Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
ACS Omega ; 9(1): 146-156, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38222605

ABSTRACT

There is no approved antiviral for the management of the Chikungunya virus (CHIKV). To develop an antiviral drug that can manage both CHIKV and arthritis induced by it, an ester conjugate of telmisartan (TM) and salicylic acid (SA) was synthesized (DDABT1). It showed higher potency (IC50 of 14.53 µM) and a good selectivity index [(SI = CC50/IC50) > 33]. On post-treatment of DDABT1, CHIKV infection was inhibited significantly by reducing CPE, viral titer, viral RNA, and viral proteins. Further, the time of addition experiment revealed >95% inhibition up to 4hpi indicating its interference predominantly in the early stages of infection. However, the late stages were also affected. This conjugate of SA and TM was found to increase the antiviral efficacy, and this might be partly attributed to modulating angiotensin II (Ang II) receptor type 1 (AT1). However, DDABT1 might have other modes of action that need further investigation. In addition, the in vivo experiments showed an LD50 of 5000 mg/kg in rats and was found to be more effective than TM, SA, or their combination against acute, subacute, and chronic inflammation/arthritis in vivo. In conclusion, DDABT1 showed remarkable anti-CHIKV properties and the ability to reduce inflammation and arthritis, making it a very good potential drug candidate that needs further experimental validation.

2.
Front Immunol ; 14: 1139808, 2023.
Article in English | MEDLINE | ID: mdl-37153546

ABSTRACT

Toll like receptor 4 (TLR4), a pathogen-associated molecular pattern (PAMP) receptor, is known to exert inflammation in various cases of microbial infection, cancer and autoimmune disorders. However, any such involvement of TLR4 in Chikungunya virus (CHIKV) infection is yet to be explored. Accordingly, the role of TLR4 was investigated towards CHIKV infection and modulation of host immune responses in the current study using mice macrophage cell line RAW264.7, primary macrophage cells of different origins and in vivo mice model. The findings suggest that TLR4 inhibition using TAK-242 (a specific pharmacological inhibitor) reduces viral copy number as well as reduces the CHIKV-E2 protein level significantly using p38 and JNK-MAPK pathways. Moreover, this led to reduced expression of macrophage activation markers like CD14, CD86, MHC-II and pro-inflammatory cytokines (TNF, IL-6, MCP-1) significantly in both the mouse primary macrophages and RAW264.7 cell line, in vitro. Additionally, TAK-242-directed TLR4 inhibition demonstrated a significant reduction of percent E2-positive cells, viral titre and TNF expression in hPBMC-derived macrophages, in vitro. These observations were further validated in TLR4-knockout (KO) RAW cells. Furthermore, the interaction between CHIKV-E2 and TLR4 was demonstrated by immuno-precipitation studies, in vitro and supported by molecular docking analysis, in silico. TLR4-dependent viral entry was further validated by an anti-TLR4 antibody-mediated blocking experiment. It was noticed that TLR4 is necessary for the early events of viral infection, especially during the attachment and entry stages. Interestingly, it was also observed that TLR4 is not involved in the post-entry stages of CHIKV infection in host macrophages. The administration of TAK-242 decreased CHIKV infection significantly by reducing disease manifestations, improving survivability (around 75%) and reducing inflammation in mice model. Collectively, for the first time, this study reports TLR4 as one of the novel receptors to facilitate the attachment and entry of CHIKV in host macrophages, the TLR4-CHIKV-E2 interactions are essential for efficient viral entry and modulation of infection-induced pro-inflammatory responses in host macrophages, which might have translational implication for designing future therapeutics to regulate the CHIKV infection.


