Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Future Microbiol ; 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38648093

ABSTRACT

The emergence of highly zoonotic viral infections has propelled bat research forward. The viral outbreaks including Hendra virus, Nipah virus, Marburg virus, Ebola virus, Rabies virus, Middle East respiratory syndrome coronavirus, SARS-CoV and the latest SARS-CoV-2 have been epidemiologically linked to various bat species. Bats possess unique immunological characteristics that allow them to serve as a potential viral reservoir. Bats are also known to protect themselves against viruses and maintain their immunity. Therefore, there is a need for in-depth understanding into bat-virus biology to unravel the major factors contributing to the coexistence and spread of viruses.


Bats are the most diverse mammalian order, with over 1400 species found worldwide. Studies on bats have revealed that they frequently carry and transmit multiple viruses. They are also known to recover from viral infections. Further, human interference and climatic changes in bats' native habitat have led to virus spillover events from bats to human populations, posing a serious public health risk. A deeper understanding of the coexistence of bats and viruses, as well as the mechanisms of disease transmission to humans, is required to minimize the risk of future viral outbreaks.

2.
Clin Ophthalmol ; 15: 871-879, 2021.
Article in English | MEDLINE | ID: mdl-33664564

ABSTRACT

PURPOSE: To study the level of serum malondialdehyde (MDA), a biomarker of oxidative stress before and after chemotherapy in various ocular malignancies and to correlate its significance with clinicopathological parameters. METHODS: Thirty two histopathologically confirmed cases of primary ocular malignancies were included in this longitudinal cohort study. Detailed clinicopathological assessment was done. Analysis of serum MDA level in the patient before and after chemotherapy was measured and its prognostic significance was analyzed. RESULTS: Maximum cases were of eyelid malignancy (n=18, 56.2%) followed by retinoblastoma (18.8%) and OSSN (6, 18.8%). About 43.75% patients were in the advanced-stage. The tumor was histopathologically well-differentiated in 20 (62.5%) cases. Most common malignancy was sebaceous gland carcinoma of the eyelid (n=10,31.25%). Serum MDA level in patients were significantly higher than controls [5.5712±0.32779 vs 2.5531±0.08056 nmol/mL, p<0.001]. Level was significantly reduced after chemotherapy (4.5146±0.23209 nmol/mL). Serum MDA was maximum in cases of rhabdomyosarcoma (5.9450±0.23335 nmol/mL) and retinoblastoma (5.7433±0.14334 nmol/mL). It was minimum in basal cell carcinoma (5.3775±0.17746 nmol/mL). Pre chemotherapy serum MDA level was significantly higher in patients having larger tumor (>20mm, p< 0.001) and having lymph node metastasis than those without lymph node metastasis [5.8350±0.17113 vs 5.4833± 0.32193 nmol/mL, p<0.006]. No significant difference was observed in post chemotherapy serum MDA level as for as size of tumor (p=0.947) and histopathological differentiation (p=0.109) was concerned. CONCLUSION: The serum MDA level is a potential biomarker in primary ocular carcinoma to assess oxidative stress and its impact on response to chemotherapy.

3.
Biochim Biophys Acta Gene Regul Mech ; 1863(11): 194639, 2020 11.
Article in English | MEDLINE | ID: mdl-32987149

ABSTRACT

Japanese Encephalitis Virus (JEV) is a single positive strand RNA virus, belongs to the Flaviviridae family. JEV is neurotropic in nature which accounts for 30-50% neurological, psychiatric sequelae and movement disorder, with 20-30% case fatality rate among children or elder population. JEV causes neuronal loss and microglial activation which leads to neuroinflammation. The microRNAs are the molecular switches, which regulate the gene expression post-transcriptionally. The microRNA-155 has been reported to be associated with CNS-related pathologies like, experimental autoimmune encephalitis, multiple sclerosis and amyotrophic lateral sclerosis. In the present study, we infected microglial cells with JEV, which resulted in the up-regulation of microRNA-155; quantified by real-time polymerase chain reaction. The gene target prediction databases revealed pellino 1 as a putative gene target for microRNA-155. The over-expression based studies of microRNA-155 mimics, scrambles, inhibitors, and cy3 negative control demonstrated the role of PELI1 in the regulation of the non-canonical NF-κB pathway via TRAF3. The luciferase assay showed the regulation of NF-κB promoter via microRNA-155 in JEV infected microglial cells. The suppression of NF-κB in JEV infected microglial cells led to the reduced expression of IL-6 and TNF-α. JEV exploits cellular microRNA-155 to suppress the expression of PELI1 in human microglial cells as a part of their immune evasion strategy.


