Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Viruses ; 16(5)2024 04 27.
Article in English | MEDLINE | ID: mdl-38793575

ABSTRACT

BACKGROUND: EcoHIV is a chimeric HIV that replicates in mice in CD4+ T cells, macrophages, and microglia (but not in neurons), causing lasting neurocognitive impairment resembling neurocognitive disease in people living with HIV. The present study was designed to develop EcoHIV-susceptible primary mouse brain cultures to investigate the indirect effects of HIV infection on neuronal integrity. RESULTS: We used two EcoHIV clones encoding EGFP and mouse bone marrow-derived macrophages (BMM), mixed mouse brain cells, or enriched mouse glial cells from two wild-type mouse strains to test EcoHIV replication efficiency, the identity of productively infected cells, and neuronal apoptosis and integrity. EcoHIV replicated efficiently in BMM. In mixed brain cell cultures, EcoHIV targeted microglia but did not cause neuronal apoptosis. Instead, the productive infection of the microglia activated them and impaired synaptophysin expression, dendritic density, and axonal structure in the neurons. EcoHIV replication in the microglia and neuronal structural changes during infection were prevented by culture with an antiretroviral. CONCLUSIONS: In murine brain cell cultures, EcoHIV replication in the microglia is largely responsible for the aspects of neuronal dysfunction relevant to cognitive disease in infected mice and people living with HIV. These cultures provide a tool for further study of HIV neuropathogenesis and its control.


Subject(s)
Brain , Microglia , Neurons , Virus Replication , Animals , Mice , Brain/virology , Brain/pathology , Neurons/virology , Neurons/pathology , Microglia/virology , Cells, Cultured , HIV Infections/virology , Macrophages/virology , Disease Models, Animal , Apoptosis , Humans , HIV-1/physiology , Primary Cell Culture , Mice, Inbred C57BL
2.
Virology ; 589: 109917, 2024 01.
Article in English | MEDLINE | ID: mdl-37951088

ABSTRACT

Antiretroviral therapy controls immunodeficiency in people with HIV but many develop mild neurocognitive disorder. Here we investigated HIV brain disease by infecting mice with the chimeric HIV, EcoHIV, and probing changes in brain gene expression during infection and reversal with polyinosinic-polycytidylic acid (poly I:C). EcoHIV-infected C57BL/6 mice were treated with poly I:C and monitored by assay of learning in radial arm water maze, RNAseq of striatum, and QPCR of virus burden and brain transcripts. Poly I:C reversed EcoHIV-associated cognitive impairment and reduced virus burden. Major pathways downregulated by infection involved neuronal function, these transcriptional changes were normalized by poly I:C treatment. Innate immune responses were the major pathways induced in EcoHIV-infected, poly I:C treated mice. Our findings provide a framework to identify brain cell genes dysregulated by HIV infection and identify a set of innate immune response genes that can block systemic infection and its associated dysfunction in the brain.


Subject(s)
HIV Infections , Humans , Animals , Mice , HIV Infections/complications , Mice, Inbred C57BL , Brain , Immunity, Innate , Cognition , Poly I
3.
STAR Protoc ; 4(3): 102368, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37342907

ABSTRACT

EcoHIV is a model of HIV infection that recapitulates aspects of HIV-1 pathology in mice. However, there are limited published protocols to guide EcoHIV virion production. Here, we present a protocol for producing infective EcoHIV virions and essential quality controls. We describe steps for viral purification, titering, and multiple techniques to analyze infection efficacy. This protocol produces high infectivity in C57BL/6 mice which will aid investigators in generating preclinical data.


Subject(s)
HIV Infections , HIV-1 , Animals , Mice , Mice, Inbred C57BL , Virion
4.
Sci Rep ; 13(1): 6577, 2023 04 21.
Article in English | MEDLINE | ID: mdl-37085605

ABSTRACT

HIV enters the brain within days of infection causing neurocognitive impairment (NCI) in up to half of infected people despite suppressive antiretroviral therapy. The virus is believed to enter the brain in infected monocytes through chemotaxis to the major monocyte chemokine, CCL2, but the roles of CCL2 in established NCI are not fully defined. We addressed this question during infection of conventional and CCL2 knockout mice with EcoHIV in which NCI can be verified in behavioral tests. EcoHIV enters mouse brain within 5 days of infection, but NCI develops gradually with established cognitive disease starting 25 days after infection. CCL2 knockout mice infected by intraperitoneal injection of virus failed to develop brain infection and NCI. However, when EcoHIV was directly injected into the brain, CCL2 knockout mice developed NCI. Knockout of CCL2 or its principal receptor, CCR2, slightly reduced macrophage infection in culture. Treatment of mice prior to and during EcoHIV infection with the CCL2 transcriptional inhibitor, bindarit, prevented brain infection and NCI and reduced macrophage infection. In contrast, bindarit treatment of mice 4 weeks after infection affected neither brain virus burden nor NCI. Based on these findings we propose that HIV enters the brain mainly through infected monocytes but that resident brain cells are sufficient to maintain NCI. These findings suggest that NCI therapy must act within the brain.


