Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Front Aging Neurosci ; 11: 342, 2019.
Article in English | MEDLINE | ID: mdl-32009938

ABSTRACT

Alzheimer's disease (AD) is a growing global threat to healthcare in the aging population. In the USA alone, it is estimated that one in nine persons over the age of 65 years is living with AD. The pathology is marked by the accumulation of amyloid-beta (Aß) deposition in the brain, which is further enhanced by the neuroinflammatory process. Nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing 3 (NLRP3) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) are the major neuroinflammatory pathways that intensify AD pathogenesis. Histone deacetylase 2 (HDAC2)-mediated epigenetic mechanisms play a major role in the genesis and neuropathology of AD. Therefore, therapeutic drugs, which can target Aß production, NLRP3 activation, and HDAC2 levels, may play a major role in reducing Aß levels and the prevention of associated neuropathology of AD. In this study, we demonstrate that withaferin A (WA), an extract from Withania somnifera plant, significantly inhibits the Aß production and NF-κB associated neuroinflammatory molecules' gene expression. Furthermore, we demonstrate that cytokine release inhibitory drug 3 (CRID3), an inhibitor of NLRP3, significantly prevents inflammasome-mediated gene expression in our in vitro AD model system. We have also observed that mithramycin A (MTM), an HDAC2 inhibitor, significantly upregulated the synaptic plasticity gene expression and downregulated HDAC2 in SH-SY5Y cells overexpressing amyloid precursor protein (SH-APP cells). Therefore, the introduction of these agents targeting Aß production, NLRP3-mediated neuroinflammation, and HDAC2 levels will have a translational significance in the prevention of neuroinflammation and associated neurodegeneration in AD patients.

2.
Front Aging Neurosci ; 10: 291, 2018.
Article in English | MEDLINE | ID: mdl-30356847

ABSTRACT

Neurological disorders are the biggest concern globally. Out of ~36 million human immunodeficiency virus (HIV) positive people, about 30%-60% exhibit neurological disorders, including dementia and Alzheimer's disease (AD) like pathology. In AD or AD like neurological disorders, the pathogenesis is mainly due to the abnormal accumulation of extracellular amyloid beta (Aß). In this era of antiretroviral therapy, the life span of the HIV-infected individuals has increased leading towards increased neurocognitive dysfunction in nearly 30% of HIV-infected individuals, specifically older people. Deposition of the Aß plaques in the CNS is one the major phenomenon happening in aging HIV patients. ART suppresses the viral replication, but the neurotoxic protein (Tat) is still produced and results in increased levels of Aß. Furthermore, drugs of abuse like cocaine (coc) is known to induce the HIV associated neurocognitive disorders as well as the Aß secretion. To target the Tat and coc induced Aß secretion, we propose a potent bifunctional molecule Withaferin A (WA) which may act as a neuro-protectant against Aß neurotoxicity. In this study, we show that WA reduces secreted Aß and induced neurotoxicity in amyloid precursor protein (APP)-plasmid transfected SH-SY5Y cells (SH-APP). In this study, we show that in SH-APP cells, Aß secretion is induced in the presence of HIV-1 Tat (neurotoxic) and drug of abuse coc. Our fluorescent microscopy studies show the increased concentration of Aß40 in Tat (50 ng/ml) and coc (0.1 µM) treated SH-APP cells as compared to control. Our dose optimization study show, lower concentrations (0.5-2 µM) of WA significantly reduce the Aß40 levels, without inducing cytotoxicity in the SH-APP cells. Additionally, WA reduces the Tat and cocaine induced Aß levels. Therefore, we propose that Aß aggregation is induced by the presence of Tat and coc and WA is potent in reducing the secreted Aß and induced neurotoxicity. Our study provides new opportunities for exploring the pathophysiology and targeting the neurological disorders.

3.
Int J Nanomedicine ; 11: 4287-98, 2016.
Article in English | MEDLINE | ID: mdl-27621622

ABSTRACT

Although the introduction of antiretroviral therapy has reduced the prevalence of severe forms of neurocognitive disorders, human immunodeficiency virus (HIV)-1-associated neurocognitive disorders were observed in 50% of HIV-infected patients globally. The blood-brain barrier is known to be impermeable to most of antiretroviral drugs. Successful delivery of antiretroviral drugs into the brain may induce an inflammatory response, which may further induce neurotoxicity. Therefore, alternate options to antiretroviral drugs for decreasing the HIV infection and neurotoxicity may help in reducing neurocognitive impairments observed in HIV-infected patients. In this study, we explored the role of magnetic nanoparticle (MNP)-bound tissue inhibitor of metalloproteinase-1 (TIMP1) protein in reducing HIV infection levels, oxidative stress, and recovering spine density in HIV-infected SK-N-MC neuroblastoma cells. We did not observe any neuronal cytotoxicity with either the free TIMP1 or MNP-bound TIMP1 used in our study. We observed significantly reduced HIV infection in both solution phase and in MNP-bound TIMP1-exposed neuronal cells. Furthermore, we also observed significantly reduced reactive oxygen species production in both the test groups compared to the neuronal cells infected with HIV alone. To observe the effect of both soluble-phase TIMP1 and MNP-bound TIMP1 on spine density in HIV-infected neuronal cells, confocal microscopy was used. We observed significant recovery of spine density in both the test groups when compared to the cells infected with HIV alone, indicting the neuroprotective effect of TIMP1. Therefore, our results suggest that the MNP-bound TIMP1 delivery method across the blood-brain barrier can be used for reducing HIV infectivity in brain tissue and neuronal toxicity in HIV-infected patients.