Subject(s)
Encephalitis Virus, Japanese/physiology , Encephalitis, Japanese/metabolism , Encephalitis, Japanese/virology , MicroRNAs/genetics , Microglia/metabolism , NF-kappa B/metabolism , Signal Transduction , Cell Line, Tumor , Encephalitis, Japanese/genetics , Gene Expression , Gene Expression Regulation , Gene Knockdown Techniques , Host-Pathogen Interactions/genetics , Humans , Models, Biological , Protein Binding , RNA Interference
4.
Virol J ; 13: 131, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27473856

ABSTRACT

Flaviviruses are emerging arthropod-borne viruses representing an immense global health problem. The prominent viruses of this group include dengue virus, yellow fever virus, Japanese encephalitis virus, West Nile virus tick borne encephalitis virus and Zika Virus. These are endemic in many parts of the world. They are responsible for the illness ranging from mild flu like symptoms to severe hemorrhagic, neurologic and cognitive manifestations leading to death. NS1 is a highly conserved non-structural protein among flaviviruses, which exist in diverse forms. The intracellular dimer form of NS1 plays role in genome replication, whereas, the secreted hexamer plays role in immune evasion. The secreted NS1 has been identified as a potential diagnostic marker for early detection of the infections caused by flaviviruses. In addition to the diagnostic marker, the importance of NS1 has been reported in the development of therapeutics. NS1 based subunit vaccines are at various stages of development. The structural details and diverse functions of NS1 have been discussed in detail in this review.


Subject(s)
Flavivirus Infections/virology , Flavivirus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Flavivirus/chemistry , Flavivirus/genetics , Flavivirus Infections/therapy , Humans , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
5.
BMC Neurosci ; 15: 80, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24965120

ABSTRACT

BACKGROUND: Human brain microvascular endothelial cells (hBMVECs) are integral part of the blood brain barrier. Post-translational modifications of adherens junction proteins regulate the permeability of human brain microvascular endothelial cells. Pro-inflammatory signals can induce tyrosine phosphorylation of adherens junction proteins. The primary objective of this work is to provide a molecular model; how the HIV-1 Tat protein can compromise the BBB integrity and eventually lead to neurological consequences. We exposed hBMVECs to recombinant HIV-1 clade C Tat protein to study the effect of HIV-1 Tat C on permeability of hBMVECs. Trans-endothelial electrical resistance and fluorescent dye migration assay have been used to check the permeability of hBMVECs. DCFDA staining has been used for intracellular reactive oxygen species (ROS) detection. Western blotting has been used to study the expression levels and co-immunoprecipitation has been used to study the interactions among adherens junction proteins. RESULTS: HIV-1 Tat C protein induced NOX2 and NOX4 expression level and increased intracellular ROS level. Redox-sensitive kinase; PYK2 activation led to increased tyrosine phosphorylation of VE-cadherin and ß-catenin, leading to disruption of junctional assembly. The dissociation of tyrosine phosphatases VE-PTP and SHP2 from cadherin complex resulted into increased tyrosine phosphorylation of VE-cadherin and ß-catenin in HIV-1 Tat C treated hBMVECs. CONCLUSION: Unrestricted phosphorylation of junctional proteins in hBMVECs, in response to HIV-1 Tat C protein; leads to the disruption of junctional complexes and increased endothelial permeability.