Subject(s)
Chikungunya Fever , Chikungunya virus , Toll-Like Receptor 4 , Animals , Mice , Inflammation , Macrophages , Molecular Docking Simulation , Viral Envelope Proteins , Virus Replication
3.
Mol Divers ; 2023 Apr 21.
Article in English | MEDLINE | ID: mdl-37085737

ABSTRACT

Chikungunya virus infection has become a global health concern because of its high rates of morbidity and mortality in patients with preexisting conditions. Inflammation and arthritis are the major symptoms of CHIKV that persist even after clearance of CHIKV. To develop an antiviral that can reduce infection and manage inflammation independent of the CHIKV infection, ibuprofen (IBU) conjugates with sulfonamide and thiosemicarbazide were synthesized. The conjugates, IBU-SULFA, IBU-ISS and IBU-IBT significantly inhibited CHIKV infection in vitro with a selectivity index (CC50/IC50) of > 11.9, > 25.1 and > 21, respectively. The reduction in infection was attributed to the interference of the conjugates in the early stages of CHIKV life cycle. With no acute oral toxicity, these compounds significantly reduced inflammation and arthritis in rats. Unlike IBU, the conjugates were not ulcerogenic. In conclusion, the conjugation imparted anti-CHIKV properties while retaining the anti-inflammatory properties of IBU. These findings can encourage further validation and research to develop an antiviral for CHIKV to manage both infection and arthritis.

4.
J Virol ; 96(23): e0133422, 2022 12 14.
Article in English | MEDLINE | ID: mdl-36377875

ABSTRACT

Viruses utilize a plethora of strategies to manipulate the host pathways and hijack host machineries for efficient replication. Several DNA and few RNA viruses are reported to interact with proteins involved in DNA damage responses (DDRs). As the DDR pathways have never been explored in alphaviruses, this investigation intended to understand the importance of the DDR pathways in chikungunya virus (CHIKV) infection in vitro, in vivo, and ex vivo models. The study revealed that CHIKV infection activated the Chk2 and Chk1 proteins associated with the DDR signaling pathways in Vero, RAW264.7, and C2C12 cells. The comet assay revealed an increase in DNA damage by 95%. Inhibition of both ATM-ATR kinases by the ATM/ATR kinase inhibitor (AAKi) showed a drastic reduction in the viral particle formation in vitro. Next, the treatment of CHIKV-infected C57BL/6 mice with this drug reduced the disease score substantially with a 93% decrease in the viral load. The same was observed in human peripheral blood mononuclear cell (hPBMC)-derived monocyte-macrophage populations. Additionally, silencing of Chk2 and Chk1 reduced viral progeny formation by 91.2% and 85.5%, respectively. Moreover, CHIKV-nsP2 was found to interact with Chk2 and Chk1 during CHIKV infection. Furthermore, CHIKV infection induced cell cycle arrest in G1 and G2 phases. In conclusion, this work demonstrated for the first time the mechanistic insights regarding the induction of the DDR pathways by CHIKV that might contribute to the designing of effective therapeutics for the control of this virus infection in the future. IMPORTANCE Being intracellular parasites, viruses require several host cell machineries for effectively replicating their genome, along with virus-encoded enzymes. One of the strategies involves hijacking of the DDR pathways. Several DNA and few RNA viruses interact with the cellular proteins involved in the DDR pathways; however, reports regarding the involvement of Chk2 and Chk1 in alphavirus infection are limited. This is the first study to report that modulation of DDR pathways is crucial for effective CHIKV infection. It also reveals an interaction of CHIKV-nsP2 with two crucial host factors, namely, Chk2 and Chk1, for efficient viral infection. Interestingly, CHIKV infection was found to cause DNA damage and arrest the cell cycle in G1 and G2 phases for efficient viral infection. This information might facilitate the development of effective therapeutics for controlling CHIKV infection in the future.


Subject(s)
Chikungunya Fever , Chikungunya virus , DNA Damage , Virus Replication , Animals , Humans , Mice , Chikungunya Fever/genetics , Chikungunya virus/physiology , Leukocytes, Mononuclear/metabolism , Mice, Inbred C57BL , RAW 264.7 Cells , Vero Cells , Chlorocebus aethiops , Cell Cycle Checkpoints
5.
Front Microbiol ; 13: 856913, 2022.
Article in English | MEDLINE | ID: mdl-35847066

ABSTRACT

The emergence of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) as a serious pandemic has altered the global socioeconomic dynamics. The wide prevalence, high death counts, and rapid emergence of new variants urge for the establishment of research infrastructure to facilitate the rapid development of efficient therapeutic modalities and preventive measures. In agreement with this, SARS-CoV-2 strains were isolated from patient swab samples collected during the first COVID-19 wave in Odisha, India. The viral isolates were adapted to in vitro cultures and further characterized to identify strain-specific variations in viral growth characteristics. The neutralization susceptibility of viral isolates to vaccine-induced antibodies was determined using sera from individuals vaccinated in the Government-run vaccine drive in India. The major goal was to isolate and adapt SARS-CoV-2 viruses in cell culture with minimum modifications to facilitate research activities involved in the understanding of the molecular virology, host-virus interactions, drug discovery, and animal challenge models that eventually contribute toward the development of reliable therapeutics.