Subject(s)
AIDS Dementia Complex , Chemokine CCL2 , HIV Infections , Animals , Mice , AIDS Dementia Complex/genetics , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cognition , HIV Infections/complications , HIV Infections/genetics , Indazoles , Mice, Inbred C57BL , Mice, Knockout , Monocytes , Receptors, CCR2/genetics , Disease Models, Animal
5.
Front Immunol ; 13: 1004985, 2022.
Article in English | MEDLINE | ID: mdl-36275760

ABSTRACT

Thirty-eight million people worldwide are living with HIV, PWH, a major public health problem. Antiretroviral therapy (ART) revolutionized HIV treatment and significantly increased the lifespan of PWH. However, approximately 15-50% of PWH develop HIV associated neurocognitive disorders (HIV-NCI), a spectrum of cognitive deficits, that negatively impact quality of life. Many PWH also have opioid use disorder (OUD), and studies in animal models of HIV infection as well as in PWH suggest that OUD can contribute to HIV-NCI. The synthetic opioid agonist, buprenorphine, treats OUD but its effects on HIV-NCI are unclear. We reported that human mature inflammatory monocytes express the opioid receptors MOR and KOR, and that buprenorphine reduces important steps in monocyte transmigration. Monocytes also serve as HIV reservoirs despite effective ART, enter the brain, and contribute to HIV brain disease. Using EcoHIV infected mice, an established model of HIV infection and HIV-NCI, we previously showed that pretreatment of mice prior to EcoHIV infection reduces mouse monocyte entry into the brain and prevents NCI. Here we show that buprenorphine treatment of EcoHIV infected mice with already established chronic NCI completely reverses the disease. Disease reversal was associated with a significant reduction in brain inflammatory monocytes and reversal of dendritic injury in the cortex and hippocampus. These results suggest that HIV-NCI persistence may require a continuing influx of inflammatory monocytes into the brain. Thus, we recommend buprenorphine as a potential therapy for mitigation of HIV brain disease in PWH with or without OUD.


Subject(s)
Brain Diseases , Buprenorphine , HIV Infections , Opioid-Related Disorders , Animals , Humans , Mice , Buprenorphine/pharmacology , Buprenorphine/therapeutic use , HIV Infections/complications , HIV Infections/drug therapy , Analgesics, Opioid/therapeutic use , Quality of Life , Opioid-Related Disorders/complications , Receptors, Opioid
6.
Neurobiol Dis ; 169: 105734, 2022 07.
Article in English | MEDLINE | ID: mdl-35462006

ABSTRACT

People living with HIV (PLH) have significantly higher rates of cognitive impairment (CI) and major depressive disorder (MDD) versus the general population. The enzyme neutral sphingomyelinase 2 (nSMase2) is involved in the biogenesis of ceramide and extracellular vesicles (EVs), both of which are dysregulated in PLH, CI, and MDD. Here we evaluated EcoHIV-infected mice for behavioral abnormalities relevant to depression and cognition deficits, and assessed the behavioral and biochemical effects of nSMase2 inhibition. Mice were infected with EcoHIV and daily treatment with either vehicle or the nSMase2 inhibitor (R)-(1-(3-(3,4-dimethoxyphenyl)-2,6-dimethylimidazo[1,2-b]pyridazin-8-yl)pyrrolidin-3-yl)-carbamate (PDDC) began 3 weeks post-infection. After 2 weeks of treatment, mice were subjected to behavior tests. EcoHIV-infected mice exhibited behavioral abnormalities relevant to MDD and CI that were reversed by PDDC treatment. EcoHIV infection significantly increased cortical brain nSMase2 activity, resulting in trend changes in sphingomyelin and ceramide levels that were normalized by PDDC treatment. EcoHIV-infected mice also exhibited increased levels of brain-derived EVs and altered microRNA cargo, including miR-183-5p, miR-200c-3p, miR-200b-3p, and miR-429-3p, known to be associated with MDD and CI; all were normalized by PDDC. In conclusion, inhibition of nSMase2 represents a possible new therapeutic strategy for the treatment of HIV-associated CI and MDD.