Subject(s)
Antigens, CD/metabolism , Blood-Brain Barrier/metabolism , Brain/metabolism , Cadherins/metabolism , Endothelium, Vascular/metabolism , HIV-1/metabolism , Microvessels/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Blotting, Western , Brain/blood supply , Capillary Permeability/physiology , Cells, Cultured , Electric Impedance , Endothelial Cells/metabolism , Fluoresceins , Fluorescent Dyes , Humans , Immunoprecipitation , Phosphorylation , Reactive Oxygen Species/metabolism , beta Catenin/metabolism
6.
J Neurosci ; 33(14): 5992-6000, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23554480

ABSTRACT

HIV-1 infection leads to the development of HIV-associated neurological disorders. The HIV-1 Tat protein has been reported to exert an adverse effect on blood-brain barrier integrity and permeability. Perturbation in permeability is mainly caused by disruptions in adherens junctions and tight junction proteins. We have identified HIV-1 Tat C-induced disruption of VE-cadherin mediated by miRNA-101 in human brain microvascular endothelial cells (BMVECs). HIV-1 Tat C increased the expression of miR-101, which led to downregulation of VE-cadherin. Overexpression of miR-101 resulted into the suppression of VE-cadherin. Inhibition of miR-101 by the miRNA inhibitor enhanced the expression of VE-cadherin. We have demonstrated that VE-cadherin is a direct target of miR-101 using a luciferase reporter assay, which showed that mutated VE-cadherin 3'UTR and miR-101 cotransfection did not change luciferase activity. By overexpression and knockdown of miR-101, we have demonstrated that the expression level of claudin-5 is governed by the expression of VE-cadherin. These findings demonstrate a novel mechanism for the regulation of barrier permeability by miR-101 via posttranscriptional regulation of VE-cadherin in human BMVECs exposed to the HIV-1 Tat C protein.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , MicroRNAs/metabolism , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Brain/anatomy & histology , Capillary Permeability/drug effects , Cells, Cultured , Claudin-5/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/metabolism , Gap Junctions/drug effects , Gap Junctions/metabolism , Green Fluorescent Proteins/metabolism , Humans , MicroRNAs/genetics , Microvessels/cytology , Models, Biological , RNA-Binding Proteins/metabolism , Transfection , Zonula Occludens-1 Protein/metabolism , tat Gene Products, Human Immunodeficiency Virus/genetics
7.
J Neuroinflammation ; 9: 131, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22709905

ABSTRACT

BACKGROUND: HIV-1 Tat protein is known to be associated with neuroinflammation, a condition that develops in almost half of patients infected with HIV-1. HIV-1 Tat can alter glial neuroprotective functions, leading to neurotoxicity within the CNS. HIV-1 Tat is known to be secreted from productively infected cells and can affect neighboring uninfected cells by modulating cellular gene expression in a bystander fashion. METHODS: We were interested to study whether exogenous exposure to HIV-1 Tat-C protein perturbs the microRNA (miRNA) expression profile of human microglial cells, leading to altered protein expression. We used protein expression and purification, miRNA overexpression, miRNA knockdown, transfection, site-directed mutagenesis, real-time PCR, luciferase assay and western blotting techniques to perform our study. RESULTS: HIV-1 Tat-C treatment of human microglial cells resulted in a dose-dependent increase in miR-32 expression. We found that tumor necrosis factor-receptor-associated factor 3 TRAF3) is a direct target for miR-32, and overexpression of miR-32 in CHME3 cells decreased TRAF3 both at the mRNA and the protein level. Recovery of TRAF3 protein expression after transfection of anti-miR-32 and the results of the luciferase reporter assay provided direct evidence of TRAF3 regulation by miR-32. We found that the regulation of interferon regulatory factor 3 (IRF3) and IRF7 is controlled by cellular levels of TRAF3 protein in microglial cells, as after overexpression of miR-32 and application of anti-miR-32, expression levels of IRF3 and IRF7 were inversely regulated by expression levels of TRAF3. Thus, our results suggest a novel miRNA mediated mechanism for regulation of TRAF3 in human microglial cells exposed to HIV-1 Tat C protein. These results may help to elucidate the detrimental neuroinflammatory consequences of HIV-1 Tat C protein in bystander fashion. CONCLUSION: HIV-1 Tat protein can modulate TRAF3 expression through miRNA mediated pathway and can change the downstream expression of IRF3 and IRF7. This study demonstrates a novel mechanism of HIV-1 Tat C protein-mediated perturbation of miRNA, resulting in dysregulation of cellular TRAF3.