Subject(s)
Anti-HIV Agents/pharmacology , HIV Infections/drug therapy , Magnetite Nanoparticles , Neuronal Plasticity/drug effects , Tissue Inhibitor of Metalloproteinase-1/pharmacology , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacokinetics , Blood-Brain Barrier/drug effects , Brain/metabolism , Cell Line , HIV-1/pathogenicity , Humans , Magnetics , Magnetite Nanoparticles/chemistry , Microscopy, Confocal , Neuroprotective Agents/chemistry , Neuroprotective Agents/pharmacokinetics , Neuroprotective Agents/pharmacology , Tissue Inhibitor of Metalloproteinase-1/chemistry , Tissue Inhibitor of Metalloproteinase-1/pharmacokinetics
4.
Sci Rep ; 6: 27864, 2016 06 20.
Article in English | MEDLINE | ID: mdl-27321752

ABSTRACT

We have observed significantly increased HIV infection in HIV infected macrophages in the presence of cocaine that could be due to the downregulation of BST2 restriction factor in these cells. In human inflammasome PCR array, among different involved in inflammasome formation, in HIV infected macrophages in the presence of cocaine, we have observed significant upregulation of NLRP3, AIM2 genes and downstream genes IL-1ß and PTGS2. Whereas negative regulatory gene MEFV was upregulated, CD40LG and PYDC1 were significantly downregulated. Among various NOD like receptors, NOD2 was significantly upregulated in both HIV alone and HIV plus cocaine treated cells. In the downstream genes, chemokine (C-C motif) ligand 2 (CCL2), CCL7 and IL-6 were significantly up regulated in HIV plus cocaine treated macrophages. We have also observed significant ROS production (in HIV and/or cocaine treated cells) which is one of the indirect-activators of inflammasomes formation. Further, we have observed early apoptosis in HIV alone and HIV plus cocaine treated macrophages which may be resultant of inflammasome formation and cspase-1 activation. These results indicate that in case of HIV infected macrophages exposed to cocaine, increased ROS production and IL-1ß transcription serve as an activators for the formation of NLRP3 and AIM2 mediated inflammasomes that leads to caspase 1 mediated apoptosis.


Subject(s)
Cocaine/pharmacology , HIV Infections/genetics , Inflammasomes/genetics , Macrophages/drug effects , Apoptosis , Caspase 1/genetics , Cells, Cultured , Cyclooxygenase 2/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , HIV Infections/metabolism , Humans , Inflammasomes/drug effects , Interleukin-1beta/genetics , Macrophages/cytology , Macrophages/metabolism , Macrophages/virology , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Reactive Oxygen Species/metabolism
5.
Mol Brain ; 9(1): 57, 2016 05 23.
Article in English | MEDLINE | ID: mdl-27216740

ABSTRACT

The human immunodeficiency virus (HIV) is a neurotropic virus. It induces neurotoxicity and subsequent brain pathologies in different brain cells. Addiction to recreational drugs remarkably affects the initiation of HIV infections and expedites the progression of acquired immunodeficiency syndrome (AIDS) associated neuropathogenesis. Symptoms of HIV-associated neurocognitive disorders (HAND) are noticed in many AIDS patients. At least 50 % of HIV diagnosed cases show one or other kind of neuropathological signs or symptoms during different stages of disease progression. In the same line, mild to severe neurological alterations are seen in at least 80 % autopsies of AIDS patients. Neurological illnesses weaken the connections between neurons causing significant altercations in synaptic plasticity. Synaptic plasticity alterations during HIV infection and recreational drug abuse are mediated by complex cellular phenomena involving changes in gene expression and subsequent loss of dendritic and spine morphology and physiology. New treatment strategies with ability to deliver drugs across blood-brain barrier (BBB) are being intensively investigated. In this context, magnetic nanoparticles (MNPs) based nanoformulations have shown significant potential for target specificity, drug delivery, drug release, and bioavailability of desired amount of drugs in non-invasive brain targeting. MNPs-based potential therapies to promote neuronal plasticity during HIV infection and recreational drug abuse are being developed.