Subject(s)
Depressive Disorder, Major , Extracellular Vesicles , HIV Infections , MicroRNAs , Animals , Ceramides , HIV Infections/complications , HIV Infections/drug therapy , Humans , Mice , MicroRNAs/genetics , MicroRNAs/pharmacology , Sphingomyelin Phosphodiesterase/genetics
8.
J Leukoc Biol ; 109(3): 675-681, 2021 03.
Article in English | MEDLINE | ID: mdl-32578908

ABSTRACT

Approximately 15-40% of people living with HIV develop HIV-associated neurocognitive disorders, HAND, despite successful antiretroviral therapy. There are no therapies to treat these disorders. HIV enters the CNS early after infection, in part by transmigration of infected monocytes. Currently, there is a major opioid epidemic in the United States. Opioid use disorder in the context of HIV infection is important because studies show that opioids exacerbate HIV-mediated neuroinflammation that may contribute to more severe cognitive deficits. Buprenorphine is an opioid derivate commonly prescribed for opiate agonist treatment. We used the EcoHIV mouse model to study the effects of buprenorphine on cognitive impairment and to correlate these with monocyte migration into the CNS. We show that buprenorphine treatment prior to mouse EcoHIV infection prevents the development of cognitive impairment, in part, by decreased accumulation of monocytes in the brain. We propose that buprenorphine has a novel therapeutic benefit of limiting the development of neurocognitive impairment in HIV-infected opioid abusers as well as in nonabusers, in addition to decreasing the use of harmful opioids. Buprenorphine may also be used in combination with HIV prevention strategies such as pre-exposure prophylaxis because of its safety profile.


Subject(s)
AIDS Dementia Complex/prevention & control , Buprenorphine/therapeutic use , HIV Infections/drug therapy , AIDS Dementia Complex/complications , AIDS Dementia Complex/virology , Animals , Antigens, Ly/metabolism , Brain/pathology , Buprenorphine/pharmacology , Chronic Disease , Cognitive Dysfunction/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/virology , Disease Models, Animal , Inflammation/pathology , Male , Mice, Inbred C57BL , Monocytes/drug effects , Phenotype , Viral Load/drug effects
9.
Article in English | MEDLINE | ID: mdl-32699842

ABSTRACT

HIV associated neurocognitive impairment afflicts roughly half of infected individuals on antiretroviral therapy. This disease currently has no treatment. We have previously shown that type I interferon is induced by and partially controls infection and neuropathogenesis in mice infected by chimeric HIV, EcoHIV. Here we investigate the intentional ligation of the pattern recognition receptor Toll-like receptor 3 (TLR3) by polyinosinic-polycytidylic acid (poly I:C) for its ability to prevent or control infection and associated cognitive disease in EcoHIV infected mice. We tested topical, injection, and intranasal application of poly I:C in mice during primary infection through injection or sexual transmission or in established infection. We measured different forms of HIV DNA and RNA in tissues by real-time PCR and the development of HIV-associated cognitive disease by the radial arm water maze behavioral test. Our results indicate that poly I:C blocks primary EcoHIV infection of mice prior to reverse transcription and reduces established EcoHIV infection. Prevention or control of viral replication by poly I:C prevents or reverses HIV associated cognitive disease in mice. These findings indicate that poly I:C or other innate immune agonists may be useful in control of HIV cognitive disease.

10.
FASEB J ; 34(2): 1996-2010, 2020 02.
Article in English | MEDLINE | ID: mdl-31907999

ABSTRACT

Despite the use of antiretroviral therapy for the treatment of HIV-1 infection, cognitive impairments, that is, HIV-1-associated neurocognitive disorders remain prevalent potentially due to persistent viral replication, production of viral proteins, associated brain inflammation or in certain instances, antiretroviral neurotoxicity. Cellular targets in the brain include microglia which in response to infection release inflammatory markers and viral proteins. Evidence suggests that PPARγ agonists exert anti-inflammatory properties in neurological disorders. However, these agonists namely, thiazolidinediones have limited use in the clinic due to reported adverse side effects. INT131 is a novel non-thiazolidinedione compound that belongs to a new class of drugs known as selective PPARγ modulators. INT131 is considered to have a safer profile; however, its neuroprotective role in vivo is not known.The goal of this study was to examine the effect of INT131 in the context of EcoHIV-induced inflammation in vitro, in primary cultures of mouse glial cells and in vivo, in a mouse model of EcoHIV-associated brain inflammation, as well as characterize its pharmacokinetic properties and brain penetration. In primary cultures of glial cells and in the in vivo mouse model, EcoHIV exposure resulted in a significant elevation of inflammatory markers such as TNFα, IL-1ß, CCL3, and C3 which were attenuated with INT131 treatment. Pharmacokinetic analyses revealed that INT131 penetrates into the brain with a brain to blood partition ratio Kp value of 8.5%. Overall, this is the first report to demonstrate that INT131 could be a potential candidate for the treatment of HIV-1-associated brain inflammation.


