Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
J Med Chem ; 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687966

ABSTRACT

Despite the record-breaking discovery, development and approval of vaccines and antiviral therapeutics such as Paxlovid, coronavirus disease 2019 (COVID-19) remained the fourth leading cause of death in the world and third highest in the United States in 2022. Here, we report the discovery and characterization of PF-07817883, a second-generation, orally bioavailable, SARS-CoV-2 main protease inhibitor with improved metabolic stability versus nirmatrelvir, the antiviral component of the ritonavir-boosted therapy Paxlovid. We demonstrate the in vitro pan-human coronavirus antiviral activity and off-target selectivity profile of PF-07817883. PF-07817883 also demonstrated oral efficacy in a mouse-adapted SARS-CoV-2 model at plasma concentrations equivalent to nirmatrelvir. The preclinical in vivo pharmacokinetics and metabolism studies in human matrices are suggestive of improved oral pharmacokinetics for PF-07817883 in humans, relative to nirmatrelvir. In vitro inhibition/induction studies against major human drug metabolizing enzymes/transporters suggest a low potential for perpetrator drug-drug interactions upon single-agent use of PF-07817883.

2.
Pathogens ; 11(2)2022 Feb 17.
Article in English | MEDLINE | ID: mdl-35215199

ABSTRACT

Transgenic mice expressing human angiotensin-converting enzyme 2 under the cytokeratin 18 promoter (K18-hACE2) have been extensively used to investigate the pathogenesis and tissue tropism of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. Neuroinvasion and the replication of SARS-CoV-2 within the central nervous system (CNS) of K18-hACE2 mice is associated with increased mortality; although, the mechanisms by which this occurs remain unclear. In this study, we generated primary neuronal cultures from K18-hACE2 mice to investigate the effects of a SARS-CoV-2 infection. We also evaluated the immunological response to SARS-CoV-2 infection in the CNS of K18-hACE2 mice and mouse neuronal cultures. Our data show that neuronal cultures obtained from K18-hACE2 mice are permissive to SARS-CoV-2 infection and support productive virus replication. Furthermore, SARS-CoV-2 infection upregulated the expression of genes involved in innate immunity and inflammation, including IFN-α, ISG-15, CXCL10, CCL2, IL-6 and TNF-α, in the neurons and mouse brains. In addition, we found that SARS-CoV-2 infection of neurons and mouse brains activates the ZBP1/pMLKL-regulated necroptosis pathway. Together, our data provide insights into the neuropathogenesis of SARS-CoV-2 infection in K18-hACE2 mice.

3.
Molecules ; 26(14)2021 Jul 16.
Article in English | MEDLINE | ID: mdl-34299596

ABSTRACT

Zika virus (ZIKV) represents a re-emerging threat to global health due to its association with congenital birth defects. ZIKV NS2B-NS3 protease is crucial for virus replication by cleaving viral polyprotein at various junctions to release viral proteins and cause cytotoxic effects in ZIKV-infected cells. This study characterized the inhibitory effects of doxycycline against ZIKV NS2B-NS3 protease and viral replication in human skin cells. The in silico data showed that doxycycline binds to the active site of ZIKV protease at a low docking energy (-7.8 Kcal/mol) via four hydrogen bonds with the protease residues TYR1130, SER1135, GLY1151, and ASP83. Doxycycline efficiently inhibited viral NS2B-NS3 protease at average human temperature (37 °C) and human temperature with a high fever during virus infection (40 °C). Interestingly, doxycycline showed a higher inhibitory effect at 40 °C (IC50 = 5.3 µM) compared to 37 °C (9.9 µM). The virus replication was considerably reduced by increasing the concentration of doxycycline. An approximately 50% reduction in virus replication was observed at 20 µM of doxycycline. Treatment with 20 µM of doxycycline reduced the cytopathic effects (CPE), and the 40 µM of doxycycline almost eliminated the CPE of human skin cells. This study showed that doxycycline binds to the ZIKV protease and inhibits its catalytic activity at a low micro-molecular concentration range. Treatment of human skin fibroblast with doxycycline eliminated ZIKV infection and protected the cells against the cytopathic effects of the infection.