Subject(s)
AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/therapy , Magnetite Nanoparticles/therapeutic use , Neuronal Plasticity , Substance-Related Disorders/physiopathology , Substance-Related Disorders/therapy , Animals , Drug Liberation , Humans
6.
Neural Plast ; 2015: 138979, 2015.
Article in English | MEDLINE | ID: mdl-26649202

ABSTRACT

Based on the type of cells or tissues they tend to harbor or attack, many of the viruses are characterized. But, in case of neurotropic viruses, it is not possible to classify them based on their tropism because many of them are not primarily neurotropic. While rabies and poliovirus are considered as strictly neurotropic, other neurotropic viruses involve nervous tissue only secondarily. Since the AIDS pandemic, the interest in neurotropic viral infections has become essential for all clinical neurologists. Although these neurotropic viruses are able to be harbored in or infect the nervous system, not all the neurotropic viruses have been reported to cause disrupted synaptic plasticity and impaired cognitive functions. In this review, we have discussed the neurotropic viruses, which play a major role in altered synaptic plasticity and neurological disorders.


Subject(s)
Brain/virology , DNA Viruses/physiology , Nervous System Diseases/virology , Neuronal Plasticity , RNA Viruses/physiology , Animals , Humans
7.
Front Microbiol ; 6: 946, 2015.
Article in English | MEDLINE | ID: mdl-26528242

ABSTRACT

Leishmania are obligate intracellular protozoan parasites of mammalian hosts. Promastigotes of Leishmania are internalized by macrophages and transformed into amastigotes in phagosomes, and replicate in phagolysosomes. Phagosomal maturation arrest is known to play a crucial role in the survival of pathogenic Leishmania within activated macrophages. Recently, tryptophan-aspartate containing coat (TACO) gene has been recognized as playing a central role in the survival of Mycobacterium tuberculosis within human macrophages by arresting the phagosome maturation process. We postulated that a similar association of TACO gene with phagosomes would prevent the vacuole from maturation in the case of Leishmania. In this study we attempted to define the effect of TACO gene downregulation on the entry/survival of Leishmania donovani intracellularly, by treatment with Vitamin D3 (Vit.D3)/Retinoic acid (RA) and chenodeoxycholic acid (CDCA)/RA combinations in human THP-1 macrophages (in vitro). Treatment with these molecules downregulated the TACO gene in macrophages, resulting in reduced parasite load and marked reduction of disease progression in L. donovani infected macrophages. Taken together, these results suggest that TACO gene downregulation may play a role in subverting macrophage machinery in establishing the L. donovani replicative niche inside the host. Our study is the first to highlight the important role of the TACO gene in Leishmania entry, survival and to identify TACO gene downregulation as potential drug target against leishmaniasis.

8.
Front Cell Neurosci ; 9: 212, 2015.
Article in English | MEDLINE | ID: mdl-26113810

ABSTRACT

The blood-brain barrier (BBB) is a diffusion barrier that has an important role in maintaining a precisely regulated microenvironment protecting the neural tissue from infectious agents and toxins in the circulating system. Compromised BBB integrity plays a major role in the pathogenesis of retroviral associated neurological diseases. Human Immunodeficiency Virus (HIV) infection in the Central Nervous System (CNS) is an early event even before the serodiagnosis for HIV positivity or the initiation of antiretroviral therapy (ART), resulting in neurological complications in many of the infected patients. Macrophages, microglia and astrocytes (in low levels) are the most productively/latently infected cell types within the CNS. In this brief review, we have discussed about the effect of HIV infection and viral proteins on the integrity and function of BBB, which may contribute to the progression of HIV associated neurocognitive disorders.

9.
J Neuroinflammation ; 12: 66, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25890101

ABSTRACT

BACKGROUND: Although highly active antiretroviral therapy (HAART) has significantly reduced the morbidity and mortality in HIV patients, virus continues to reside in the central nervous system (CNS) reservoir. Hence, a complete eradication of virus remains a challenge. HIV productively infects microglia/macrophages, but astrocytes are generally restricted to HIV infection. The relative importance of the possible replication blocks in astrocytes, however, is yet to be delineated. A recently identified restriction factor, sterile alpha motif and histidine/aspartic acid domain-containing protein 1 (SAMHD1), restricts HIV infection in resting CD4(+)T cells and in monocyte-derived dendritic cells. However, SAMHD1 expression and HIV-1 restriction activity regulation in the CNS cells are unknown. Though, certain miRNAs have been implicated in HIV restriction in resting CD4(+)T cells, their role in the CNS HIV restriction and their mode of action are not established. We hypothesized that varying SAMHD1 expression would lead to restricted HIV infection and host miRNAs would regulate SAMHD1 expression in astrocytes. RESULTS: We found increased SAMHD1 expression and decreased miRNA expression (miR-181a and miR-155) in the astrocytes compared to microglia. We report for the first time that miR-155 and miR-181a regulated the SAMHD1 expression. Overexpression of these cellular miRNAs increased viral replication in the astrocytes, through SAMHD1 modulation. Reactivation of HIV replication was accompanied by decrease in SAMHD1 expression. CONCLUSIONS: Here, we provide a proof of concept that increased SAMHD1 in human astrocytes is in part responsible for the HIV restriction, silencing of which relieves this restriction. At this time, this concept is of theoretical nature. Further experiments are needed to confirm if HIV replication can be reactivated in the CNS reservoir.


