Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Methods Mol Biol ; 2807: 271-283, 2024.
Article in English | MEDLINE | ID: mdl-38743235

ABSTRACT

The blood-brain barrier (BBB) is one of several barriers between the brain and the peripheral blood system to maintain homeostasis. Understanding the interactions between infectious agents such as human immunodeficiency virus type 1 (HIV-1), which are capable of traversing the BBB and causing neuroinflammation requires modeling an authentic BBB in vitro. Such an in vitro BBB model also helps develop means of targeting viruses that reside in the brain via natural immune effectors such as antibodies. The BBB consists of human brain microvascular endothelial cells (HBMECs), astrocytes, and pericytes. Here we report in vitro methods to establish a dual-cell BBB model consisting of primary HBMECs and primary astrocytes to measure the integrity of the BBB and antibody penetration of the BBB, as well as a method to establish a single cell BBB model to study the impact of HIV-1 infected medium on the integrity of such a BBB.


Subject(s)
Astrocytes , Blood-Brain Barrier , Endothelial Cells , HIV Infections , HIV-1 , Blood-Brain Barrier/virology , Blood-Brain Barrier/metabolism , Humans , Astrocytes/virology , Astrocytes/metabolism , Astrocytes/immunology , Endothelial Cells/virology , Endothelial Cells/metabolism , Endothelial Cells/immunology , HIV-1/immunology , HIV-1/physiology , HIV Infections/virology , HIV Infections/immunology , Pericytes/virology , Pericytes/metabolism , Pericytes/immunology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/immunology , Coculture Techniques/methods , Cells, Cultured , Brain/virology , Brain/immunology , Brain/metabolism
2.
J Infect Dis ; 227(Suppl 1): S58-S61, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36930635

ABSTRACT

Despite effective suppressive antiretroviral therapy, central nervous system (CNS) complications related to human immunodeficiency virus (HIV) remain a significant problem for people with HIV (PWH). Numerous studies have contributed data to define the mechanisms underlying HIV-associated CNS pathophysiology, but causality remains elusive, with no effective therapies to prevent, reduce, or reverse HIV-associated CNS complications. Multiple physiological, clinical, cognitive, behavioral, social, and environmental factors contribute to the observed heterogeneity of adverse CNS outcomes among PWH. The National Institute of Mental Health in collaboration with investigators engaged in research related to HIV associated CNS complications organized a series of meetings to review the state of the science and facilitate the development of biologically based measures to identify the phenotypic heterogeneity of CNS outcomes linked to pathophysiology (biotypes). In this article, we summarize the proceedings of these meetings and explore the precision medicine framework to identify critical factors linked to the etiopathogenesis of CNS outcomes in PWH.


Subject(s)
HIV Infections , HIV-1 , United States/epidemiology , Humans , National Institute of Mental Health (U.S.) , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/pathology , Central Nervous System , Delivery of Health Care
3.
J Leukoc Biol ; 112(5): 1233-1243, 2022 11.
Article in English | MEDLINE | ID: mdl-36073341

ABSTRACT

Macrophages play a significant role in HIV infection and contribute to pathogenesis of comorbidities as well as establishment of the viral reservoir in people living with HIV. While CD4+ T cells are considered the main targets of HIV infection, infected macrophages resist the cytopathic effects of infection, contributing to the persistent HIV reservoir. Furthermore, activated macrophages drive inflammation and contribute to the development of comorbidities, including HIV-associated CNS dysfunction. Better understanding the role of macrophages in HIV infection, persistence, and comorbidities can lead to development of innovative therapeutic strategies to address HIV-related outcomes in people living with HIV. In October 2021, the National Institute of Mental Health and the Ragon Institute of MGH, MIT, and Harvard conducted a virtual meeting on role of macrophages in HIV infection, pathogenesis, and cure. This review article captures the key highlights from this meeting and provides an overview of interests and activities of various NIH institutes involved in supporting research on macrophages and HIV.