Subject(s)
Doxycycline/pharmacology , Fibroblasts/metabolism , Skin/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Proteins/antagonists & inhibitors , Virus Replication/drug effects , Zika Virus/physiology , Animals , Chlorocebus aethiops , Doxycycline/chemistry , Fibroblasts/virology , Humans , Serine Endopeptidases/chemistry , Serine Endopeptidases/metabolism , Skin/virology , Vero Cells , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Viral Proteins/chemistry , Viral Proteins/metabolism , Zika Virus/chemistry
4.
Mol Biotechnol ; 63(3): 240-248, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33464543

ABSTRACT

The global public health has been compromised since the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) emerged in late December 2019. There are no specific antiviral drugs available to combat SARS-CoV-2 infection. Besides the rapid dissemination of SARS-CoV-2, several variants have been identified with a potential epidemiologic and pathogenic variation. This fact has forced antiviral drug development strategies to stay innovative, including new drug discovery protocols, combining drugs, and establishing new drug classes. Thus, developing novel screening methods and direct-targeting viral enzymes could be an attractive strategy to combat SARS-CoV-2 infection. In this study, we designed, optimized, and validated a cell-based assay protocol for high-throughput screening (HTS) antiviral drug inhibitors against main viral protease (3CLpro). We applied the split-GFP complementation to develop GFP-split-3CLpro HTS system. The system consists of GFP-based reporters that become fluorescent upon cleavage by SARS-CoV-2 protease 3CLpro. We generated a stable GFP-split-3CLpro HTS system valid to screen large drug libraries for inhibitors to SARS-CoV-2 main protease in the bio-safety level 2 laboratory, providing real-time antiviral activity of the tested compounds. Using this assay, we identified a new class of viral protease inhibitors derived from quinazoline compounds that worth further in vitro and in vivo validation.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases/antagonists & inhibitors , High-Throughput Screening Assays/methods , SARS-CoV-2/drug effects , COVID-19/virology , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Drug Development , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries
5.
Viruses ; 13(1)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33477869

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection can cause neurological disease in humans, but little is known about the pathogenesis of SARS-CoV-2 infection in the central nervous system (CNS). Herein, using K18-hACE2 mice, we demonstrate that SARS-CoV-2 neuroinvasion and encephalitis is associated with mortality in these mice. Intranasal infection of K18-hACE2 mice with 105 plaque-forming units of SARS-CoV-2 resulted in 100% mortality by day 6 after infection. The highest virus titers in the lungs were observed on day 3 and declined on days 5 and 6 after infection. By contrast, very high levels of infectious virus were uniformly detected in the brains of all the animals on days 5 and 6. Onset of severe disease in infected mice correlated with peak viral levels in the brain. SARS-CoV-2-infected mice exhibited encephalitis hallmarks characterized by production of cytokines and chemokines, leukocyte infiltration, hemorrhage and neuronal cell death. SARS-CoV-2 was also found to productively infect cells within the nasal turbinate, eye and olfactory bulb, suggesting SARS-CoV-2 entry into the brain by this route after intranasal infection. Our data indicate that direct infection of CNS cells together with the induced inflammatory response in the brain resulted in the severe disease observed in SARS-CoV-2-infected K18-hACE2 mice.


Subject(s)
Brain/virology , COVID-19/pathology , Encephalitis, Viral/pathology , Lung/virology , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Animals , Brain/pathology , COVID-19/mortality , Cytokines/blood , Disease Models, Animal , Encephalitis, Viral/virology , Lung/pathology , Mice , Mice, Transgenic , Viral Load
6.
Pediatr Allergy Immunol ; 32(1): 17-22, 2021 01.
Article in English | MEDLINE | ID: mdl-32897642

ABSTRACT

Multisystem inflammatory syndrome in children (MIS-C) during the COVID-19 pandemic raised a global alert from the Centers for Disease Control and Prevention's Health Alert Network. The main manifestations of MIS-C (also known as pediatric MIS (PMIS)) in the setting of a severe inflammatory state include fever, diarrhea, shock, and variable presence of rash, conjunctivitis, extremity edema, and mucous membrane changes. In some cases, these symptoms progressed to multi-organ failure. The low percentage of children with asymptomatic cases compared with mild illness and moderate illness could be correlated with the rare cases of MIS-C. One potential explanation for the progression to severe MIS-C disease despite the presence of readily detectable anti-SARS-CoV-2 antibodies could be due to the potential role of antibody-dependent enhancement (ADE). We reason that the incidence of the ADE phenomenon whereby the pathogen-specific antibodies can promote pathology should be considered in vaccine development against SARS-CoV-2.