Subject(s)
Astrocytes/metabolism , Astrocytes/virology , HIV Infections/virology , HIV-1 , MicroRNAs/genetics , Monomeric GTP-Binding Proteins/genetics , Gene Silencing , HIV Infections/genetics , Humans , Kinetics , MicroRNAs/antagonists & inhibitors , Microglia/metabolism , Microglia/virology , SAM Domain and HD Domain-Containing Protein 1 , Virus Activation/genetics , Virus Replication/genetics
10.
J Basic Clin Physiol Pharmacol ; 26(1): 1-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25046311

ABSTRACT

To understand HIV pathogenesis or development is no simple undertaking and neither is the cell cycle which is highly complex that requires the coordination of multiple events and machinery. It is interesting that these two processes are interrelated, intersect and interact as HIV-1 infection results in cell cycle arrest at the G2 phase which is accompanied by massive CD4+ T cell death. For its own benefit, in an impressive manner and with the overabundance of tactics, HIV maneuvers DNA damage responses and cell cycle check points for viral replication at different stages from infection, to latency and to pathogenesis. Although the cell cycle is the most critical aspect involved in both viral and cellular replication, in this review, our main focus is on recent developments, including our own observations in the field of cell cycle proteins, checkpoints and strategies utilized by the viruses to manipulate these pathways to promote their own replication and survival. We will also discuss the emerging concept of targeting the replication initiation machinery for HIV therapy.


Subject(s)
Cell Cycle Checkpoints , HIV Infections/physiopathology , HIV-1/pathogenicity , CD4-Positive T-Lymphocytes/pathology , DNA Damage , HIV Infections/drug therapy , HIV Infections/virology , Humans , Virus Replication
11.
PLoS One ; 9(11): e112818, 2014.
Article in English | MEDLINE | ID: mdl-25415340

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive dysfunction of memory and higher cognitive functions with abnormal accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles throughout cortical and limbic brain regions. Withania somnifera (WS) also known as 'ashwagandha' (ASH) is used widely in Ayurvedic medicine as a nerve tonic and memory enhancer. However, there is paucity of data on potential neuroprotective effects of ASH against ß-Amyloid (1-42) (Aß) induced neuropathogenesis. In the present study, we have tested the neuroprotective effects of Methanol: Chloroform (3:1) extract of ASH and its constituent Withanolide A (WA) against Aß induced toxicity, HIV-1(Ba-L) (clade B) infection and the effects of drugs of abuse using a human neuronal SK-N-MC cell line. Aß when tested individually, induced cytotoxic effects in SK-N-MC cells as shown by increased trypan blue stained cells. However, when ASH was added to Aß treated cells the toxic effects were neutralized. This observation was supported by cellular localization of Aß, MTT formazan exocytosis, and the levels of acetylcholinesterase activity, confirming the chemopreventive or protective effects of ASH against Aß induced toxicity. Further, the levels of MAP2 were significantly increased in cells infected with HIV-1(Ba-L) (clade B) as well as in cells treated with Cocaine (COC) and Methamphetamine (METH) compared with control cells. In ASH treated cells the MAP2 levels were significantly less compared to controls. Similar results were observed in combination experiments. Also, WA, a purified constituent of ASH, showed same pattern using MTT assay as a parameter. These results suggests that neuroprotective properties of ASH observed in the present study may provide some explanation for the ethnopharmacological uses of ASH in traditional medicine for cognitive and other HIV associated neurodegenerative disorders and further ASH could be a potential novel drug to reduce the brain amyloid burden and/or improve the HIV-1 associated neurocognitive impairments.