Subject(s)
Anti-Inflammatory Agents , HIV Infections/drug therapy , HIV-1/metabolism , Neurocognitive Disorders/drug therapy , PPAR gamma/agonists , Quinolines , Sulfonamides , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/genetics , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Mice , Neurocognitive Disorders/genetics , Neurocognitive Disorders/metabolism , Neurocognitive Disorders/pathology , Neuroglia/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Quinolines/pharmacokinetics , Quinolines/pharmacology , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
11.
Sci Rep ; 9(1): 9428, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31263138

ABSTRACT

The widespread use of combination antiretroviral therapy (cART) has resulted in significantly reduced deaths from HIV-1 associated complications and opportunistic infections. However, it is estimated that up to 50% of HIV-1 infected individuals still develop HIV-1 associated neurocognitive disorders (HAND). With no treatment currently available for patients, there is a critical need to identify therapeutic approaches that can treat this disorder. Evidence suggests that targeting Peroxisome Proliferator-Activated Receptor-gamma (PPARγ) can be anti-inflammatory in neurological disorders. Here we show that treatment with PPARγ agonists (rosiglitazone or pioglitazone) in primary cultures of mouse glial cells reversed EcoHIV-induced inflammatory genes (TNFα, IL-1ß, CCL2, CCL3, CXCL10) and indicator of oxidative stress (iNOS). Furthermore, in vivo, mice administered with EcoHIV through intracranial injection resulted in upregulation of inflammatory genes (TNFα, IL-1ß, IFNγ, CCL2, CCL3, CXCL10) and oxidative stress marker (iNOS) in the brain which was reversed through intraperitoneal administration of PPARγ agonists (rosiglitazone or pioglitazone). Finally, we demonstrated that treatment with these compounds in vivo reduced EcoHIV p24 protein burden in the brain. Our results suggest that treatment with PPARγ agonists are anti-inflammatory and antiviral in an in vivo model of EcoHIV infection. These drugs hold promise as potential candidates for HAND treatment in the future.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , HIV-1/drug effects , PPAR gamma/agonists , Pioglitazone/pharmacology , Rosiglitazone/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Antiviral Agents/therapeutic use , Brain/drug effects , Brain/metabolism , Cells, Cultured , Disease Models, Animal , HIV Core Protein p24/metabolism , HIV Infections/drug therapy , HIV Infections/pathology , HIV-1/genetics , HIV-1/physiology , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , PPAR gamma/metabolism , Pioglitazone/therapeutic use , Rosiglitazone/therapeutic use , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
mBio ; 10(4)2019 07 02.
Article in English | MEDLINE | ID: mdl-31266862

ABSTRACT

HIV causes neurodegeneration and dementia in AIDS patients, but its function in milder cognitive impairments in virologically suppressed patients on antiretroviral therapy is unknown. Such patients are immunocompetent, have low peripheral and brain HIV burdens, and show minimal brain neuropathology. Using the model of HIV-related memory impairment in EcoHIV-infected conventional mice, we investigated the neurobiological and cognitive consequences of efficient EcoHIV expression in the mouse brain after intracerebral infection. HIV integrated and persisted in an expressed state in brain tissue, was detectable in brain monocytic cells, and caused neuroinflammatory responses and lasting spatial, working, and associative memory impairment. Systemic antiretroviral treatment prevented direct brain infection and memory dysfunction indicating the requirement for HIV expression in the brain for disease. Similarly inoculated murine leukemia virus used as a control replicated in mouse brain but not in monocytic cells and was cognitively benign, linking the disease to HIV-specific functions. Memory impairment correlated in real time with hippocampal dysfunction shown by defective long-term potentiation in hippocampal slices ex vivo and with diffuse synaptodendritic injury in the hippocampus reflected in significant reduction in microtubule-associated protein 2 and synapsin II staining. In contrast, there was no evidence of overt neuronal loss in this region as determined by neuron-specific nuclear protein quantification, TUNEL assay, and histological observations. Our results reveal a novel capacity of HIV to induce neuronal dysfunction and memory impairment independent of neurotoxicity, distinct from the neurotoxicity of HIV infection in dementia.IMPORTANCE HIV neuropathogenesis has been attributed in large measure to neurotoxicity of viral proteins and inflammatory factors produced by infected monocytic cells in the brain. We show here that HIV expression in mouse brain causes lasting memory impairment by a mechanism involving injury to hippocampal synaptodendritic arbors and neuronal function but not overt neuronal loss in the region. Our results mirror the observation of minimal neurodegeneration in cognitively impaired HIV patients on antiretroviral therapy and demonstrate that HIV is nonneurotoxic in certain brain abnormalities that it causes. If neurons comprising the cognition-related networks survive HIV insult, at least for some time, there is a window of opportunity for disease treatment.