Subject(s)
COVID-19/epidemiology , Systemic Inflammatory Response Syndrome/epidemiology , Adolescent , Antibodies, Viral/immunology , Antibody-Dependent Enhancement/immunology , COVID-19/immunology , Child , Child, Preschool , Conjunctivitis/epidemiology , Diarrhea/epidemiology , Exanthema/epidemiology , Humans , Infant , Macrophage Activation/immunology , Pandemics , SARS-CoV-2/immunology , Severity of Illness Index , Systemic Inflammatory Response Syndrome/immunology , Young Adult
7.
Pathogens ; 9(7)2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32640525

ABSTRACT

In the absence of therapeutic interventions, and a possible vaccine candidate, the spread of COVID-19 disease and associated fatalities are on the rise. The high mutation frequency in the genomic material of these viruses supports their ability to adapt to new environments, resulting in an efficient alteration in tissue tropism and host range. Therefore, the coronavirus' health threats could be relevant for the long-term. The epidemiological data indicate that age, sex, and cardio-metabolic disease have a significant impact on the spread and severity of COVID-19. In this review, we highlight recent updates on the pathogenesis of SARS-CoV-2 among men and women, including children. We also discuss the role of the cellular receptors and coreceptors used by the virus to enter host cells on differential infection among men, women, and cardio-metabolic patients.

8.
Virology ; 547: 7-11, 2020 08.
Article in English | MEDLINE | ID: mdl-32442105

ABSTRACT

SARS-COV-2 has recently emerged as a new public health threat. Herein, we report that the FDA-approved drug, auranofin, inhibits SARS-COV-2 replication in human cells at low micro molar concentration. Treatment of cells with auranofin resulted in a 95% reduction in the viral RNA at 48 h after infection. Auranofin treatment dramatically reduced the expression of SARS-COV-2-induced cytokines in human cells. These data indicate that auranofin could be a useful drug to limit SARS-CoV-2 infection and associated lung injury due to its antiviral, anti-inflammatory and anti-reactive oxygen species (ROS) properties. Further animal studies are warranted to evaluate the safety and efficacy of auranofin for the management of SARS-COV-2 associated disease.


Subject(s)
Auranofin/pharmacology , Betacoronavirus/drug effects , Virus Replication/drug effects , Antiviral Agents/pharmacology , Betacoronavirus/physiology , COVID-19 , Cell Line , Coronavirus Infections , Cytokines , Drug Evaluation, Preclinical , Gold , Humans , Inflammation , Pandemics , Pneumonia, Viral , SARS-CoV-2
9.
Mol Biotechnol ; 62(5): 289-296, 2020 May.
Article in English | MEDLINE | ID: mdl-32185600

ABSTRACT

Pasteurella multocida is the main cause of haemorrhagic septicaemia (HS) outbreak in livestock, such as cattle and buffaloes. Conventional vaccines such as alum-precipitated or oil-adjuvant broth bacterins were injected subcutaneously to provide protection against HS. However, the immunity developed is only for short term and needed to be administered frequently. In our previous study, a short gene fragment from Pasteurella multocida serotype B was obtained via shotgun cloning technique and later was cloned into bacterial expression system. pQE32-ABA392 was found to possess immunogenic activity towards HS when tested in vivo in rat model. In this study, the targeted gene fragment of ABA392 was sub-cloned into a DNA expression vector pVAX1 and named as pVAX1-ABA392. The new recombinant vaccine was stable and expressed on mammalian cell lines. Serum sample collected from a group of vaccinated rats for ELISA test shows that the antibody in immunized rats was present at high titer and can be tested as a vaccine candidate with challenge in further studies. This successful recombinant vaccine is immunogenic and potentially could be used as vaccine in future against HS.