6.
Antimicrob Agents Chemother ; 66(7): e0046322, 2022 07 19.
Article in English | MEDLINE | ID: mdl-35766508

ABSTRACT

The increase in disease incidences and persistent Chikungunya virus (CHIKV)-induced arthritis have been a huge burden on public health globally. In the absence of specific antivirals or vaccines, it is essential to continue efforts to develop effective anti-CHIKV strategies. Our previous study showing the in vitro anti-CHIKV potential of a novel molecule 1-[(2-methylbenzimidazol-1-yl) methyl]-2-oxo-indolin-3-ylidene] amino] thiourea (MBZM-N-IBT) encouraged us to further validate its efficacy. Here, the effect of MBZM-N-IBT was evaluated in vitro in RAW 264.7 cells, in vivo in C57BL/6 mice, and ex vivo in human peripheral blood mononuclear cells (hPBMCs). The study demonstrated that CHIKV infection was efficiently abrogated in RAW 264.7 cells (IC50 = 22.34 µM) with significant inhibition in viral proteins. The inhibition was effective in the postentry step, and MBZM-N-IBT predominately interfered in the early stages of CHIKV life cycle. It was further supported when the protease activity of CHIKV-nsP2 was hindered by the compound. Moreover, it diminished the CHIKV-induced inflammatory responses in vitro through significant downregulation of all the major mitogen-activated protein kinases (MAPKs), NF-κB, cyclooxygenase (COX)-2, and cytokines. Furthermore, MBZM-N-IBT restricted CHIKV infection and inflammation in vivo, leading to reduced clinical scores and complete survival of C57BL/6 mice. Additionally, it has been noticed that the CHIKV infection was reduced remarkably in hPBMC-derived monocyte-macrophage populations ex vivo by the compound. In conclusion, it can be suggested that this novel compound MBZM-N-IBT has been demonstrated to be a potential anti-CHIKV molecule in vitro, in vivo, and ex vivo and fulfilled all the criteria to investigate further for successful treatment of CHIKV infection.


Subject(s)
Chikungunya Fever , Chikungunya virus , Animals , Benzimidazoles , Chikungunya Fever/drug therapy , Humans , Isatin/analogs & derivatives , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , Peptide Hydrolases/metabolism , Virus Replication
7.
J Ethnopharmacol ; 296: 115458, 2022 Oct 05.
Article in English | MEDLINE | ID: mdl-35728708

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Cameroon is one of the sub-Saharan African countries affected by Chikungunya virus (CHIKV). With the absence of approved treatment, this disease represents globally a major public health concern. Several plants are traditionally used in Cameroon for the treatment of virus induced fever and arthralgia. But to date there is no study that validate the efficacy of these plants for the treatment of Chikungunya virus infection. AIM OF THE STUDY: This study aims to explore the inhition effect, mechanism of action of plant extracts against Chikungunya virus. MATERIAL AND METHODS: An ethnobotanical survey conducted in some regions of Cameroon, led to the identification of nine medicinal plants used in traditional medicine for the healing of fever-related diseases and arthritis. Crude hydro-ethanolic extracts of each plant were prepared by maceration and their effects against CHIKV infection were investigated. CHIKV S27 strain was used to infection in Vero cell line. The antiviral activities were determined by plaque assay and/or RT-PCR targeting E1 envelope gene of CHIKV. Dose-response studies of the active plants were also determined by flow cytometry and Western blot. RESULTS: Four extracts, Entada africana Guill et Pers. (E4), Entandrophragma cylindricum Sprague (EI), Khaya grandifoliola C. D.C. Sapindales (E2) and Macaranga hurifolia Beille (E6) showed antiviral activity with the half-maximal inhibitory concentration of 8.29; 8.14; 12.81 and 26.89 µg/mL respectively. All extracts were nontoxic up to the concentration of 100 µg/µL. Entandrophragma cylindricum Sprague (EI), Khaya grandifoliola C. D.C. Sapindales (E2), and Entada africana Guill et Pers. (E4) showed strong inhibition on the entry step of viral infection. At the same time, only Entandrophragma cylindricum Sprague (EI) inhibited the viral titer significantly in replication and intercellular assembly steps. Four plant extracts namely Entandrophragma cylindricum Sprague (EI), Macaranga hurifolia Beille (E6), Phragmentera capitata (Sprengel) Balle (E12), and Detarium microcarpum (E13) were effective against egression step. CONCLUSIONS: Together, the results of this study showed anti-chikungunya activities of Entandrophragma cylindricum Sprague (EI) and Macaranga hurifolia Beille (E6), with therapeutics perspectives and can be promising sources of the development of anti-CHIKV molecule in future.