Subject(s)
AIDS Dementia Complex/physiopathology , Cognitive Dysfunction/physiopathology , Hippocampus/pathology , Hippocampus/virology , Memory Disorders/complications , Memory Disorders/physiopathology , Animals , Behavior, Animal , Disease Models, Animal , Male , Mice , Viral Load
13.
J Neuroimmune Pharmacol ; 14(3): 391-400, 2019 09.
Article in English | MEDLINE | ID: mdl-31209775

ABSTRACT

HIV-associated neurocognitive disorders (HAND) have been linked to dysregulation of glutamate metabolism in the central nervous system (CNS) culminating in elevated extracellular glutamate and disrupted glutamatergic neurotransmission. Increased glutamate synthesis via upregulation of glutaminase (GLS) activity in brain immune cells has been identified as one potential source of excess glutamate in HAND. However, direct evidence for this hypothesis in an animal model is lacking, and the viability of GLS as a drug target has not been explored. In this brief report, we demonstrate that GLS inhibition with the glutamine analogue 6-diazo-5-oxo-L-norleucine (DON) can reverse cognitive impairment in the EcoHIV-infected mouse model of HAND. However, due to peripheral toxicity DON is not amenable to clinical use in a chronic disease such as HAND. We thus tested JHU083, a novel, brain penetrant DON prodrug predicted to exhibit improved tolerability. Systemic administration of JHU083 reversed cognitive impairment in EcoHIV-infected mice similarly to DON, and simultaneously normalized EcoHIV-induced increases in cerebrospinal fluid (CSF) glutamate and GLS activity in microglia-enriched brain CD11b + cells without observed toxicity. These studies support the mechanistic involvement of elevated microglial GLS activity in HAND pathogenesis, and identify JHU083 as a potential treatment option. Graphical Abstract Please provide Graphical Abstract caption.Glutamine Antagonist JHU083 Normalizes Aberrant Glutamate Production and Cognitive Deficits in the EcoHIV Murine Model of HIV-Associated Neurocognitive Disorders .


Subject(s)
AIDS Dementia Complex , Azo Compounds/therapeutic use , Caproates/therapeutic use , Cognition Disorders/drug therapy , Glutamates/biosynthesis , Glutamine/antagonists & inhibitors , Prodrugs/therapeutic use , Animals , Azo Compounds/pharmacokinetics , CD11b Antigen/analysis , Caproates/pharmacokinetics , Cognition Disorders/cerebrospinal fluid , Cognition Disorders/etiology , Cognition Disorders/virology , Conditioning, Classical/drug effects , Fear , Glutamates/cerebrospinal fluid , HIV-1/genetics , HIV-1/pathogenicity , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , Norleucine/analogs & derivatives , Norleucine/therapeutic use , Prodrugs/pharmacokinetics , Reassortant Viruses/genetics , Reassortant Viruses/pathogenicity , Spatial Learning/drug effects
14.
AIDS ; 33(6): 973-984, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30946151