Subject(s)
DNA, Bacterial/genetics , Hemorrhagic Septicemia/microbiology , Pasteurella Infections/prevention & control , Pasteurella multocida/genetics , Vaccines, DNA/administration & dosage , Animals , Cloning, Molecular , DNA, Bacterial/immunology , Disease Models, Animal , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Hemorrhagic Septicemia/prevention & control , Pasteurella multocida/immunology , Plasmids/genetics , Rats , Sequence Analysis, DNA , Vaccination , Vaccines, DNA/immunology
10.
J Autoimmun ; 109: 102433, 2020 05.
Article in English | MEDLINE | ID: mdl-32113704

ABSTRACT

Coronavirus disease (COVID-19) is caused by SARS-COV2 and represents the causative agent of a potentially fatal disease that is of great global public health concern. Based on the large number of infected people that were exposed to the wet animal market in Wuhan City, China, it is suggested that this is likely the zoonotic origin of COVID-19. Person-to-person transmission of COVID-19 infection led to the isolation of patients that were subsequently administered a variety of treatments. Extensive measures to reduce person-to-person transmission of COVID-19 have been implemented to control the current outbreak. Special attention and efforts to protect or reduce transmission should be applied in susceptible populations including children, health care providers, and elderly people. In this review, we highlights the symptoms, epidemiology, transmission, pathogenesis, phylogenetic analysis and future directions to control the spread of this fatal disease.


Subject(s)
Betacoronavirus/classification , Betacoronavirus/pathogenicity , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Animals , COVID-19 , Coronavirus Infections/prevention & control , Humans , Pandemics/prevention & control , Phylogeny , Pneumonia, Viral/prevention & control , Public Health , SARS-CoV-2 , Zoonoses/epidemiology , Zoonoses/virology
11.
Viruses ; 12(1)2019 12 19.
Article in English | MEDLINE | ID: mdl-31861621

ABSTRACT

West Nile virus (WNV) is a flavivirus that has disseminated globally as a significant cause of viral encephalitis in humans. MircoRNA-155 (miR-155) regulates various aspects of innate and adaptive immune responses. We previously reported that WNV infection induces upregulation of miR-155 in mice brains. In the current study, we demonstrate the critical role of miR-155 in restricting the pathogenesis of WNV infection in mice. Compared to wild-type (WT) mice, miR-155 knockout mice exhibited significantly higher morbidity and mortality after infection with either a lethal strain, WNV NY99, or a non-lethal strain, WNV Eg101. Increased mortality in miR-155-/- mice was associated with significantly high WNV burden in the serum and brains. Protein levels of interferon (IFN)-α in the serum and brains were higher in miR-155-/- mice. However, miR-155-/- mice exhibited significantly lower protein levels of anti-viral interleukin (IL)-1ß, IL-12, IL-6, IL-15, and GM-CSF despite the high viral load. Primary mouse cells lacking miR-155 were more susceptible to infection with WNV compared to cells derived from WT mice. Besides, overexpression of miR-155 in human neuronal cells modulated anti-viral cytokine response and resulted in significantly lower WNV replication. These data collectively indicate that miR-155 restricts WNV production in mouse and human cells and protects against lethal WNV infection in mice.


Subject(s)
Gene Expression Regulation , Host-Pathogen Interactions/genetics , MicroRNAs/genetics , West Nile Fever/genetics , West Nile Fever/virology , West Nile virus/physiology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Host-Pathogen Interactions/immunology , Humans , Inflammation Mediators/metabolism , Mice , Mice, Knockout , Viral Load , Virus Replication , West Nile Fever/immunology , West Nile Fever/pathology
12.
Front Microbiol ; 10: 2089, 2019.
Article in English | MEDLINE | ID: mdl-31572318

