type 2 (CAV-2), canine coronavirus (CCoV), canine distemper virus (CDV), canine parvovirus type 2 (CPV-2), equine arteritis virus (EAV), equine influenza virus (EIV), feline calicivirus (FCV) and feline herpesvirus type 1 (FHV-1). The results showed that the peptide P34 exhibited antiviral activity against EAV and FHV-1. The peptide P34 inhibited the replication of EAV by 99.9% and FHV-1 by 94.4%. Virucidal activity was detected only against EAV. When P34 and EAV were incubated for 6 h at 37 °C the viral titer reduced from 10(4.5) TCID50 to 10(2.75) TCID50, showing a percent of inhibition of 98.6%. In conclusion, our results demonstrated that P34 inhibited EAV and FHV-1 replication in infected cell cultures and it showed virucidal activity against EAV. Since there is documented resistance to the current drugs used against herpesviruses and there is no treatment for equine viral arteritis, it is advisable to search for new antiviral compounds to overcome these infections.


Subject(s)
Animals , Animals, Domestic/virology , Antimicrobial Cationic Peptides/pharmacology , Antiviral Agents/pharmacology , Bacillus/metabolism , Viruses/drug effects , Antimicrobial Cationic Peptides/isolation & purification , Antiviral Agents/isolation & purification , Brazil , Bacillus/isolation & purification , Bacterial Proteins/isolation & purification , Bacterial Proteins/pharmacology , Fishes/microbiology , Gastrointestinal Tract/microbiology , Microbial Viability/drug effects , Temperature , Time Factors , Viral Load , Virus Replication/drug effects
13.
Gut and Liver ; : 471-479, 2014.
Article in English | WPRIM | ID: wpr-108135

ABSTRACT

The combination of pegylated interferon (PEG-IFN) and ribavirin (RBV), the current therapy for hepatitis C virus (HCV) infection, has saved the lives of many HCV-infected patients. Direct-acting antivirals (DAAs) target several sites of HCV nonstructural proteins, resulting in the cessation of viral replication. The first NS3/4A protease inhibitors consisted of boceprevir and telaprevir, which have shown superior efficacy against genotype 1 HCV infection when combined with PEG-IFN/RBV compared with the standard therapy in both treatment-naive and -experienced patients. Simeprevir, faldaprevir, and asunaprevir are second-wave, first-generation NS3/4A inhibitors that have already been or will soon be approved. Second-generation protease inhibitors are in clinical trials. Daclatasvir is the first approved DAA belonging to the class of NS5A replication complex inhibitors. The potency of daclatasvir is very high, and this drug is an important and essential component of combination regimens for all genotypes. Sofosbuvir, the first approved NS5B polymerase inhibitor, is characterized by high potency and genetic barriers to resistance. Sofosbuvir combined with RBV achieved an interferon-free regimen in genotype 2 or 3 patients with a reduced treatment duration. It can also be used in combination with PEG-IFN/RBV in genotype 1 patients for 12 weeks. DAAs have provided new hope for curing HCV infections with a short treatment duration and acceptable adverse events.


Subject(s)
Humans , Antiviral Agents/therapeutic use , Drug Therapy, Combination , Hepacivirus/drug effects , Hepatitis C/drug therapy , Protease Inhibitors/therapeutic use , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects
14.
Braz. j. microbiol ; 44(3): 879-882, July-Sept. 2013. tab
Article in English | LILACS | ID: lil-699783

ABSTRACT

Rabies is a zoonotic disease that affects all mammals and leads to more than 55,000 human deaths every year, caused by rabies virus (RABV) (Mononegavirales: Rhabdoviridae: Lyssavirus). Currently, human rabies treatment is based on the Milwaukee Protocol which consists on the induction of coma and massive antiviral therapy. The aim of this study was to assess the decrease in the titer of rabies virus both in vitro and in vivo using short-interfering RNAs. To this end, three siRNAs were used with antisense strands complementary to rabies virus nucleoprotein (N) mRNA. BHK-21 cells monolayers were infected with 1000 to 0.1 TCID50 of PV and after 2 hours the cells were transfected with each of tree RNAs in separate using Lipofectamine-2000. All three siRNAs reduced the titer of PV strain in a least 0.72 logTCID50/mL and no cytotoxic effect was observed in the monolayers treated with Lipofectamine-2000. Swiss albino mice infected with 10.000 to 1 LD of PV strain by the intracerebral route were also transfected after two hours of infection with a pool 3 siRNAs with Lipofectamine-2000 by the intracerebral route, resulting in a survival rate of 30% in mice inoculated with 100 LD50, while the same dose led to 100% mortality in untreated animals. Lipofectamine-2000 showed no toxic effect in control mice. These results suggest that intracerebral administration of siRNAs might be an effective antiviral strategy for rabies.