ABSTRACT

OBJECTIVE: Almost half of HIV-positive people on antiretroviral therapy have demonstrable mild neurocognitive impairment (HIV-NCI), even when virologically suppressed. Intranasal insulin therapy improves cognition in Alzheimer's disease and diabetes. Here we tested intranasal insulin therapy in a model of HIV-NCI in EcoHIV-infected conventional mice. DESIGN AND METHODS: Insulin pharmacokinetics following intranasal administration to mice was determined by ELISA. Mice were inoculated with EcoHIV to cause NCI; 23 days or 3 months after infection they were treated daily for 9 days with intranasal insulin (2.4 IU/mouse) and examined for NCI in behavioral tests and HIV burdens by quantitative PCR. Some animals were tested for hippocampal neuronal integrity by immunostaining and expression of neuronal function-related genes by real time-quantitative PCR. The effect of insulin treatment discontinuation on cognition and neuropathology was also examined. RESULTS: Intranasal insulin administration to mice resulted in µIU/ml levels of insulin in cerebrospinal fluid with a half-life of about 2 h, resembling pharmacokinetic parameters of patients receiving 40 IU. Intranasal insulin treatment starting 23 days or 3 months after infection completely reversed NCI in mice. Murine NCI correlated with reductions in hippocampal dendritic arbors and downregulation of neuronal function genes; intranasal insulin reversed these changes coincident with restoration of cognitive acuity, but they returned within 24 h of treatment cessation. Intranasal insulin treatment reduced brain HIV DNA when started 23 but not 90 days after infection. CONCLUSION: Our preclinical studies support the use of intranasal insulin administration for treatment of HIV-NCI and suggest that some dendritic injury in this condition is reversible.


Subject(s)
AIDS Dementia Complex/drug therapy , Cognitive Dysfunction/drug therapy , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Administration, Intranasal , Animals , Behavior, Animal , Disease Models, Animal , Hippocampus/pathology , Hypoglycemic Agents/pharmacokinetics , Immunohistochemistry , Insulin/pharmacokinetics , Mice, Inbred C57BL , Treatment Outcome , Viral Load
15.
PLoS Pathog ; 14(6): e1007061, 2018 06.
Article in English | MEDLINE | ID: mdl-29879225

ABSTRACT

Suppression of HIV replication by antiretroviral therapy (ART) or host immunity can prevent AIDS but not other HIV-associated conditions including neurocognitive impairment (HIV-NCI). Pathogenesis in HIV-suppressed individuals has been attributed to reservoirs of latent-inducible virus in resting CD4+ T cells. Macrophages are persistently infected with HIV but their role as HIV reservoirs in vivo has not been fully explored. Here we show that infection of conventional mice with chimeric HIV, EcoHIV, reproduces physiological conditions for development of disease in people on ART including immunocompetence, stable suppression of HIV replication, persistence of integrated, replication-competent HIV in T cells and macrophages, and manifestation of learning and memory deficits in behavioral tests, termed here murine HIV-NCI. EcoHIV established latent reservoirs in CD4+ T lymphocytes in chronically-infected mice but could be induced by epigenetic modulators ex vivo and in mice. In contrast, macrophages expressed EcoHIV constitutively in mice for up to 16 months; murine leukemia virus (MLV), the donor of gp80 envelope in EcoHIV, did not infect macrophages. Both EcoHIV and MLV were found in brain tissue of infected mice but only EcoHIV induced NCI. Murine HIV-NCI was prevented by antiretroviral prophylaxis but once established neither persistent EcoHIV infection in mice nor NCI could be reversed by long-acting antiretroviral therapy. EcoHIV-infected, athymic mice were more permissive to virus replication in macrophages than were wild-type mice, suffered cognitive dysfunction, as well as increased numbers of monocytes and macrophages infiltrating the brain. Our results suggest an important role of HIV expressing macrophages in HIV neuropathogenesis in hosts with suppressed HIV replication.


Subject(s)
CD4-Positive T-Lymphocytes/virology , Disease Reservoirs , HIV Infections/complications , HIV/physiology , Macrophages, Peritoneal/virology , Neurocognitive Disorders/virology , Adoptive Transfer , Aged , Animals , Anti-Retroviral Agents/therapeutic use , Brain/virology , Female , HIV/genetics , HIV/immunology , HIV/pathogenicity , HIV Infections/drug therapy , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Middle Aged , Plasmids , Spleen/cytology , Spleen/immunology
16.
J Neurovirol ; 24(4): 398-410, 2018 08.
Article in English | MEDLINE | ID: mdl-29594983

ABSTRACT

The widespread use of antiretroviral therapy for treatment of human immunodeficiency virus (HIV) infections has dramatically improved the quality and duration of life for HIV-positive individuals. Despite this success, HIV persists for the life of an infected person in tissue reservoirs including the nervous system. Thus, whether HIV exacerbates age-related brain disorders such as Parkinson's disease (PD) is of concern. In support of this idea, HIV infection can be associated with motor and gait abnormalities that parallel late-stage manifestations of PD including dopaminergic neuronal loss. With these findings in hand, we investigated whether viral infection could affect nigrostriatal degeneration or exacerbate chemically induced nigral degeneration. We now demonstrate an additive effect of EcoHIV on dopaminergic neuronal loss and neuroinflammation induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intoxication. HIV-1-infected humanized mice failed to recapitulate these EcoHIV results suggesting species-specific neural signaling. The results demonstrate a previously undefined EcoHIV-associated neurodegenerative response that may be used to model pathobiological aspects of PD.