ABSTRACT

West Nile virus (WNV), a neurotropic flavivirus, is the leading cause of viral encephalitis in the United States. Recently, Zika virus (ZIKV) infections have caused serious neurological diseases and birth defects, specifically Guillain-Barrè syndrome and microcephaly. Z-DNA binding protein 1 (ZBP1) is a cytoplasmic sensor that that has been shown to play a significant role in initiating a robust immune response. We previously reported that WNV and ZIKV infections induce dramatic up-regulation of ZBP1 in mouse brains as well as in infected primary mouse cells. Herein, we show the critical role of ZBP1 in restricting the pathogenesis of WNV and ZIKV infections. Deletion of ZBP1 resulted in significantly higher morbidity and mortality after infection with a pathogenic WNV NY99 strain in mice. No mortality was observed in wild-type (WT) mice infected with the non-pathogenic WNV strain, Eg101. Interestingly, infection of ZBP1-/- mice with WNV Eg101 was lethal resulting in 100% mortality, suggesting that ZBP1 is required for survival after WNV infection. Viremia and brain viral load were significantly higher in ZBP1-/- mice compared to WT mice. In addition, protein levels of interferon (IFN)-α, and inflammatory cytokines and chemokines were significantly higher in the serum and brains of infected ZBP1-/- mice compared to the WT mice. Primary mouse cortical neurons and mouse embryonic fibroblasts (MEFs) derived from ZBP1-/- mice produced higher virus titers compared to WT cells after infection with WNV NY99 and WNV Eg101. Similarly, neurons and MEFs lacking ZBP1 exhibited significantly enhanced replication of PRVABC59 (Asian) and MR766 (African) ZIKV compared to WT cells. The knockout of ZBP1 function in MEFs inhibited ZBP1-dependent virus-induced cell death. In conclusion, these data reveal that ZBP1 restricts WNV and ZIKV production in mouse cells and is required for survival of a peripheral WNV infection in mice.

13.
Pathogens ; 8(3)2019 Sep 12.
Article in English | MEDLINE | ID: mdl-31547236

ABSTRACT

Flavivirus replication in host cells requires the formation of replication and assembly complexes on the cytoplasmic side of the endoplasmic reticulum (ER) membrane. These complexes consist of an ER membrane, viral proteins, and host proteins. Genome-wide investigations have identified a number of ER multiprotein complexes as vital factors for flavivirus replication. The detailed mechanisms of the role of ER complexes in flavivirus replication are still largely elusive. This review highlights the fact that the ER multiprotein complexes are crucial for the formation of flavivirus replication and assembly complexes, and the ER complexes could be considered as a target for developing successful broad-spectrum anti-flavivirus drugs.

14.
Antiviral Res ; 171: 104590, 2019 11.
Article in English | MEDLINE | ID: mdl-31421166

ABSTRACT

Two major flaviviruses, dengue virus (DENV) and Zika virus (ZIKV), cause severe health and economic burdens worldwide. Recently, genome-wide screenings have uncovered the importance of regulators of the Hrd1 ubiquitin ligase-mediated endoplasmic reticulum (ER)-associated degradation (ERAD) pathway for flavivirus replication in host cells. Here we report the identification of the compound Bardoxolone methyl (CDDO-me) as a potent inhibitor of the Hrd1 ubiquitin ligase-mediated ERAD, which possesses a broad-spectrum activity against both DENV and ZIKV. Cellular thermal shift assay (CETSA) suggested that CDDO-me binds to grp94, a key component of the Hrd1 pathway, at a low nanomolar concentration, whereas interaction was not detected with its paralog Hsp90. CDDO-me and the grp94 inhibitor PU-WS13 substantially suppressed DENV2 replication and the cytopathic effects caused by DENV and ZIKV infection. The antiviral activities of both compounds were demonstrated for all four DENV serotypes and four ZIKV strains in multiple human cell lines. This study defines grp94 as a crucial host factor for flavivirus replication and identified CDDO-me as a potent small molecule inhibitor of flavivirus infection. Inhibition of grp94 may contribute to the antiviral activity of CDDO-me. Further investigation of grp94 inhibitors may lead to a new class of broad-spectrum anti-flaviviral medications.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Dengue/virology , Membrane Glycoproteins/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , Zika Virus Infection/virology , Zika Virus/drug effects , Cell Survival , Dengue/drug therapy , Dengue/metabolism , Humans , Oleanolic Acid/pharmacology , Virus Replication/drug effects , Zika Virus Infection/drug therapy , Zika Virus Infection/metabolism
15.
J Pept Sci ; 25(8): e3196, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31290226