Subject(s)
HIV Infections , Humans , HIV Infections/drug therapy , Virus Latency , Macrophages/pathology , CD4-Positive T-Lymphocytes
5.
Elife ; 82019 06 07.
Article in English | MEDLINE | ID: mdl-31172941

ABSTRACT

Cellular ESCRT machinery plays pivotal role in HIV-1 budding and release. Extracellular stimuli that modulate HIV-1 egress are currently unknown. We found that CCL2 induced by HIV-1 clade B (HIV-1B) infection of macrophages enhanced virus production, while CCL2 immuno-depletion reversed this effect. Additionally, HIV-1 clade C (HIV-1C) was refractory to CCL2 levels. We show that CCL2-mediated increase in virus production requires Gag late motif LYPX present in HIV-1B, but absent in HIV-1C, and ALIX protein that recruits ESCRT III complex. CCL2 immuno-depletion sequestered ALIX to F-actin structures, while CCL2 addition mobilized it to cytoplasm facilitating Gag-ALIX binding. The LYPX motif improves virus replication and its absence renders the virus less fit. Interestingly, novel variants of HIV-1C with PYRE/PYKE tetrapeptide insertions in Gag-p6 conferred ALIX binding, CCL2-responsiveness and enhanced virus replication. These results, for the first time, indicate that CCL2 mediates ALIX mobilization from F-actin and enhances HIV-1 release and fitness.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Chemokine CCL2/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , HIV-1/growth & development , Host-Pathogen Interactions , Virus Release , gag Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Humans , Macrophages/virology
6.
J Virus Erad ; 2(4): 243-250, 2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27781109

ABSTRACT

CSF HIV escape is a recently recognised phenomenon that suggests that despite suppressive treatment, HIV RNA may be detected in the CNS compartment in some individuals. In rare cases this is associated with clinical neurological disease, while in most cases, neurological consequences are not apparent. Attempts at characterising the biological substrates of CSF escape and further investigating the neurological consequences need to be made to better understand the implications of this condition for the HIV cure agenda as well as for clinical outcomes. The Global CSF HIV-1 Escape Consortium meeting, convened by the US National Institute of Mental Health, was a first step to gather investigators from diverse sites to discuss opportunities for future collaborative work on this emerging issue. To better understand CSF HIV escape and allow cross-site data reconciliation, it will be useful to reach a consensus set of definitions of the distinct forms of CSF escape, without which concerted cross-site efforts are difficult.

7.
Curr HIV Res ; 14(5): 389-399, 2016.
Article in English | MEDLINE | ID: mdl-27009096

ABSTRACT

BACKGROUND: Over the past three decades, the clinical presentation of HIV infection of the Central Nervous System (CNS) has evolved. Prior to wide spread use of effective antiretroviral therapy (ART), more than a third of infected individuals exhibited a range of neurocognitive and motor deficits that frequently progressed to severe dementia and paralysis. However, the use of ART has significantly decreased the prevalence of severe forms of HIV-1 associated neurocognitive disorders (HAND). Studies of neurocognitive dysfunction have reported variable prevalence, ranging from 21% to 77.6%, defined primarily by mild to moderate neurocognitive impairment. HIV-associated chronic inflammation and associated neurotoxicity of long term ART, as well as the aging of the HIV-infected population, likely influence the pathogenesis of HAND. Despite significant research efforts directed towards a better understanding of the mechanisms underlying HIV neuropathogenesis, definitive causal pathophysiology of HAND and thus effective prevention or treatment remain elusive. Furthermore, HIV therapeutic research now includes efforts to effect a cure, by eliminating or silencing HIV within infected cells, which must include efforts to target the latently infected cells within the CNS. CONCLUSION: Prevention and treatment of the neurological complications of HIV, and eradication of persistent virus from the CNS compartment are major priorities for the HIV-CNS research. Here we give an overview of the progress of research on HIV-CNS disease, define new challenges and research areas, and highlight domestic and global priorities.


Subject(s)
AIDS Dementia Complex/prevention & control , AIDS Dementia Complex/physiopathology , HIV Infections/complications , HIV-1/pathogenicity , Host-Pathogen Interactions , Biomedical Research/trends , Humans
8.
Methods Mol Biol ; 1354: 367-76, 2016.
Article in English | MEDLINE | ID: mdl-26714725

ABSTRACT

Despite the inability of HIV-1 to infect neurons, over half of the HIV-1-infected population in the USA suffers from neurocognitive dysfunction. HIV-infected immune cells in the periphery enter the central nervous system by causing a breach in the blood-brain barrier. The damage to the neurons is mediated by viral and host toxic products released by activated and infected immune and glial cells. To evaluate the toxicity of any viral isolate, viral protein, or host inflammatory protein, we describe a protocol to assess the neuronal apoptosis and synaptic compromise in primary cultures of human neurons and astrocytes.