Subject(s)
Animals , Cricetinae , Mice , Antiviral Agents/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rabies virus/drug effects , Rabies virus/physiology , Rabies/drug therapy , Virus Replication/drug effects , Cell Line , Disease Models, Animal , Nucleocapsid Proteins/antagonists & inhibitors , RNA, Small Interfering/genetics , Survival Analysis , Viral Load , Virus Cultivation
15.
Clinical and Molecular Hepatology ; : 195-209, 2013.
Article in English | WPRIM | ID: wpr-202394

ABSTRACT

Although much advancement has been achieved in the treatment of chronic hepatitis B, antiviral resistance is still a challenging issue. Previous generation antiviral agents have already developed resistance in a number of patients, and it is still being used especially in resource limited countries. Once antiviral resistance occurs, it predisposes to subsequent resistance, resulting in multidrug resistance. Therefore, prevention of initial antiviral resistance is the most important strategy, and appropriate choice and modification of therapy would be the cornerstone in avoiding treatment failures. Until now, management of antiviral resistance has been evolving from sequential therapy to combination therapy. In the era of tenofovir, the paradigm shifts again, and we have to decide when to switch and when to combine on the basis of newly emerging clinical data. We expect future eradication of chronic hepatitis B virus infection by proper prevention and optimal management of antiviral resistance.


Subject(s)
Humans , Adenine/analogs & derivatives , Antiviral Agents/pharmacology , Drug Resistance, Viral/drug effects , Drug Therapy, Combination , Hepatitis B virus/genetics , Hepatitis B, Chronic/drug therapy , Mutation , Nucleosides/chemistry , Organophosphonates/pharmacology , Virus Replication/drug effects
16.
Braz. j. infect. dis ; 16(2): 129-135, May-Apr. 2012. ilus, tab
Article in English | LILACS | ID: lil-622732

ABSTRACT

BACKGROUND: Due to the emergence of drug resistance in herpes simplex virus type 1 (HSV-1), researchers are trying to find other methods for treating herpes simplex virus type 1 infections. Probiotic bacteria are effective in macrophage activation and may have antiviral activities. OBJECTIVE: This study aimed at verifying the direct effect of Lactobacillus rhamnosus, a probiotic bacterium, in comparison with Escherichia coli, a non-probiotic one, on HSV-1 infection, and determining its effect on macrophage activation for in vitro elimination of HSV-1 infection. METHODS: The above bacteria were introduced into HSV-1 infected Vero cells, and their effects were examined using both MTT and plaque assay. To determine macrophage activation against in vitro HSV-1 infection, J774 cells were exposed to these bacteria; then, macrophage viability was examined with the MTT method, and tumor necrosis factor alpha (TNF-α), interferon-gamma (IFN-γ), and nitric oxide (NO) assessments were performed using the ELISA method. RESULTS: A significant increased viability of macrophages was observed (p < 0.05) in the presence of Lactobacillus rhamnosus before and after HSV-1 infection when compared with Escherichia coli as a non-probiotic bacterium. However, tumor necrosis factor α concentration produced by Escherichia coli-treated J774 cells was significantly higher than Lactobacillus rhamnosus-treated J774 cells (p < 0.05). interferon-gamma and NO production were not different in the groups treated with Escherichia coli or with Lactobacillus rhamnosus. CONCLUSION: The results of this study indicate that Lactobacillus rhamnosus enhances macrophage viability for HSV-1 elimination and activation against HSV-1 more effectively, when compared with non-probiotic Escherichia coli. it also seems that receptor occupation of macrophage sites decreases HSV-1 infectivity by both of the studied bacteria.