Subject(s)
Chikungunya Fever , Chikungunya virus , Fabaceae , Meliaceae , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cameroon , Chikungunya Fever/drug therapy , Chikungunya virus/genetics , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Virus Replication
8.
Mol Omics ; 18(6): 490-505, 2022 07 11.
Article in English | MEDLINE | ID: mdl-35506682

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a major global health concern. This virus infects the upper respiratory tract and causes pneumonia-like symptoms. So far, few studies have shown alterations in nasopharyngeal (NP) microbial diversity, enrichment of opportunistic pathogens and their role in co-infections during respiratory infections. Therefore, we hypothesized that microbial diversity changes, with increase in the population of opportunistic pathogens, during SARS-CoV2 infection in the nasopharynx, which may be involved in co-infection in COVID-19 patients. The 16S rRNA variable regions, V1-V9, of NP samples of control and COVID-19 (symptomatic and asymptomatic) patients were sequenced using the Oxford Nanopore™ technology. Comprehensive bioinformatics analysis for determining alpha/beta diversities, non-metric multidimensional scaling, correlation studies, canonical correspondence analysis, linear discriminate analysis, and dysbiosis index were used to analyze the control and COVID-19-specific NP microbiomes. We observed significant dysbiosis in the COVID-19 NP microbiome with an increase in the abundance of opportunistic pathogens at genus and species levels in asymptomatic/symptomatic patients. The significant abundance of Mycobacteria spp. and Mycoplasma spp. in symptomatic patients suggests their association and role in co-infections in COVID-19 patients. Furthermore, we found strong correlation of enrichment of Mycobacteria and Mycoplasma with the occurrences of chest pain and fever in symptomatic COVID-19 patients. This is the first study from India to show the abundance of Mycobacteria and Mycoplasma opportunistic pathogens in non-hospitalized COVID-19 patients and their relationship with symptoms, indicating the possibility of co-infections.


Subject(s)
COVID-19 , Coinfection , Mycobacterium , Mycoplasma , Coinfection/epidemiology , Dysbiosis , Humans , Nasopharynx , RNA, Ribosomal, 16S/genetics , RNA, Viral , SARS-CoV-2
9.
Antimicrob Agents Chemother ; 66(1): e0148921, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34748384

ABSTRACT

Chikungunya virus (CHIKV) has reemerged as a global public health threat. The inflammatory pathways of the renin-angiotensin system (RAS) and peroxisome proliferator-activated receptor-gamma (PPAR-γ) are usually involved in viral infections. Thus, telmisartan (TM), which is known to block the angiotensin 1 (AT1) receptor and activate PPAR-γ, was investigated for activity against CHIKV. The anti-CHIKV effect of TM was investigated in vitro (Vero cells, RAW 264.7 cells, and human peripheral blood mononuclear cells [hPBMCs]) and in vivo (C57BL/6 mice). TM was found to abrogate CHIKV infection efficiently (50% inhibitory concentration (IC50) of 15.34 to 20.89 µM in the Vero cells and RAW 264.7 cells, respectively). Viral RNA and proteins were reduced remarkably. Additionally, TM interfered in the early and late stages of the CHIKV life cycle with efficacy during pretreatment and posttreatment. Moreover, the agonist of the AT1 receptor and an antagonist of PPAR-γ increased CHIKV infection, suggesting that the antiviral potential of TM occurs through modulating host factors. In addition, reduced activation of all major mitogen-activated protein kinases (MAPKs), NF-κB (p65), and cytokines by TM occurred through the inflammatory axis and supported the fact that the anti-CHIKV efficacy of TM is partly mediated through the AT1/PPAR-γ/MAPKs pathways. Interestingly, at a human equivalent dose, TM abrogated CHIKV infection and inflammation significantly, leading to reduced clinical scores and complete survival of C57BL/6 mice. Additionally, TM reduced infection in hPBMC-derived monocyte-macrophage populations in vitro. Hence, TM was found to reduce CHIKV infection by targeting both viral and host factors. Considering its safety and in vivo efficacy, it can be a suitable candidate in the future for repurposing against CHIKV.