Subject(s)
HIV Infections/complications , HIV-1/physiology , Neurons/pathology , Neurons/virology , Neurotoxicity Syndromes/virology , Retroviridae Proteins/metabolism , Apoptosis , Astrocytes/pathology , Astrocytes/virology , Cell Culture Techniques/methods , Cells, Cultured , HIV Infections/pathology , HIV Infections/virology , Humans , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/pathology , Viral Proteins
9.
PLoS One ; 9(9): e107074, 2014.
Article in English | MEDLINE | ID: mdl-25188269

ABSTRACT

Regional differences in neurovirulence have been documented among subtype/clade-C HIV-1 isolates in India and Southern Africa. We previously demonstrated that a C31S substitution in Clade-C Tat dicysteine motif reduces monocyte recruitment, cytokine induction and direct neurotoxicity. Therefore, this polymorphism is considered to be a causative factor for these differences in neurovirulence. We previously reported on the genotypic differences in Tat protein between clade-C and rest of the clades showing that approximately 90% of clade-C HIV-1 Tat sequences worldwide contained this C31S polymorphism, while 99% of non-clade C isolates lacked this Tat polymorphism at C31 residue (Ranga et al. (2004) J Virol 78:2586-2590). Subsequently, we documented intra-clade-C differences in the frequency of Tat dicysteine variants between India and Southern Africa, as the basis for differential disease severity and showed the importance of the Tat dicysteine motif for neuropathogenesis using small animal models. We have now examined if determinants of neurovirulence besides Tat are different between the clade-C HIV-1 isolates from Southern Africa and India. Envelope glycoprotein gp120 is a well-documented contributor to neurotoxicity. We found that gp120 sequences of HIV-1 isolates from these two regions are genetically distinct. In order to delineate the contribution of gp120 to neurovirulence, we compared direct in vitro neurotoxicity of HIV-infected supernatants of a representative neurovirulent US clade-B isolate with two isolates each from Southern Africa and India using primary human neurons and SH-SY5Y neuroblastoma cells. Immunodepletion of gp120 of both US clade B and the Southern African clade C isolates revealed robust decreases in neurotoxicity, while that of the Indian isolates showed minimal effect on neurotoxicity. The gp120 as a cause of differential neurotoxicity was further confirmed using purified recombinant gp120 from HIV isolates from these regions. We conclude that gp120 is one of the key factors responsible for the decreased neurovirulence of Indian clade C HIV-1 isolates when compared to South African clade C HIV-1.


Subject(s)
HIV Envelope Protein gp120/genetics , HIV-1/genetics , HIV-1/pathogenicity , Neurons/drug effects , Polymorphism, Genetic , Africa, Southern , Cell Line, Tumor , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cerebral Cortex/virology , Culture Media, Conditioned/toxicity , Fetus , Gene Expression , Genotype , HIV Envelope Protein gp120/chemistry , HIV Envelope Protein gp120/metabolism , HIV-1/classification , HIV-1/metabolism , Humans , India , Molecular Typing , Neurons/pathology , Neurons/virology , Phylogeny , Phylogeography , Primary Cell Culture , Virulence
10.
AIDS ; 28(15): 2319-22, 2014 Sep 24.
Article in English | MEDLINE | ID: mdl-25102091

ABSTRACT

A novel tetra-peptide insertion was identified in Gag-p6 ALIX-binding region, which appeared in protease inhibitor failure Indian HIV-1C sequences (odds ratio=17.1, P < 0.001) but was naturally present in half of untreated Ethiopian HIV-1C sequences. The insertion is predicted to restore ALIX-mediated virus release pathway, which is lacking in HIV-1C. The clinical importance of the insertion needs to be evaluated in HIV-1C dominating regions wherein the use of protease inhibitor drugs are being scaled up.


Subject(s)
Drug Resistance, Viral , HIV Infections/drug therapy , HIV Infections/virology , HIV Protease Inhibitors/therapeutic use , HIV-1/genetics , Mutagenesis, Insertional , gag Gene Products, Human Immunodeficiency Virus/genetics , Adult , Cohort Studies , Female , Genotype , HIV-1/classification , HIV-1/isolation & purification , Humans , India , Male , Middle Aged , Treatment Failure
11.
AIDS Res Ther ; 11: 13, 2014.
Article in English | MEDLINE | ID: mdl-24894206