Subject(s)
Humans , Escherichia coli/physiology , Herpesvirus 1, Human , Lacticaseibacillus rhamnosus/chemistry , Probiotics/pharmacology , Cell Line , Interferon-gamma/analysis , Lacticaseibacillus rhamnosus/physiology , Macrophage Activation/drug effects , Nitric Oxide/analysis , Tumor Necrosis Factor-alpha/analysis , Virus Replication/drug effects
17.
Invest. clín ; 53(1): 71-83, mar. 2012. ilus, tab
Article in Spanish | LILACS | ID: lil-664567

ABSTRACT

La infección por VIH (virus de la inmunodeficiencia humana) en la actualidad es un grave problema de salud pública a nivel mundial, que requiere de nuevas estrategias vacunales para detener su propagación así como para su efectivo tratamiento. Algunos estudios relacionados con la inmunidad innata en contra de VIH, han demostrado que los péptidos antimicrobianos (AMP´s) pueden generar resistencia a las infecciones virales. En la presente revisión, se describen a los péptidos antimicrobianos de humano y su actividad en contra de VIH así como péptidos de otras especies como plantas, anfibios, insectos y varias especies de animales que poseen un potencial terapéutico o profiláctico en la infección por VIH. Se describen brevemente algunos mecanismos mediante los cuales estos péptidos pueden bloquear la replicación e infección por el VIH.


HIV (human immunodeficiency virus) infection is today a very important health issue worldwide, which demands new ways and strategies for its prevention and treatment. Several studies on the innate immunity against HIV infection have shown that antimicrobial peptides are associated with increased resistance to infection. In the present review, we briefly summarize the major characteristics of antimicrobial peptides from human and several species of plants, amphibians, insects and other animal species that have significant potential to be used as therapeutic or prophylactic agents. The mechanisms of infection inhibition and viral replication blockade are also described in the context of the biology of infection.


Subject(s)
Animals , Humans , Anti-HIV Agents/therapeutic use , Antimicrobial Cationic Peptides/therapeutic use , HIV Infections/drug therapy , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/pharmacology , Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Drug Discovery , Drug Evaluation, Preclinical , HIV , Invertebrates/chemistry , Plants/chemistry , Species Specificity , Vertebrates/metabolism , Virus Replication/drug effects
18.
Armaghane-danesh. 2011; 16 (2): 149-141
in Persian | IMEMR | ID: emr-129743

ABSTRACT

In the recent years, following the increase of drug resistant strain of viruses, there has been an increasing interest in the use of natural substances with antiviral activity with fewer side effects. One of these herbal medicines, Quercqus persica L, has shown some therapeutic effects, such as anti-bacterial and anti-viral activities. Therefore, this study was aimed to determine the minimum inhibitory concentration of hydroalcoholic extract of this plant on Herpes simplex virus-1 [HSV-1]. In this interventional study conducted at Shahrekord University of Medical Sciences in 2010, the hydroalcoholic extract of Quercqus persica L. was prepared using 70% ethanol by the maceration method. Baby Hamster Kidney [BHK] cells were grown in monolayer culture with Dulbecco's modified Eagle's medium [DMEM] supplemented with 5% fetal calf serum and plated into 48-well culture plates. Fifty percent of cytotoxic concentration [CC50%] of the extract on BHK cells was determined and subsequently, 50% inhibitory concentrations [IC50%] of the herbal extract on replication of HSV-1 both in extracellular and interacellular cases were assessed. The statistical data was analyzed by the SPSS software using Probit analysis. Based on Probit analysis, the extract had no cyto-toxicity up to concentration of 200 mg/ml. ICSOs of the extract on the virus before cellular attachment and after entering the cells were 1.2 and 0.257 mg/ml, respectively. Based on the model, with the increasing of the extract concentration, the percentage of inhibition of cytopathic effect [CPE] in both of the stages were increased [p<0.05]. In addition to low cytotoxicity, hydroalcoholic extract of Quercqus persica L. has promising inhibitory effects on HSV-1 replication in cell culture. Thus, it should be considered as a promising herbal medicine and should be thoroughly evaluated through a comprehensive study