ABSTRACT

Dengue virus (DENV) and Zika virus (ZIKV) are flaviviruses transmitted to humans by their common vector, Aedes mosquitoes. DENV infection represents one of the most widely spread mosquito-borne diseases whereas ZIKV infection occasionally re-emerged in the past causing outbreaks. Although there have been considerable advances in understanding the pathophysiology of these viruses, no effective vaccines or antiviral drugs are currently available. In this study, we evaluated the antiviral activity of carnosine, an endogenous dipeptide (ß-alanyl-l-histidine), against DENV serotype 2 (DENV2) and ZIKV infection in human liver cells (Huh7). Computational studies were performed to predict the potential interactions between carnosine and viral proteins. Biochemical and cell-based assays were performed to validate the computational results. Mode-of-inhibition, plaque reduction, and immunostaining assays were performed to determine the antiviral activity of carnosine. Exogenous carnosine showed minimal cytotoxicity in Huh7 cells and rescued the viability of infected cells with EC50 values of 52.3 and 59.5 µM for DENV2 and ZIKV infection, respectively. Based on the mode-of-inhibition assays, carnosine inhibited DENV2 mainly by inhibiting viral genome replication and interfering with virus entry. Carnosine antiviral activity was verified with immunostaining assay where carnosine treatment diminished viral fluorescence signal. In conclusion, carnosine exhibited significant inhibitory effects against DENV2 and ZIKV replication in human liver cells and could be utilized as a lead peptide for the development of effective and safe antiviral agents against DENV and ZIKV.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Zika Virus/drug effects , Animals , Carnosine/pharmacology , Cell Survival/drug effects , Chlorocebus aethiops , Dengue/drug therapy , Humans , Microbial Sensitivity Tests , Vero Cells , Zika Virus Infection/drug therapy
16.
Sci Rep ; 9(1): 10901, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358863

ABSTRACT

Infection with flaviviruses, such as dengue virus (DENV) and the recently re-emerging Zika virus (ZIKV), represents an increasing global risk. Targeting essential host elements required for flavivirus replication represents an attractive approach for the discovery of antiviral agents. Previous studies have identified several components of the Hrd1 ubiquitin ligase-mediated endoplasmic reticulum (ER)-associated degradation (ERAD) pathway, a cellular protein quality control process, as host factors crucial for DENV and ZIKV replication. Here, we report that CP26, a small molecule inhibitor of protein dislocation from the ER lumen to the cytosol, which is an essential step for ERAD, has broad-spectrum anti-flavivirus activity. CP26 targets the Hrd1 complex, inhibits ERAD, and induces ER stress. Ricin and cholera toxins are known to hijack the protein dislocation machinery to reach the cytosol, where they exert their cytotoxic effects. CP26 selectively inhibits the activity of cholera toxin but not that of ricin. CP26 exhibits a significant inhibitory activity against both DENV and ZIKV, providing substantial protection to the host cells against virus-induced cell death. This study identified a novel dislocation inhibitor, CP26, that shows potent anti-DENV and anti-ZIKV activity in cells. Furthermore, this study provides the first example of the targeting of host ER dislocation with small molecules to combat flavivirus infection.


Subject(s)
Antiviral Agents/pharmacology , Dengue Virus/drug effects , Virus Replication/drug effects , Zika Virus/drug effects , Animals , Chlorocebus aethiops , Dengue/drug therapy , Dengue Virus/physiology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum-Associated Degradation/drug effects , HeLa Cells , Host Microbial Interactions/drug effects , Humans , Ubiquitin-Protein Ligases/metabolism , Vero Cells , Zika Virus/physiology , Zika Virus Infection/drug therapy
17.
Mol Neurobiol ; 56(4): 2551-2557, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30043260

ABSTRACT

Zika virus (ZIKV) infection is associated with abnormal functions of neuronal cells causing neurological disorders such as microcephaly in the newborns and Guillain-Barré syndrome in the adults. Typically, healthy brain growth is associated with normal neural stem cell proliferation, differentiation, and maturation. This process requires a controlled cellular metabolism that is essential for normal migration, axonal elongation, and dendrite morphogenesis of newly generated neurons. Thus, the remarkable changes in the cellular metabolism during early stages of neuronal stem cell differentiation are crucial for brain development. Recent studies show that ZIKV directly infects neuronal stem cells in the fetus and impairs brain growth. In this review, we highlighted the fact that the activation of P53 and inhibition of the mTOR pathway by ZIKV infection to neuronal stem cells induces early shifting from glycolysis to oxidative phosphorylation (OXPHOS) may induce immature differentiation, apoptosis, and stem cell exhaustion. We hypothesize that ZIKV infection to mature myelin-producing cells and resulting metabolic shift may lead to the development of neurological diseases, such as Guillain-Barré syndrome. Thus, the effects of ZIKV on the cellular metabolism of neuronal cells may lead to the incidence of neurological disorders as observed recently during ZIKV infection.