Subject(s)
HIV-1/physiology , MicroRNAs/physiology , Microglia/physiology , TNF Receptor-Associated Factor 3/metabolism , tat Gene Products, Human Immunodeficiency Virus/physiology , Gene Expression Regulation/immunology , HeLa Cells , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Microglia/drug effects , TNF Receptor-Associated Factor 3/biosynthesis
8.
Drug Discov Today ; 16(21-22): 948-56, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21889604

ABSTRACT

The AIDS vaccine development effort has already been facing various scientific and economic challenges. The fundamental challenge resides at the level of understanding the basic biology of HIV-1 infection and an effective antiviral immune response. There is a need to design immunogens that can elicit cross-clade neutralizing antibodies (NAbs) along with effective T-cell responses against a wide variety of primary HIV isolates. We must exploit the capabilities of the vaccine-elicited cytotoxic T cells and the NAb responses in controlling HIV-1 replication. A coordinated approach is required to understand the intricacies involved in the basic immune responses against HIV infection as well as the cross-clade effectiveness of an AIDS vaccine.


Subject(s)
AIDS Vaccines/immunology , Drug Discovery , HIV Infections/immunology , HIV Infections/prevention & control , AIDS Vaccines/metabolism , AIDS Vaccines/therapeutic use , HIV Antibodies/immunology , HIV Antibodies/physiology , HIV-1/immunology , HIV-1/physiology , Humans
9.
Rev Med Virol ; 21(2): 78-88, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21412934

ABSTRACT

Chikungunya is a re-emerging arthropod-borne viral disease caused by Chikungunya virus (CHIKV) belonging to the Togaviridae family of genus Alphavirus. It is a virus with a single stranded, positive sense RNA, as its genome. It is maintained in a sylvatic and urban cycle involving humans and the mosquito species Aedes aegypti and Aedes albopictus. It has garnered the attention of scientists in the past 5-6 years due to the massive outbreaks in the Indian Ocean region in 2005-2006. It has a major health impact on humans as it causes fever, rashes, arthralgia and myalgia. Polyarthralgia is the most important feature of CHIKV infection which primarily affects the small joints of the wrists and fingers along with the large joints like shoulders and knees. Currently, there are no vaccines or treatment regimens available for CHIKV infection. The molecular mechanism underlying the chronic polyarthralgia observed in patients is not well understood. In this review we have summarized the CHIKV organization, replication, epidemiology, clinical manifestations and pathogenesis with emphasis on the arthralgia.


Subject(s)
Chikungunya Fever/virology , Chikungunya virus/physiology , Host-Pathogen Interactions , Aedes/virology , Animals , Arthralgia/epidemiology , Arthralgia/pathology , Arthralgia/virology , Chikungunya Fever/epidemiology , Chikungunya Fever/pathology , Disease Outbreaks , Humans , Virus Replication
10.
Drug Discov Today ; 14(17-18): 851-8, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19540928

ABSTRACT

RNA interference (RNAi) is a novel and essential biological process, as well as a powerful experimental tool with the potential to be used in therapeutic development. RNAi-based strategies have the capability of being able to be driven from bench to bedside. It is very important to develop the precise tools for designing the siRNAs to get the most efficient knockdown of the target genes and to reduce any off-target effects. In this review we have discussed the strategies and parameters required for effective siRNA designing and synthesis, based on already published literature.


Subject(s)
Drug Design , Gene Knockdown Techniques/standards , RNA, Small Interfering/chemical synthesis , Drug Stability , Humans , Models, Genetic , RNA Interference/drug effects , RNA, Small Interfering/therapeutic use , Software
11.
Drug Discov Today ; 14(17-18): 859-65, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19540929

ABSTRACT

RNA interference (RNAi) is a sequence-specific mechanism to control the expression of target genes. This technique has proven potentials both in vivo and in vitro. The main hurdle for using RNAi-based therapy is the effective delivery of RNAi-based drugs to the target cells or tissues in vivo. The aspects of off-target effects, delivery methods, induction of immune response and dose determination for delivery should, however, be considered carefully. If these challenges associated with siRNA can be met, then the potentials of RNAi could be exploited to the full for the development of therapeutic tools and drugs.