Subject(s)
Animals , Plant Extracts , Herpesvirus 1, Human/drug effects , Virus Replication/drug effects , Cricetinae , Antiviral Agents
19.
The Korean Journal of Hepatology ; : 213-219, 2011.
Article in English | WPRIM | ID: wpr-194177

ABSTRACT

BACKGROUND/AIMS: JX-594 is an oncolytic virus derived from the Wyeth vaccinia strain that causes replication-dependent cytolysis and antitumor immunity. Starting with a cross-examination of clinical-trial samples from advanced hepatocellular carcinoma patients having high levels of aldosterone and virus amplification in JX-594 treatment, we investigated the association between virus amplification and aldosterone in human cancer cell lines. METHODS: Cell proliferation was determined by a cell-counting-kit-based colorimetric assay, and vaccinia virus quantitation was performed by quantitative polymerase chain reaction (qPCR) and a viral plaque assay. Also, the intracellular pH was measured using a pH-sensitive dye. RESULTS: Simultaneous treatment with JX-594 and aldosterone significantly increased viral replication in A2780, PC-3, and HepG2 cell lines, but not in U2OS cell lines. Furthermore, the aldosterone treatment time altered the JX-594 replication according to the cell line. The JX-594 replication peaked after 48 and 24 hours of treatment in PC-3 and HepG2 cells, respectively. qPCR showed that JX-594 entry across the plasma membrane was increased, however, the changes are not significant by the treatment. This was inhibited by treatment with spironolactone (an aldosterone-receptor inhibitor). JX-594 entry was significantly decreased by treatment with EIPA [5-(N-ethyl-N-isopropyl)amiloride; a Na+/H+-exchange inhibitor], but aldosterone significantly restored JX-594 entry even in the presence of EIPA. Intracellular alkalization was observed after aldosterone treatment but was acidified by EIPA treatment. CONCLUSIONS: Aldosterone stimulates JX-594 amplification via increased virus entry by affecting the H+ gradient.


Subject(s)
Animals , Humans , Rabbits , Aldosterone/pharmacology , Mineralocorticoid Receptor Antagonists/pharmacology , Amiloride/analogs & derivatives , Carcinoma, Hepatocellular/blood , Cell Line, Tumor , Hydrocortisone/blood , Hydrogen-Ion Concentration , Liver Neoplasms/blood , Neuroprotective Agents/pharmacology , Oncolytic Virotherapy , Spironolactone/pharmacology , Vaccinia virus/drug effects , Virus Replication/drug effects
20.
Mem. Inst. Oswaldo Cruz ; 105(3): 304-309, May 2010. graf, tab
Article in English | LILACS | ID: lil-547301

ABSTRACT

The inhibitory effect of Lippia alba and Lippia citriodora essential oils on dengue virus serotypes replication in vitro was investigated. The cytotoxicity (CC50) was evaluated by the MTT assay and the mode of viral inhibitory effect was investigated with a plaque reduction assay. The virus was treated with the essential oil for 2 h at 37ºC before cell adsorption and experiments were conducted to evaluate inhibition of untreated-virus replication in the presence of oil. Antiviral activity was defined as the concentration of essential oil that caused 50 percent reduction of the virus plaque number (IC50). L. alba oil resulted in less cytotoxicity than L. citriodora oil (CC50: 139.5 vs. 57.6 μg/mL). Virus plaque reduction for all four dengue serotypes was observed by treatment of the virus before adsorption on cell. The IC50 values for L. alba oil were between 0.4-32.6 μg/mL and between 1.9-33.7 μg/mL for L. citriodora oil. No viral inhibitory effect was observed by addition of the essential oil after virus adsorption. The inhibitory effect of the essential oil seems to cause direct virus inactivation before adsorption on host cell.


Subject(s)
Animals , Antiviral Agents/pharmacology , Dengue Virus/drug effects , Lippia/chemistry , Oils, Volatile/pharmacology , Plant Oils/pharmacology , Virus Replication/drug effects , Chlorocebus aethiops , Dengue Virus/growth & development , Microbial Sensitivity Tests , Vero Cells , Viral Plaque Assay
SELECTION OF CITATIONS
SEARCH DETAIL