Subject(s)
Chikungunya Fever , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases , PPAR gamma , Receptor, Angiotensin, Type 1 , Animals , Chikungunya Fever/drug therapy , Chlorocebus aethiops , Leukocytes, Mononuclear/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/metabolism , Receptor, Angiotensin, Type 1/metabolism , Telmisartan/pharmacology , Vero Cells
10.
PLoS Pathog ; 17(11): e1009667, 2021 11.
Article in English | MEDLINE | ID: mdl-34780576

ABSTRACT

Chikungunya virus (CHIKV) epidemics around the world have created public health concern with the unavailability of effective drugs and vaccines. This emphasizes the need for molecular understanding of host-virus interactions for developing effective targeted antivirals. Microarray analysis was carried out using CHIKV strain (Prototype and Indian) infected Vero cells and two host isozymes, MAPK activated protein kinase 2 (MK2) and MAPK activated protein kinase 3 (MK3) were selected for further analysis. The substrate spectrum of both enzymes is indistinguishable and covers proteins involved in cytokines production, endocytosis, reorganization of the cytoskeleton, cell migration, cell cycle control, chromatin remodeling and transcriptional regulation. Gene silencing and drug treatment were performed in vitro and in vivo to unravel the role of MK2/MK3 in CHIKV infection. Gene silencing of MK2 and MK3 abrogated around 58% CHIKV progeny release from the host cell and a MK2 activation inhibitor (CMPD1) treatment demonstrated 68% inhibition of viral infection suggesting a major role of MAPKAPKs during late CHIKV infection in vitro. Further, it was observed that the inhibition in viral infection is primarily due to the abrogation of lamellipodium formation through modulation of factors involved in the actin cytoskeleton remodeling pathway. Moreover, CHIKV-infected C57BL/6 mice demonstrated reduction in the viral copy number, lessened disease score and better survivability after CMPD1 treatment. In addition, reduction in expression of key pro-inflammatory mediators such as CXCL13, RAGE, FGF, MMP9 and increase in HGF (a CHIKV infection recovery marker) was observed indicating the effectiveness of the drug against CHIKV. Taken together it can be proposed that MK2 and MK3 are crucial host factors for CHIKV infection and can be considered as important target for developing effective anti-CHIKV strategies.


Subject(s)
Actins/metabolism , Anilides/pharmacology , Antiviral Agents/pharmacology , Chikungunya Fever/prevention & control , Chikungunya virus/drug effects , Cytoskeleton/drug effects , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology , Actins/drug effects , Animals , Chikungunya Fever/virology , Chlorocebus aethiops , Male , Mice , Mice, Inbred C57BL , Vero Cells , Virus Release
11.
EMBO Rep ; 22(11): e52948, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34467632

ABSTRACT

The type I interferon (IFN) response is the major host arsenal against invading viruses. IRGM is a negative regulator of IFN responses under basal conditions. However, the role of human IRGM during viral infection has remained unclear. In this study, we show that IRGM expression is increased upon viral infection. IFN responses induced by viral PAMPs are negatively regulated by IRGM. Conversely, IRGM depletion results in a robust induction of key viral restriction factors including IFITMs, APOBECs, SAMHD1, tetherin, viperin, and HERC5/6. Additionally, antiviral processes such as MHC-I antigen presentation and stress granule signaling are enhanced in IRGM-deficient cells, indicating a robust cell-intrinsic antiviral immune state. Consistently, IRGM-depleted cells are resistant to the infection with seven viruses from five different families, including Togaviridae, Herpesviridae, Flaviviverdae, Rhabdoviridae, and Coronaviridae. Moreover, we show that Irgm1 knockout mice are highly resistant to chikungunya virus (CHIKV) infection. Altogether, our work highlights IRGM as a broad therapeutic target to promote defense against a large number of human viruses, including SARS-CoV-2, CHIKV, and Zika virus.