Subject(s)
Amyloid beta-Peptides/physiology , HIV-1/physiology , Illicit Drugs/toxicity , Neuroprotective Agents/pharmacology , Peptide Fragments/physiology , Withanolides/pharmacology , Cell Line , Humans , Neurons/drug effects , Neurons/metabolism , Neurons/virology , Withania/chemistry
12.
Biomed Res Int ; 2014: 904046, 2014.
Article in English | MEDLINE | ID: mdl-25215297

ABSTRACT

Serum and urine samples were collected from 33 NCC patients before the albendazole treatment, 3-6 and 12 months PT. At 3 months PT, 24 (72.7%) patients had no detectable CT/MRI lesions and 9 (27.2%) patients had persistent lesions. Antibody response to crude soluble extract (CSE), excretory secretory (ES), and lower molecular mass (LMM) (10-30 KDa) antigenic fraction of T. solium cysticerci was detected in serum and urine samples by ELISA. Before the treatment, out of 33 NCC children, 14 (42.4%), 22 (66.6%), and 11 (33.3%) serum samples were found positive with the use of CSE, ES, and LMM antigen, respectively. At 3-6 months PT, positivity rate was 5 (15.1%), 2 (6%), and 4 (12.1%) and at 12 months PT, positivity rate was 5 (15.1%), 0, and 3 (9%) with the use of CSE, ES, and LMM antigen, respectively. There was no significant difference in the positivity with the use of three antigens in pretreatment and PT urine samples. The study suggests that the use of ES antigen to detect antibody in serum samples may serve better purpose to evaluate the therapeutic response in patients with NCC.


Subject(s)
Antibodies, Helminth/immunology , Epilepsy/immunology , Neurocysticercosis/parasitology , Taenia solium/pathogenicity , Animals , Antigens, Helminth/blood , Antigens, Helminth/immunology , Antigens, Helminth/urine , Cell Extracts/immunology , Child , Child, Preschool , Epilepsy/blood , Epilepsy/parasitology , Epilepsy/urine , Female , Follow-Up Studies , Humans , Larva/parasitology , Male , Neurocysticercosis/blood , Neurocysticercosis/immunology , Neurocysticercosis/urine , Taenia solium/drug effects , Taenia solium/immunology
13.
Mol Brain ; 7: 37, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24886748

ABSTRACT

BACKGROUND: HIV-associated neurocognitive disorder (HAND) is characterized by development of cognitive, behavioral and motor abnormalities, and occurs in approximately 50% of HIV infected individuals. In the United States, the prevalence of cigarette smoking ranges from 35-70% in HIV-infected individuals compared to 20% in general population. Cognitive impairment in heavy cigarette smokers has been well reported. However, the synergistic effects of nicotine and HIV infection and the underlying mechanisms in the development of HAND are unknown. RESULTS: In this study, we explored the role of nicotine in the progression of HAND using SK-N-MC, a neuronal cell line. SK-N-MC cells were infected with HIV-1 in the presence or absence of nicotine for 7 days. We observed significant increase in HIV infectivity in SK-N-MC treated with nicotine compared to untreated HIV-infected neuronal cells. HIV and nicotine synergize to significantly dysregulate the expression of synaptic plasticity genes and spine density; with a concomitant increase of HDAC2 levels in SK-N-MC cells. In addition, inhibition of HDAC2 up-regulation with the use of vorinostat resulted in HIV latency breakdown and recovery of synaptic plasticity genes expression and spine density in nicotine/HIV alone and in co-treated SK-N-MC cells. Furthermore, increased eIF2 alpha phosphorylation, which negatively regulates eukaryotic translational process, was observed in HIV alone and in co-treatment with nicotine compared to untreated control and nicotine alone treated SK-N-MC cells. CONCLUSIONS: These results suggest that nicotine and HIV synergize to negatively regulate the synaptic plasticity gene expression and spine density and this may contribute to the increased risk of HAND in HIV infected smokers. Apart from disrupting latency, vorinostat may be a useful therapeutic to inhibit the negative regulatory effects on synaptic plasticity in HIV infected nicotine abusers.


Subject(s)
AIDS Dementia Complex/genetics , AIDS Dementia Complex/pathology , Dendritic Spines/genetics , Gene Expression Regulation , Hydroxamic Acids/therapeutic use , Neuronal Plasticity/genetics , Nicotine/adverse effects , AIDS Dementia Complex/drug therapy , AIDS Dementia Complex/physiopathology , Cell Line, Tumor , Dendritic Spines/drug effects , Disease Progression , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation/drug effects , Gene Regulatory Networks , Histone Deacetylase 2/genetics , Histone Deacetylase 2/metabolism , Humans , Hydroxamic Acids/pharmacology , Models, Biological , Neuronal Plasticity/drug effects , Phosphorylation/drug effects , Synapses/drug effects , Synapses/metabolism , Vorinostat
14.
Free Radic Biol Med ; 69: 136-44, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24480751