Subject(s)
HIV Infections/complications , MPTP Poisoning/complications , Substantia Nigra/pathology , Substantia Nigra/virology , Animals , HIV Infections/pathology , HIV-1 , Humans , MPTP Poisoning/pathology , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Nerve Degeneration/virology
17.
ACS Chem Neurosci ; 9(4): 809-816, 2018 04 18.
Article in English | MEDLINE | ID: mdl-29257872

ABSTRACT

Insulin delivery to the brain has emerged as an important therapeutic target for cognitive disorders associated with abnormal brain energy metabolism. Although insulin is transported across the blood-brain barrier, peripheral routes of administration are problematic due to systemic effects of insulin on blood glucose. Intranasal (IN) administration is being investigated as an alternative route. We conducted a head-to-head comparison of subcutaneous (SC) and IN insulin, assessing plasma and brain pharmacokinetics and blood glucose levels in the mouse. SC insulin (2.4 IU) achieved therapeutically relevant concentrations in the brain (AUCbrain = 2537 h·µIU/mL) but dramatically increased plasma insulin (AUCplasma = 520 351 h·*µIU/mL), resulting in severe hypoglycemia and in some cases death. IN administration of the same dose resulted in similar insulin levels in the brain (AUCbrain = 3442 h·µIU/mL) but substantially lower plasma concentrations (AUCplasma = 354 h·µIU/mL), amounting to a ∼ 2000-fold increase in the AUCbrain:plasma ratio relative to SC. IN dosing also had no significant effect on blood glucose. When administered daily for 9 days, IN insulin increased brain glucose and energy metabolite concentrations (e.g., adenosine triphosphate and phosphocreatine) without causing overt toxicity, suggesting that IN insulin may be a safe therapeutic option for cognitively impaired patients.


Subject(s)
Blood Glucose/metabolism , Brain/metabolism , Insulin/blood , Insulin/pharmacokinetics , Administration, Intranasal , Animals , Blood-Brain Barrier/metabolism , Cognition Disorders/metabolism , Energy Metabolism/physiology , Insulin/administration & dosage , Insulin/cerebrospinal fluid , Male , Mice
18.
J Med Chem ; 60(16): 7186-7198, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28759224

ABSTRACT

Aberrant excitatory neurotransmission associated with overproduction of glutamate has been implicated in the development of HIV-associated neurocognitive disorders (HAND). The glutamine antagonist 6-diazo-5-oxo-l-norleucine (DON, 14) attenuates glutamate synthesis in HIV-infected microglia/macrophages, offering therapeutic potential for HAND. We show that 14 prevents manifestation of spatial memory deficits in chimeric EcoHIV-infected mice, a model of HAND. 14 is not clinically available, however, because its development was hampered by peripheral toxicities. We describe the synthesis of several substituted N-(pivaloyloxy)alkoxy-carbonyl prodrugs of 14 designed to circulate inert in plasma and be taken up and biotransformed to 14 in the brain. The lead prodrug, isopropyl 6-diazo-5-oxo-2-(((phenyl(pivaloyloxy)methoxy)carbonyl)amino)hexanoate (13d), was stable in swine and human plasma but liberated 14 in swine brain homogenate. When dosed systemically in swine, 13d provided a 15-fold enhanced CSF-to-plasma ratio and a 9-fold enhanced brain-to-plasma ratio relative to 14, opening a possible clinical path for the treatment of HAND.


Subject(s)
Aminocaproates/pharmacology , Azo Compounds/pharmacology , Diazooxonorleucine/pharmacology , Neurocognitive Disorders/drug therapy , Nootropic Agents/pharmacology , Prodrugs/pharmacology , Aminocaproates/administration & dosage , Aminocaproates/chemical synthesis , Animals , Azo Compounds/administration & dosage , Azo Compounds/chemical synthesis , Blood/metabolism , Brain/metabolism , Diazooxonorleucine/administration & dosage , Drug Stability , Female , Glutamic Acid/metabolism , Glutaminase/antagonists & inhibitors , HIV Infections/complications , Humans , Male , Mice, Inbred C57BL , Neurocognitive Disorders/etiology , Nootropic Agents/administration & dosage , Nootropic Agents/chemical synthesis , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Swine , Viral Load/drug effects
19.
Am J Physiol Lung Cell Mol Physiol ; 312(4): L500-L509, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28104604