Subject(s)
GTP-Binding Proteins/antagonists & inhibitors , Virus Diseases/immunology , Animals , Antiviral Agents/pharmacology , Humans , Mice , Virus Replication
12.
Virol J ; 18(1): 103, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039377

ABSTRACT

INTRODUCTION: The emergence of drug resistance and cross-resistance to existing drugs has warranted the development of new antivirals for Herpes simplex viruses (HSV). Hence, we have designed this study to evaluate the anti-viral activity of 1-[(2-methyl benzimidazole-1-yl) methyl]-2-oxo-indolin-3-ylidene] amino] thiourea (MBZM-N-IBT), against HSV-1. METHOD: Molecular docking was performed to assess the affinity of MBZM-N-IBT for HSV-1 targets. This was validated by plaque assay, estimation of RNA and protein levels as well as time of addition experiments in vitro. RESULT: Molecular docking analysis suggested the inhibitory capacity of MBZM-N-IBT against HSV-1. This was supported by the abrogation of the HSV-1 infectious viral particle formation with the IC50 value of 3.619 µM. Viral mRNA levels were also reduced by 72% and 84% for UL9 and gC respectively. MBZM-N-IBT also reduced the protein synthesis for gC and ICP8 significantly. While mRNA of ICP8 was not significantly affected, its protein synthesis was reduced by 47%. The time of addition experiment revealed the capacity of MBZM-N-IBT to inhibit HSV-1 at early as well as late stages of infection in the Vero cells. Similar effect of MBZM-N-IBT was also noticed in the Raw 264.7 and BHK 21 cells after HSV-1 infection. Supported by the in silico data, this can be attributed to possible interference with multiple HSV targets including the ICP8, ICP27, UL42, UL25, UL15 and gB proteins. CONCLUSION: These results along with the lack of acute oral toxicity and significant anti-inflammatory effects suggest its suitability for further evaluation as a non-nucleoside inhibitor of HSV.


Subject(s)
Benzimidazoles/pharmacology , Herpes Simplex , Herpesvirus 1, Human , Isatin/analogs & derivatives , Animals , Chlorocebus aethiops , Cricetinae , Herpes Simplex/drug therapy , Herpesvirus 1, Human/drug effects , Isatin/pharmacology , Mice , Molecular Docking Simulation , RAW 264.7 Cells , RNA, Messenger , Vero Cells , Viral Proteins/genetics , Virus Replication
13.
Asian Pac J Cancer Prev ; 22(4): 1171-1181, 2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33906310