ABSTRACT

As the HIV-1 epidemic enters its fourth decade, HIV-1 associated neurological disorders (HAND) continue to be a major concern in the infected population, despite the widespread use of anti-retroviral therapy. Advancing age and increased life expectancy of the HIV-1 infected population have been shown to increase the risk of cognitive dysfunction. Over the past 10 years, there has been a significant progress in our understanding of the mechanisms and the risk factors involved in the development of HAND. Key events that lead up to neuronal damage in HIV-1 infected individuals can be categorized based on the interaction of HIV-1 with the various cell types, including but not limited to macrophages, brain endothelial cells, microglia, astrocytes and the neurons. This review attempts to decipher these interactions, beginning with HIV-1 infection of macrophages and ultimately resulting in the release of neurotoxic viral and host products. These include: interaction with endothelial cells, resulting in the impairment of the blood brain barrier; interaction with the astrocytes, leading to metabolic and neurotransmitter imbalance; interactions with resident immune cells in the brain, leading to release of toxic cytokines and chemokines. We also review the mechanisms underlying neuronal damage caused by the factors mentioned above. We have attempted to bring together recent findings in these areas to help appreciate the viral and host factors that bring about neurological dysfunction. In addition, we review host factors and viral genotypic differences that affect phenotypic pathological outcomes, as well as recent advances in treatment options to specifically address the neurotoxic mechanisms in play.

12.
Retrovirology ; 10: 61, 2013 Jun 08.
Article in English | MEDLINE | ID: mdl-23758766

ABSTRACT

BACKGROUND: HIV-1 Clade C (Subtype C; HIV-1C) is responsible for greater than 50% of infections worldwide. Unlike clade B HIV-1 (Subtype B; HIV-1B), which is known to cause HIV associated dementia (HAD) in approximately 15% to 30% of the infected individuals, HIV-1C has been linked with lower prevalence of HAD (0 to 6%) in India and Ethiopia. However, recent studies report a higher prevalence of HAD in South Africa, Zambia and Botswana, where HIV-1C infections predominate. Therefore, we examined whether Southern African HIV-1C is genetically distinct and investigated its neurovirulence. HIV-1 Tat protein is a viral determinant of neurocognitive dysfunction. Therefore, we focused our study on the variations seen in tat gene and its contribution to HIV associated neuropathogenesis. RESULTS: A phylogenetic analysis of tat sequences of Southern African (South Africa and Zambia) HIV isolates with those from the geographically distant Southeast Asian (India and Bangladesh) isolates revealed that Southern African tat sequences are distinct from Southeast Asian isolates. The proportion of HIV - 1C variants with an intact dicysteine motif in Tat protein (C30C31) was significantly higher in the Southern African countries compared to Southeast Asia and broadly paralleled the high incidence of HAD in these countries. Neuropathogenic potential of a Southern African HIV-1C isolate (from Zambia; HIV-1C 1084i), a HIV-1C isolate (HIV-1 IndieC1) from Southeast Asia and a HIV-1B isolate (HIV-1 ADA) from the US were tested using in vitro assays to measure neurovirulence and a SCID mouse HIV encephalitis model to measure cognitive deficits. In vitro assays revealed that the Southern African isolate, HIV-1C 1084i exhibited increased monocyte chemotaxis and greater neurotoxicity compared to Southeast Asian HIV-1C. In neurocognitive tests, SCID mice injected with MDM infected with Southern African HIV-1C 1084i showed greater cognitive dysfunction similar to HIV-1B but much higher than those exposed to Southeast Asian HIV - 1C. CONCLUSIONS: We report here, for the first time, that HIV-1C from Southern African countries is genetically distinct from Southeast Asian HIV-1C and that it exhibits a high frequency of variants with dicysteine motif in a key neurotoxic HIV protein, Tat. Our results indicate that Tat dicysteine motif determines neurovirulence. If confirmed in population studies, it may be possible to predict neurocognitive outcomes of individuals infected with HIV-1C by genotyping Tat.


Subject(s)
AIDS Dementia Complex/epidemiology , AIDS Dementia Complex/virology , HIV Infections/complications , HIV Infections/virology , HIV-1/genetics , tat Gene Products, Human Immunodeficiency Virus/genetics , Adult , Africa, Southern/epidemiology , Animals , Asia, Southeastern/epidemiology , Female , Genotype , HIV-1/classification , HIV-1/isolation & purification , HIV-1/pathogenicity , Humans , Male , Mice , Middle Aged , Molecular Sequence Data , Prevalence , Sequence Analysis, DNA
13.
Methods Mol Biol ; 485: 295-309, 2009.
Article in English | MEDLINE | ID: mdl-19020833

ABSTRACT

HIV-associated dementia (HAD) is a multi-factorial disease set in motion by the presence of HIV-infected cells in the brain. A characteristic feature of HAD is the infiltration of mononuclear phagocytes into the brain, which is aided by HIV-1 Tat protein and other chemokines secreted by both HIV-infected cells and uninfected cells in their vicinity. Both direct and indirect chemokine activity of HIV-1 Tat protein has been demonstrated employing purified recombinant Tat protein. However, a corroboration of a key role for Tat or other chemokines in monocyte migration, in the context of HIV-infection, has not yet been demonstrated. Here we describe methods, to measure the role of soluble factors, such as chemokines and Tat, released by HIV-infected cells or uninfected cells in their vicinity, in monocyte migration in vitro.