ABSTRACT

Previous studies have shown that, during infection, HIV-1 clade B and clade C differentially contribute to the neuropathogenesis and development of HIV-associated neurocognitive disorders (HANDs). The low-molecular-weight tripeptide glutathione (GSH) alters the redox balance and leads to the generation of reactive oxygen species, which play a significant role in the neuropathogenesis of HANDs. We hypothesized that the HIV-1 clade B and clade C viruses and their respective Tat proteins exert differential effects on monocyte-derived immature dendritic cells (IDCs) and neuroblastoma cells (SK-N-MC) by redox activation, which leads to immunoneuropathogenesis. The GSH/GSSG ratio and mRNA expression levels and protein modification of glutathione synthetase (GSS), glutathione peroxidase 1 (GPx1), superoxide dismutase 1 (SOD1), and catalase (CAT) were analyzed in IDCs infected with HIV-1 clade B or clade C as well as in cells treated with the respective Tat proteins. The results indicated that HIV-1 clade B virus and its Tat protein significantly increased the production of reactive oxygen species and reduced the GSH/GSSG ratio and subsequent downregulation of gene expression and protein modification of GSS, GPx1, SOD1, and CAT compared to infection with the clade C virus or treatment with the clade C Tat protein. Thus, our studies demonstrate that HIV-1 clades B and C exert differential effects of redox expression and thiol modification. HIV-1 clade B potentially induces oxidative stress, leading to more immunoneuropathogenesis than infection with HIV-1 clade C.


Subject(s)
AIDS Dementia Complex/virology , HIV-1/genetics , Oxidation-Reduction , Superoxide Dismutase/metabolism , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , Astrocytes/metabolism , Astrocytes/pathology , Dendritic Cells/pathology , Glutathione/metabolism , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/administration & dosage , Superoxide Dismutase-1
15.
Nanotechnology ; 25(5): 055101, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24406534

ABSTRACT

The blood-brain barrier (BBB) is considered as the primary impediment barrier for most drugs. Delivering therapeutic agents to the brain is still a big challenge to date. In our study, a dual mechanism, receptor mediation combined with external non-invasive magnetic force, was incorporated into ferrous magnet-based liposomes for BBB transmigration enhancement. The homogenous magnetic nanoparticles (MNPs), with a size of ∼10 nm, were synthesized and confirmed by TEM and XRD respectively. The classical magnetism assay showed the presence of the characteristic superparamagnetic property. These MNPs encapsulated in PEGylated fluorescent liposomes as magneto-liposomes (MLs) showed mono-dispersion, ∼130 ± 10 nm diameter, by dynamic laser scattering (DLS) using the lipid-extrusion technique. Remarkably, a magnetite encapsulation efficiency of nearly 60% was achieved. Moreover, the luminescence and hydrodynamic size of the MLs was stable for over two months at 4 ° C. Additionally, the integrity of the ML structure remained unaffected through 120 rounds of circulation mimicking human blood fluid. After biocompatibility confirmation by cytotoxicity evaluation, these fluorescent MLs were further embedded with transferrin and applied to an in vitro BBB transmigration study in the presence or absence of external magnetic force. Comparing with magnetic force- or transferrin receptor-mediated transportation alone, their synergy resulted in 50-100% increased transmigration without affecting the BBB integrity. Consequently, confocal microscopy and iron concentration in BBB-composed cells further confirmed the higher cellular uptake of ML particles due to the synergic effect. Thus, our multifunctional liposomal magnetic nanocarriers possess great potential in particle transmigration across the BBB and may have a bright future in drug delivery to the brain.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Drug Delivery Systems , Magnetite Nanoparticles , Transferrin , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Biological Transport, Active , Cell Survival/drug effects , Cells, Cultured , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fluorescent Dyes , Humans , Liposomes/administration & dosage , Magnetics , Magnetite Nanoparticles/administration & dosage , Magnetite Nanoparticles/toxicity , Magnetite Nanoparticles/ultrastructure , Models, Biological , Nanotechnology , Particle Size , Receptors, Transferrin/metabolism , Transferrin/administration & dosage
16.
PLoS One ; 8(10): e77624, 2013.
Article in English | MEDLINE | ID: mdl-24147038

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive dysfunction of memory and higher cognitive functions with abnormal accumulation of extracellular amyloid plaques and intracellular neurofibrillary tangles throughout cortical and limbic brain regions. At present no curative treatment is available, and research focuses on drugs for slowing disease progression or providing prophylaxis. Withania somnifera (WS) also known as 'ashwagandha' is used widely in Ayurvedic medicine as a nerve tonic and memory enhancer. However, there is a paucity of data on the potential neuroprotective effects of W.somnifera against ß-Amyloid (1-42)-induced neuropathogenesis. In the present study, we have tested the neuroprotective effects of methanol:Chloroform (3:1) extract of ashwagandha against ß-amyloid induced toxicity and HIV-1Ba-L (clade B) infection using a human neuronal SK-N-MC cell line. Our results showed that ß-amyloid induced cytotoxic effects in SK-N-MC cells as shown by decreased cell growth when tested individually. Also, confocal microscopic analysis showed decreased spine density, loss of spines and decreased dendrite diameter, total dendrite and spine area in clade B infected SK-N-MC cells compared to uninfected cells. However, when ashwagandha was added to ß-amyloid treated and HIV-1 infected samples, the toxic effects were neutralized. Further, the MTT cell viability assays and the peroxisome proliferator-activated receptor-γ (PPARγ) levels supported these observations indicating the neuroprotective effect of WS root extract against ß-amyloid and HIV-1Ba-L (clade B) induced neuro-pathogenesis.


Subject(s)
Amyloid beta-Peptides/toxicity , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Plant Extracts/pharmacology , Withania/chemistry , Cell Line , Cell Survival/drug effects , Chromatography, High Pressure Liquid , Dendrites/drug effects , HIV Infections/complications , Humans , Hydro-Lyases/metabolism , Mass Spectrometry , Nervous System Diseases/etiology , Neurons/drug effects , Neuroprotective Agents/chemistry , PPAR gamma/metabolism , Plant Extracts/chemistry
17.
PLoS One ; 8(4): e62241, 2013.
Article in English | MEDLINE | ID: mdl-23653680

ABSTRACT

Parenteral use of drugs; such as opiates exert immunomodulatory effects and serve as a cofactor in the progression of HIV-1 infection, thereby potentiating HIV related neurotoxicity ultimately leading to progression of NeuroAIDS. Morphine exposure is known to induce apoptosis, down regulate cAMP response element-binding (CREB) expression and decrease in dendritic branching and spine density in cultured cells. Use of neuroprotective agent; brain derived neurotropic factor (BDNF), which protects neurons against these effects, could be of therapeutic benefit in the treatment of opiate addiction. Previous studies have shown that BDNF was not transported through the blood brain barrier (BBB) in-vivo.; and hence it is not effective in-vivo. Therefore development of a drug delivery system that can cross BBB may have significant therapeutic advantage. In the present study, we hypothesized that magnetically guided nanocarrier may provide a viable approach for targeting BDNF across the BBB. We developed a magnetic nanoparticle (MNP) based carrier bound to BDNF and evaluated its efficacy and ability to transmigrate across the BBB using an in-vitro BBB model. The end point determinations of BDNF that crossed BBB were apoptosis, CREB expression and dendritic spine density measurement. We found that transmigrated BDNF was effective in suppressing the morphine induced apoptosis, inducing CREB expression and restoring the spine density. Our results suggest that the developed nanocarrier will provide a potential therapeutic approach to treat opiate addiction, protect neurotoxicity and synaptic density degeneration.


Subject(s)
Blood-Brain Barrier/drug effects , Brain-Derived Neurotrophic Factor/pharmacology , Drug Carriers/chemistry , Endothelial Cells/drug effects , Magnetite Nanoparticles/chemistry , Morphine/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Astrocytes/cytology , Astrocytes/drug effects , Astrocytes/metabolism , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Brain/blood supply , Brain/cytology , Capillary Permeability/drug effects , Cell Count , Dendritic Spines/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Models, Biological , Morphine/pharmacology , Primary Cell Culture
18.
PLoS One ; 8(4): e61399, 2013.
Article in English | MEDLINE | ID: mdl-23620748

ABSTRACT

HIV-associated neurocognitive disorders (HAND) is characterized by development of cognitive, behavioral and motor abnormalities, and occur in approximately 50% of HIV infected individuals. Our current understanding of HAND emanates mainly from HIV-1 subtype B (clade B), which is prevalent in USA and Western countries. However very little information is available on neuropathogenesis of HIV-1 subtype C (clade C) that exists in Sub-Saharan Africa and Asia. Therefore, studies to identify specific neuropathogenic mechanisms associated with HAND are worth pursuing to dissect the mechanisms underlying this modulation and to prevent HAND particularly in clade B infection. In this study, we have investigated 84 key human synaptic plasticity genes differential expression profile in clade B and clade C infected primary human astrocytes by using RT(2) Profile PCR Array human Synaptic Plasticity kit. Among these, 31 and 21 synaptic genes were significantly (≥3 fold) down-regulated and 5 genes were significantly (≥3 fold) up-regulated in clade B and clade C infected cells, respectively compared to the uninfected control astrocytes. In flow-cytometry analysis, down-regulation of postsynaptic density and dendrite spine morphology regulatory proteins (ARC, NMDAR1 and GRM1) was confirmed in both clade B and C infected primary human astrocytes and SK-N-MC neuroblastoma cells. Further, spine density and dendrite morphology changes by confocal microscopic analysis indicates significantly decreased spine density, loss of spines and decreased dendrite diameter, total dendrite and spine area in clade B infected SK-N-MC neuroblastoma cells compared to uninfected and clade C infected cells. We have also observed that, in clade B infected astrocytes, induction of apoptosis was significantly higher than in the clade C infected astrocytes. In conclusion, this study suggests that down-regulation of synaptic plasticity genes, decreased dendritic spine density and induction of apoptosis in astrocytes may contribute to the severe neuropathogenesis in clade B infection.


Subject(s)
AIDS Dementia Complex/genetics , AIDS Dementia Complex/physiopathology , Central Nervous System/virology , Dendritic Spines/metabolism , Neuronal Plasticity/genetics , Synapses/genetics , Transcriptome , AIDS Dementia Complex/virology , Apoptosis , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Carbocyanines/metabolism , Cells, Cultured , Central Nervous System/metabolism , Central Nervous System/pathology , Dendritic Spines/pathology , Dendritic Spines/virology , Down-Regulation/genetics , Flow Cytometry , HIV-1/physiology , Humans , Membrane Proteins/metabolism , Microscopy, Confocal
19.
Acta Trop ; 118(2): 165-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21354092

ABSTRACT

Neurocysticercosis (NCC) caused by T. solium metacestode is an increasingly important health issue in Indian children. The sensitivity and specificity of available serological techniques were low in case of single cysticercus granuloma cases which is a more common feature in Indian patients who are children. Serum samples were collected from 13 clinically and radiologically suggestive NCC children and seropositive by ELISA, 25 clinically and radiologically suggestive NCC children and seronegative by ELISA and 25 control subjects. The 10-30 kDa antigens of T. solium metacestode were subjected to 2-dimensional gel electrophoresis (2D-PAGE) followed by enzyme-linked immunoelectrotransfer blot (EITB) assay to detect antibody in serum. Analysis of 10-30 kDa antigenic fraction 2D-PAGE map showed 31 proteins between 10 and ≤28 kDa and innumerable proteins between >28 and 30 kDa with the Isoelectric point of 3-10. All the 13 (100%) NCC seropositive and 15 (60%) out of 25 NCC seronegative samples were reactive with 2D fraction antigens. In the control group, none of the serum was reactive except 2 hydatid samples (92% specificity). The sensitivity and specificity of 2D-PAGE EITB assay were significantly higher than the ELISA which is the routine diagnostic method used in the endemic countries for the serodiagnosis of neurocysticercosis.


Subject(s)
Antibodies, Helminth/blood , Antigens, Helminth/analysis , Helminth Proteins/analysis , Neurocysticercosis/diagnosis , Parasitology/methods , Taenia solium/chemistry , Animals , Child , Child, Preschool , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Infant , Male , Sensitivity and Specificity , Serologic Tests/methods
20.
Mol Biochem Parasitol ; 175(1): 83-90, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20888371

ABSTRACT

Leishmania major aquaglyceroporin LmAQP1 allows adventitious passage of antimonite, an activated form of the drug Pentostam, which is used as the first line treatment for leishmaniasis. The extracellular C-loop of an aquaglyceroporin confers substrate specificity. Alteration of Glu125 to serine in the Plasmodium falciparum aquaglyceroporin PfAQP has been shown to selectively affect water but not glycerol permeability. The C-loop of LmAQP1 is twelve residues longer than PfAQP, and Ala163 is at an equivalent position as Glu125 of PfAQP. The role of Ala163 in LmAQP1 solute permeability was investigated. Alteration of Ala163 to serine or threonine did not significantly affect conduction of solutes. However, alteration to aspartate, glutamate, and glutamine blocked passage of water, glycerol, and other organic solutes. While LmAQP1 is a mercurial insensitive water channel, mutation of the adjacent threonine (Thr164) to cysteine led to inhibition of water passage by Hg(2+). This inhibition could be reversed upon addition of ß-mercaptoethanol. These data suggest that, unlike Glu125 (PfAQP), Ala163 is not involved in stabilization of the C-loop and selective solute permeability. Ala163 is located near the pore mouth of the channel, and replacement of Ala163 by bulkier residue sterically hinders the passage of solutes. Alteration of Ala163 to serine or threonine affected metalloid uptake in the order, wild-type>A163S>A163T. Metalloid conduction was near completely blocked when Ala163 was mutagenized to aspartate, glutamate, or glutamine. Mutations such as A163S and A163T that reduced the permeability to antimonite, without a significant loss in water or solute conductivity raises the possibility that, subtle changes in the side chain of the amino acid residue in position 163 of LmAQP1 may play a role in drug resistance.


Subject(s)
Alanine/genetics , Antimony/metabolism , Aquaglyceroporins/genetics , Aquaglyceroporins/metabolism , Drug Resistance , Leishmania major/genetics , Leishmania major/metabolism , Amino Acid Sequence , Amino Acid Substitution/genetics , Enzyme Inhibitors/metabolism , Glycerol/metabolism , Mercaptoethanol/metabolism , Mercury/metabolism , Metalloids/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutant Proteins/genetics , Mutant Proteins/metabolism , Organic Chemicals/metabolism , Protein Structure, Tertiary , Reducing Agents/metabolism , Water/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...