ABSTRACT

BACKGROUND: The association of BAX -248 G>A and BCL2 -938 C>A with different cancers created conflicts.  We studied the correlation and the effect of these polymorphisms in patients with Nasopharyngeal Carcinoma (NPC).  Methods: PCR-RFLP and Sanger sequencing were used to detect polymorphisms. Statistical analysis including forest plot and Kaplan-Meier Log-rank test was conducted to investigate the association and effect of these SNPs on the NPC patients' survival. The computational study was performed to investigate the possible regulatory role between these polymorphisms and the poor survival of NPC patients. Meta-analysis was executed to check the tissue-specific association of these polymorphisms in the context of global cancer prognosis. RESULTS: We observed an increased and significant association of BAX -248 G>A [GA:OR=5.29, 95%CI=1.67,16.67, P=0.004; GA+AA:OR=5.71, 95%CI=1.82,17.90, P =0.002; A:OR=5.33, 95%CI=1.76,16.13, P=0.003], and BCL2 -938 C>A [CA:OR=2.26, 95%CI=1.03,4.96, P=0.04; AA:OR=3.56, 95%CI=0.97,13.05, P=0.05; CA+AA:OR=3.10, 95%CI=1.51,6.35, P=0.002; A:OR=2.90, 95% CI=1.59,5.29, P=0.0005] with the risk of NPC. Also, these SNPs were strongly correlated with poor survival in NPC patients (lower estimated survival mean, lower estimated proportion surviving at 5 years with p <0.05). The computational study showed that these SNPs altered the binding affinity of transcription factors HIF1, SP1, PAX3, PAX9 and CREB towards promoter (Lower p indicates strong affinity). The meta-analysis revealed the tissue-specific association of these polymorphisms. BAX -248 G>A showed a significant correlation with carcinomas [A vs G:OR=1.60, 95%CI=1.09,2.34, P=0.01; AA vs GG:OR=2.61, 95%CI=1.68,4.06, p <0.001; AA+GA vs GG:OR=1.53,95%CI=1.04,2.25, P=0.02); AA vs GG+GA:OR=2.53, 95%CI=1.65,3.87, p <0.001], and BCL2 -938 C>A with other malignancies [A vs C:OR=1.45, 95%CI=1.26,1.66, p <0.001; AA vs CC:OR=2.07, 95%CI: 1.15,3.72, P=0.01; AA+CA vs CC:OR=1.42, 95%CI=1.18,1.72, p <0.001; AA vs CC+CA:OR=1.89, 95%CI=1.02,3.50, P=0.04]. CONCLUSIONS: BAX -248 G>A and BCL2 -938 C>A was associated with poor survival in NPC patients. It may increase cancer susceptibility through transcriptional regulation. Moreover, these SNPs' effects could be tissue-specific.
.


Subject(s)
Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/mortality , Proto-Oncogene Proteins c-bcl-2/genetics , bcl-2-Associated X Protein/genetics , Case-Control Studies , Female , Genetic Predisposition to Disease/genetics , Humans , India , Kaplan-Meier Estimate , Male , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Polymorphism, Genetic/genetics , Survival Rate
14.
Arch Virol ; 166(1): 139-155, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33125586

ABSTRACT

Chikungunya virus (CHIKV), a virus that induces pathogenic inflammatory host immune responses, is re-emerging worldwide, and there are currently no established antiviral control measures. Transient receptor potential vanilloid 1 (TRPV1), a non-selective Ca2+-permeable ion channel, has been found to regulate various host inflammatory responses including several viral infections. Immune responses to CHIKV infection in host macrophages have been reported recently. However, the possible involvement of TRPV1 during CHIKV infection in host macrophages has not been studied. Here, we investigated the possible role of TRPV1 in CHIKV infection of the macrophage cell line RAW 264.7. It was found that CHIKV infection upregulates TRPV1 expression in macrophages. To confirm this observation, the TRPV1-specific modulators 5'-iodoresiniferatoxin (5'-IRTX, a TRPV1 antagonist) and resiniferatoxin (RTX, a TRPV1 agonist) were used. Our results indicated that TRPV1 inhibition leads to a reduction in CHIKV infection, whereas TRPV1 activation significantly enhances CHIKV infection. Using a plaque assay and a time-of-addition assay, it was observed that functional modulation of TRPV1 affects the early stages of the viral lifecycle in RAW 264.7 cells. Moreover, CHIKV infection was found to induce of pNF-κB (p65) expression and nuclear localization. However, both activation and inhibition of TRPV1 were found to enhance the expression and nuclear localization of pNF-κB (p65) and production of pro-inflammatory TNF and IL-6 during CHIKV infection. In addition, it was demonstrated by Ca2+ imaging that TRPV1 regulates Ca2+ influx during CHIKV infection. Hence, the current findings highlight a potentially important regulatory role of TRPV1 during CHIKV infection in macrophages. This study might also have broad implications in the context of other viral infections as well.


Subject(s)
Antiviral Agents/pharmacology , Chikungunya Fever/drug therapy , Chikungunya virus/drug effects , Macrophages/drug effects , TRPV Cation Channels/metabolism , Animals , Cell Line , Chikungunya Fever/metabolism , Chikungunya Fever/virology , Diterpenes/pharmacology , Macrophages/metabolism , Macrophages/virology , Mice , RAW 264.7 Cells , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...