Subject(s)
Cell Migration Assays, Leukocyte/methods , Cell Movement , HIV-1/immunology , Monocytes/immunology , Cell Line , Cells, Cultured , Chemokines/immunology , Humans , tat Gene Products, Human Immunodeficiency Virus/immunology
14.
J Neurosci ; 28(40): 10010-6, 2008 Oct 01.
Article in English | MEDLINE | ID: mdl-18829958

ABSTRACT

Human immunodeficiency virus (HIV)-associated dementia (HAD) is common among clade B HIV-infected individuals, but less common and less severe among individuals infected with clade C HIV-1, suggesting clade-specific differences in neuropathogenicity. Although differences in neuropathogenicity have been investigated in vitro using viral proteins responsible for HAD, to date there are no virological studies using animal models to address this issue. Therefore, we investigated neuropathogenesis induced by HIV-1 clades using the severe combined immune deficiency (SCID) mouse HIV encephalitis model, which involves intracranial injection of macrophages infected with representative clade B (HIV-1(ADA)) or clade C (HIV-1(Indie-C1)) HIV-1 isolates into SCID mice. In cognitive tests, mice exposed to similar inputs of HIV-1 clade C made fewer memory errors than those exposed to HIV-1 clade B. Histopathological analysis of mice exposed to clade B exhibited greater astrogliosis and increased loss of neuronal network integrity. In vitro experiments revealed differences in a key characteristic of HIV-1 that influences HAD, increased monocyte infiltration. HIV-1(Indie-C1)-infected macrophages recruited monocytes poorly in vitro compared with HIV-1(ADA)-infected macrophages. Monocyte recruitment was HIV-1 Tat and CCL2 dependent. This is the first demonstration, ever since HIV neuropathogenesis was first recognized, that viral genetic differences between clades can affect disease severity and that such studies help identify key players in neuropathogenesis by HIV-1.


Subject(s)
HIV Infections/pathology , HIV Infections/virology , HIV-1/physiology , AIDS Dementia Complex/etiology , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Animals , Cells, Cultured , Gene Products, tat/physiology , HIV Infections/etiology , HIV-1/isolation & purification , Humans , Mice , Mice, Inbred C57BL , Mice, SCID
15.
J Biol Chem ; 281(11): 7421-8, 2006 Mar 17.
Article in English | MEDLINE | ID: mdl-16423828

ABSTRACT

Single-base deletions at nucleotide runs or -1 frameshifting by human immunodeficiency virus type 1 (HIV-1) reverse transcriptase (RT) result from template slippage during polymerization. In crystal structures of HIV-1 RT complexed with DNA-DNA template-primer, the palm subdomain in the template cleft contacts the template backbone near the proposed site of slippage via the Glu(89) side chain. We investigated the role of Glu(89) in frameshifting by perturbing this interaction. Substitutions with Asp, Gly, Ala, Val, Ser, Thr, Asn, or Lys were created in recombinant HIV RT, and frameshift frequencies of the resulting mutant RTs were measured. All substitutions led to reduced -1 frameshifting by HIV-1 RT (2-40-fold). Interestingly, the suppression of -1 frameshifting frequently coincided with an enhancement of +1 frameshifting (3-47-fold) suggesting that Glu(89) can influence the slippage of both strands. Glu(89) substitutions also led to reduced rates of dNTP misincorporation that paralleled reductions in -1 frameshifting, suggesting a common structural mechanism for both classes of RT error. Our results reveal a major influence of Glu(89) on slippage-mediated errors and dNTP incorporation fidelity. The crystal structure of HIV-1 RT reveals a salt bridge between Glu(89) and Lys(154), which may facilitate -1 frameshifting; this concept is supported by the observed reduction in -1 frameshifting for K154A and K154R mutants.


Subject(s)
HIV Reverse Transcriptase/genetics , Mutation , Base Sequence , DNA/chemistry , DNA Primers/chemistry , DNA, Viral/genetics , Drug Resistance, Viral , Frameshift Mutation , Gene Deletion , Genes, Viral , Glutamic Acid/chemistry , HIV-1/metabolism , Lac Operon , Lysine/chemistry , Models, Genetic , Models, Molecular , Molecular Sequence Data , Oligonucleotides/chemistry , Protein Conformation , RNA-Directed DNA Polymerase , Recombinant Proteins/chemistry , Templates, Genetic , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...