Subject(s)
Neurons/metabolism , Neurons/virology , Zika Virus/physiology , Animals , Cell Differentiation , Humans , Models, Biological , Zika Virus Infection/metabolism , Zika Virus Infection/virology
18.
Mol Neurobiol ; 56(4): 2558, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30209773

ABSTRACT

The original version of this article unfortunately contained mistake.

19.
J Autoimmun ; 89: 11-20, 2018 05.
Article in English | MEDLINE | ID: mdl-29352633

ABSTRACT

Dissemination of vector-borne viruses, such as Zika virus (ZIKV), in tropical and sub-tropical regions has a complicated impact on the immunopathogenesis of other endemic viruses such as dengue virus (DENV), chikungunya virus (CHIKV) and human immunodeficiency virus (HIV). The consequences of the possible co-infections with these viruses have specifically shown significant impact on the treatment and vaccination strategies. ZIKV is a mosquito-borne flavivirus from African and Asian lineages that causes neurological complications in infected humans. Many of DENV and CHIKV endemic regions have been experiencing outbreaks of ZIKV infection. Intriguingly, the mosquitoes, Aedes Aegypti and Aedes Albopictus, can simultaneously transmit all the combinations of ZIKV, DENV, and CHIKV to the humans. The co-circulation of these viruses leads to a complicated immune response due to the pre-existence or co-existence of ZIKV infection with DENV and CHIKV infections. The non-vector transmission of ZIKV, especially, via sexual intercourse and placenta represents an additional burden that may hander the treatment strategies of other sexually transmitted diseases such as HIV. Collectively, ZIKV co-circulation and co-infection with other viruses have inevitable impact on the host immune response, diagnosis techniques, and vaccine development strategies for the control of these co-infections.


Subject(s)
Arboviruses/physiology , Chikungunya Fever/epidemiology , HIV Infections/epidemiology , HIV/physiology , Viral Vaccines/immunology , Zika Virus Infection/epidemiology , Zika Virus/physiology , Aedes/physiology , Animals , Chikungunya Fever/immunology , Coinfection , Disease Vectors , Endemic Diseases , HIV Infections/immunology , Humans , Infection Control , Vaccination , Zika Virus Infection/immunology
20.
Sci Rep ; 7: 45540, 2017 03 30.
Article in English | MEDLINE | ID: mdl-28358047

ABSTRACT

N-methyl-D-aspartate receptors (NMDAR) play a central role in epileptogensis and NMDAR antagonists have been shown to have antiepileptic effects in animals and humans. Despite significant progress in the development of antiepileptic therapies over the previous 3 decades, a need still exists for novel therapies. We screened an in-house library of small molecules targeting the NMDA receptor. A novel indolyl compound, 2-(1,1-Dimethyl-1,3-dihydro-benzo[e]indol-2-ylidene)-malonaldehyde, (DDBM) showed the best binding with the NMDA receptor and computational docking data showed that DDBM antagonised the binding sites of the NMDA receptor at lower docking energies compared to other molecules. Using a rat electroconvulsive shock (ECS) model of epilepsy we showed that DDBM decreased seizure duration and improved the histological outcomes. Our data show for the first time that indolyls like DDBM have robust anticonvulsive activity and have the potential to be developed as novel anticonvulsants.


Subject(s)
Anticonvulsants/administration & dosage , Epilepsy/prevention & control , Indoles/administration & dosage , Malondialdehyde/administration & dosage , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Seizures/prevention & control , Animals , Anticonvulsants/chemical synthesis , Disease Models, Animal , Epilepsy/complications , Hippocampus/drug effects , Hippocampus/pathology , Indoles/chemical synthesis , Male , Malondialdehyde/chemical synthesis , Molecular Docking Simulation , Protein Binding , Quinazolines/chemical synthesis , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/metabolism , Seizures/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...