Subject(s)
Drug Delivery Systems/methods , RNA, Small Interfering/administration & dosage , Gene Transfer Techniques , Genetic Therapy/methods , Humans , RNA Interference/drug effects , RNA, Small Interfering/therapeutic use
13.
Pharmacogenomics ; 8(8): 971-8, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17716230

ABSTRACT

miRNAs are reported to sequence-specifically control the translation of target mRNAs by binding to 3 UTRs. The abundant expression of miRNAs in the brain highlights their biological significance in neurodevelopment. Many studies have shown that miRNAs are involved in a variety of functions, including developmental transitions and neuronal patterning, apoptosis, fat metabolism and regulation of hematopoietic lineage differentiation in different organisms. miRNAs act as regulatory switches in the determination of developmental fate through their distinct patterns of expression. The tissue-specific expression of miRNAs during brain development could possibly direct the development of cells in different subtypes. Several miRNAs are localized to neuronal subtypes and exhibit a more diverse or specific expression pattern within various neuronal cell types such as glial cells and neuronal progenitor cells. Perturbations in the expression pattern of miRNAs could lead to defects in human brain development and neurological disorders. The bioinformatic prediction tools suggest that some genes involved in synaptic formations and mental retardation are putative targets for miRNAs. miRNAs have been shown to specify cell fates in the nervous system of worms and brain morphogenesis in fish, and their distinct expression patterns during mammalian brain development. This suggests a potential role of miRNAs in neurodevelopment of mammals and other organisms. In this review, I have focused on the role of miRNAs in brain development and possible neurological disorders.


Subject(s)
MicroRNAs/genetics , MicroRNAs/physiology , Nerve Degeneration/genetics , Neurons/physiology , Stem Cells/physiology , Animals , Gene Expression Regulation, Developmental/physiology , Humans , MicroRNAs/biosynthesis , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Stem Cells/cytology , Stem Cells/metabolism , Stem Cells/pathology
14.
Future Microbiol ; 2(3): 269-75, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17661701

ABSTRACT

Endogenous retroviruses (ERVs) integrated into human genome after cross-species infectious events millions of years ago. Few of the ERVs retain the basic structure of the integrated proviral form of infectious retroviruses. Human endogenous retroviruses (HERVs) represent a key molecular link between the host genome and infectious viral particles. They constitute a large reservoir of viral genes that could be activated by various factors such as mutations or carcinogenic chemical exposures and so forth. Endogenous retroviruses are transmitted genetically in a Mendelian fashion through the germline as proviral DNA. Expression pattern of HERVs can influence the outcome of their presence in many forms, which can be either beneficial or detrimental to the host. ERVs have been reported to play a role in both human physiology and human pathology. Along with their expression in many disease conditions, they have also been reported to be expressed on normal peripheral blood lymphocytes, salivary glands and keratinocytes.


Subject(s)
Endogenous Retroviruses/genetics , Genes, Viral , Genome, Human/genetics , Endogenous Retroviruses/physiology , Humans , Models, Genetic , Virus Integration
15.
Lancet Infect Dis ; 4(9): 575-83, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15336225

ABSTRACT

Lyme disease is a tick-transmitted disease caused by the spirochete Borrelia burgdorferi. The bacterium adopts different strategies for its survival inside the immunocompetent host from the time of infection until dissemination in different parts of body tissues. The success of this spirochete depends on its ability to colonise the host tissues and counteract the host's defence mechanisms. During this process borrelia seems to maintain its vitality to ensure long-term survival in the host. Borrelia's proteins are encoded by plasmid and chromosomal genes. These genes are differentially regulated and expressed by different environmental factors in ticks as well as in the mammalian host during infection. In addition, antigenic diversity enables the spirochete to escape host defence mechanisms and maintain infection. In this review we focus on the differential expression of proteins and genes, and further molecular mechanisms used by borrelia to maintain its survival in the host. In light of these pathogenetic mechanisms, further studies on spirochete host interaction are needed to understand the complex interplay that finally lead to host autoimmunity.


Subject(s)
Arachnid Vectors/microbiology , Borrelia burgdorferi/physiology , Gene Expression Regulation, Bacterial , Lyme Disease/immunology , Lyme Disease/microbiology , Ticks/microbiology , Adaptation, Physiological , Animals , Antigen-Antibody Complex/immunology , Antigen-Antibody Complex/metabolism , Antigenic Variation , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Borrelia burgdorferi/genetics , Borrelia burgdorferi/immunology , Host-Parasite Interactions , Humans , Lyme Disease/transmission , Polymorphism, Genetic , Recombination, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...