ABSTRACT

Cigarette smoke usage is prevalent in human immunodeficiency virus (HIV)-positive patients, and, despite highly active antiretroviral therapy, these individuals develop an accelerated form of chronic obstructive pulmonary disease (COPD). Studies investigating the mechanisms of COPD development in HIV have been limited by the lack of suitable mouse models. Here we describe a model of HIV-induced COPD in wild-type mice using EcoHIV, a chimeric HIV capable of establishing chronic infection in immunocompetent mice. A/J mice were infected with EcoHIV and subjected to whole body cigarette smoke exposure. EcoHIV was detected in alveolar macrophages of mice. Compared with uninfected mice, concomitant EcoHIV infection significantly reduced forced expiratory flow 50%/forced vital capacity and enhanced distal airspace enlargement following cigarette smoke exposure. Lung IL-6, granulocyte-macrophage colony-stimulating factor, neutrophil elastase, cathepsin G, and matrix metalloproteinase-9 expression was significantly enhanced in smoke-exposed EcoHIV-infected mice. These changes coincided with enhanced IκBα, ERK1/2, p38, and STAT3 phosphorylation and lung cell apoptosis. Thus, the EcoHIV smoke exposure mouse model reproduces several of the pathophysiological features of HIV-related COPD in humans, indicating that this murine model can be used to determine key parameters of HIV-related COPD and to test future therapies for this disorder.


Subject(s)
HIV Infections/complications , Pulmonary Disease, Chronic Obstructive/complications , Animals , Apoptosis , Disease Models, Animal , Humans , Lung/enzymology , Lung/immunology , Lung/pathology , Lung/virology , Macrophages, Alveolar/pathology , Macrophages, Alveolar/virology , Male , Mice , Neutrophils/metabolism , Peptide Hydrolases/metabolism , Pneumonia/pathology , Smoking/adverse effects
20.
J Neuroinflammation ; 14(1): 23, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28122624

ABSTRACT

BACKGROUND: Abnormal activation of the complement system contributes to some central nervous system diseases but the role of complement in HIV-associated neurocognitive disorder (HAND) is unclear. METHODS: We used real-time PCR and immunohistochemistry to detect complement expression in postmortem brain tissue from HAND patients and controls. To further investigate the basis for viral induction of gene expression in the brain, we studied the effect of HIV on C3 expression by astrocytes, innate immune effector cells, and targets of HIV. Human fetal astrocytes (HFA) were infected with HIV in culture and cellular pathways and factors involved in signaling to C3 expression were elucidated using pharmacological pathway inhibitors, antisense RNA, promoter mutational analysis, and fluorescence microscopy. RESULTS: We found significantly increased expression of complement components including C3 in brain tissues from patients with HAND and C3 was identified by immunocytochemistry in astrocytes and neurons. Exposure of HFA to HIV in culture-induced C3 promoter activity, mRNA expression, and protein production. Use of pharmacological inhibitors indicated that induction of C3 expression by HIV requires NF-κB and protein kinase signaling. The relevance of NF-κB regulation to C3 induction was confirmed through detection of NF-κB translocation into nuclei and inhibition through overexpression of the physiological NF-κB inhibitor, I-κBα. C3 promoter mutation analysis revealed that the NF-κB and SP binding sites are dispensable for the induction by HIV, while the proximal IL-1ß/IL-6 responsive element is essential. HIV-treated HFA secreted IL-6, exogenous IL-6 activated the C3 promoter, and anti-IL-6 antibodies blocked HIV activation of the C3 promoter. The activation of IL-6 transcription by HIV was dependent upon an NF-κB element within the IL-6 promoter. CONCLUSIONS: These results suggest that HIV activates C3 expression in primary astrocytes indirectly, through NF-κB-dependent induction of IL-6, which in turn activates the C3 promoter. HIV induction of C3 and IL-6 in astrocytes may contribute to HIV-mediated inflammation in the brain and cognitive dysfunction.


Subject(s)
Astrocytes/metabolism , Complement C3/metabolism , HIV Infections/pathology , Interleukin-6/metabolism , NF-kappa B/metabolism , Adult , Astrocytes/virology , Caffeic Acids/pharmacology , Complement C3/genetics , Enzyme Activation/drug effects , Enzyme Activators/pharmacology , Enzyme Inhibitors/pharmacology , Female , Fetus , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation, Viral/physiology , Glial Fibrillary Acidic Protein/metabolism , Humans , Interleukin-6/genetics , Male , Middle Aged , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Signal Transduction/drug effects , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL