Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Neurovirol ; 26(6): 838-845, 2020 12.
Article in English | MEDLINE | ID: mdl-32901392

ABSTRACT

Despite combined antiretroviral therapy (cART), HIV infection in the CNS persists with reported increases in activation of macrophages (MΦ), microglia, and surrounding astrocytes/neurons, conferring HIV-induced inflammation. Chronic inflammation results in HIV-associated neurocognitive disorders (HAND) with reported occurrence of up to half of individuals with HIV infection. The existing HAND mouse model used by laboratories including ours, and the effect of novel agents on its pathology present with labor-intensive and time-consuming limitations since brain sections and immunohistochemistry assays have to be performed and analyzed. A novel flow cytometry-based system to objectively quantify phenotypic effects of HIV using a SCID mouse HAND model was developed which demonstrated that the HIV-infected mice had significant increases in astrogliosis, loss of neuronal dendritic marker, activation of murine microglia, and human macrophage explants compared to uninfected control mice. HIV p24 could also be quantified in the brains of the infected mice. Correlation of these impairments with HIV-induced brain inflammation and previous behavioral abnormalities studies in mice suggests that this model can be used as a fast and relevant throughput methodology to quantify preclinical testing of novel treatments for HAND.


Subject(s)
Brain/metabolism , Cognitive Dysfunction/genetics , Disease Models, Animal , Gliosis/genetics , HIV Infections/genetics , HIV-1/genetics , Animals , Astrocytes/metabolism , Astrocytes/virology , Biomarkers/metabolism , Brain/virology , Cognitive Dysfunction/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/virology , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/metabolism , Gliosis/virology , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Inflammation , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, SCID , Microglia/metabolism , Microglia/virology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/virology , Phenotype
2.
Sci Rep ; 10(1): 11696, 2020 07 16.
Article in English | MEDLINE | ID: mdl-32678115

ABSTRACT

Pulmonary Arterial Hypertension (PAH) is overrepresented in People Living with Human Immunodeficiency Virus (PLWH). HIV protein gp120 plays a key role in the pathogenesis of HIV-PAH. Genetic changes in HIV gp120 determine viral interactions with chemokine receptors; specifically, HIV-X4 viruses interact with CXCR4 while HIV-R5 interact with CCR5 co-receptors. Herein, we leveraged banked samples from patients enrolled in the NIH Lung HIV studies and used bioinformatic analyses to investigate whether signature sequences in HIV-gp120 that predict tropism also predict PAH. Further biological assays were conducted in pulmonary endothelial cells in vitro and in HIV-transgenic rats. We found that significantly more persons living with HIV-PAH harbor HIV-X4 variants. Multiple HIV models showed that recombinant gp120-X4 as well as infectious HIV-X4 remarkably increase arachidonate 5-lipoxygenase (ALOX5) expression. ALOX5 is essential for the production of leukotrienes; we confirmed that leukotriene levels are increased in bronchoalveolar lavage fluid of HIV-infected patients. This is the first report associating HIV-gp120 genotype to a pulmonary disease phenotype, as we uncovered X4 viruses as potential agents in the pathophysiology of HIV-PAH. Altogether, our results allude to the supplementation of antiretroviral therapy with ALOX5 antagonists to rescue patients with HIV-X4 variants from fatal PAH.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , HIV Infections/complications , HIV-1/genetics , Lung/metabolism , Pulmonary Arterial Hypertension/complications , Viral Tropism/genetics , Adult , Animals , Anti-HIV Agents/therapeutic use , Cells, Cultured , Cohort Studies , Disease Models, Animal , Endothelial Cells/metabolism , Female , Genotype , HIV Envelope Protein gp120/genetics , HIV Infections/drug therapy , HIV Infections/virology , HIV-1/metabolism , Humans , Male , Middle Aged , Phenotype , Pulmonary Arterial Hypertension/virology , Pulmonary Artery/cytology , Rats , Rats, Inbred F344 , Rats, Transgenic , Receptors, CXCR4/metabolism
3.
J Investig Med ; 68(2): 321-330, 2020 02.
Article in English | MEDLINE | ID: mdl-31582425

ABSTRACT

Although Multiple Sclerosis is the most common central nervous system (CNS) inflammatory demyelinating disorder, other CNS inflammatory disorders should be included as diagnostic considerations. Neuromyelitis Optica Spectrum Disorder (NMOSD) and myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease are less common but share some clinical characteristics, such as optic neuritis and myelitis, which can make a specific diagnosis challenging. However, these disorders have distinctive and generally different clinical phenotypes, prognosis and management. It is imperative to distinguish each from one another, especially since the treatments (not discussed in this review) can be different. The advent of reliable testing for anti-aquaporin-4 for NMOSD and anti-MOG antibodies has helped significantly; however, diagnosis can remain challenging, especially in sero-negative cases. Clinical indicators are important to guide diagnostic work-up. Careful review of the history, neurological exam, imaging, and/or spinal fluid results are essential to making an accurate diagnosis. In this review, we will examine the clinical presentation, diagnosis, and natural history of these inflammatory CNS disorders.


Subject(s)
Autoantibodies/blood , Multiple Sclerosis/blood , Multiple Sclerosis/diagnostic imaging , Myelin-Oligodendrocyte Glycoprotein/blood , Neuromyelitis Optica/blood , Neuromyelitis Optica/diagnostic imaging , Demyelinating Autoimmune Diseases, CNS/blood , Demyelinating Autoimmune Diseases, CNS/diagnostic imaging , Humans , Neuroimaging/methods
4.
J Neuroinflammation ; 16(1): 182, 2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31561750

ABSTRACT

BACKGROUND: Since HIV-associated neurocognitive disorders (HANDs) occur in up to half of HIV-positive individuals, even with combined antiretroviral therapy (cART), adjunctive therapies are needed. Chronic CNS inflammation contributes to HAND and HIV encephalitis (HIVE). Baricitinib is a JAK 1/2 inhibitor approved in the USA, EU, and Japan for rheumatoid arthritis, demonstrating potent inhibition of IL-6, D-dimer, CRP, TNF-α, IFN-α/ß, and other pro-inflammatory cytokines. METHODS: Our modified murine HAND model was used to evaluate the ability of baricitinib to cross the blood-brain barrier (BBB) and modulate monocyte/macrophage-driven HAND. Severity of HAND was measured by assessing cognitive performance of low- and high-dose baricitinib treated versus untreated HAND mice. The severity of brain neuroinflammation was evaluated in these mouse groups after flow cytometric analyses. We also assessed the ability of baricitinib to block events in myeloid and lymphoid cells in vitro that may undergird the persistence of HIV in the central nervous system (CNS) in primary human macrophages (Mϕ) and lymphocytes including HIV replication, HIV-induced activation, reservoir expansion, and reservoir maintenance. RESULTS: In vivo, both doses of 10 and 50 mg/kg qd baricitinib crossed the BBB and reversed behavioral abnormalities conferred by HIV infection. Moreover, baricitinib significantly reduced HIV-induced neuroinflammation marked by glial activation: activated microglia (MHCII+/CD45+) and astrogliosis (GFAP). Baricitinib also significantly reduced the percentage of p24+ human macrophages in mouse brains (p < 0.05 versus HAND mice; t test). In vitro, baricitinib significantly reduced markers of persistence, reservoir size, and reseeding in Mϕ. CONCLUSION: These results show that blocking the JAK/STAT pathway reverses cognitive deficits and curtails inflammatory markers in HAND in mice. Our group recently reported safety and tolerability of ruxolitinib in HIV-infected individuals (Marconi et al., Safety, tolerability and immunologic activity of ruxolitinib added to suppressive ART, 2019), underscoring potential safety and utility of JAK inhibitors for additional human trials. The data reported herein coupled with our recent human trial with JAK inhibitors provide compelling preclinical data and impetus for considering a trial of baricitinib in HAND individuals treated with cART to reverse cognitive deficits and key events driving viral persistence.


Subject(s)
AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Azetidines/pharmacology , Sulfonamides/pharmacology , Virus Activation/drug effects , Virus Replication/drug effects , Animals , Disease Models, Animal , Humans , Mice , Mice, SCID , Purines , Pyrazoles , Virus Latency/drug effects
5.
Sci Rep ; 9(1): 8733, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217522

ABSTRACT

HIV-associated neurocognitive disorder (HAND) is a common condition in both developed and developing nations, but its cause is largely unknown. Previous research has inconsistently linked Alzheimer's disease (AD), viral burden, and inflammation to the onset of HAND in HIV-infected individuals. Here we simultaneously measured cerebrospinal fluid (CSF) levels of established amyloid and tau biomarkers for AD, viral copy numbers, and six key cytokines in 41 HIV-infected individuals off combination anti-retroviral therapy (14 with HAND) who underwent detailed clinical and neuropsychological characterization, and compared their CSF patterns with those from young healthy subjects, older healthy subjects with normal cognition, and older people with AD. HAND was associated with the lowest CSF levels of phosphorylated tau (p-Tau181) after accounting for age and race. We also found very high CSF levels of the pro-inflammatory interferon gamma-induced protein 10 (IP-10/CXCL10) in HIV regardless of cognition, but elevated CSF interleukin 8 (IL-8/CXCL8) only in HIV-NC but not HAND. Eleven HIV-infected subjects underwent repeat CSF collection six months later and showed strongly correlated longitudinal changes in p-Tau181 and IL-8 levels (R = 0.841). These data suggest reduced IL-8 relative to IP-10 and reduced p-Tau181 to characterize HAND.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , HIV Infections/cerebrospinal fluid , Interleukin-8/cerebrospinal fluid , tau Proteins/cerebrospinal fluid , Adult , Aged , Alzheimer Disease/drug therapy , Anti-Retroviral Agents/administration & dosage , Biomarkers/cerebrospinal fluid , Chemokine CXCL10/cerebrospinal fluid , Female , HIV Infections/drug therapy , Humans , Male , Middle Aged , Phosphorylation
6.
Front Immunol ; 10: 480, 2019.
Article in English | MEDLINE | ID: mdl-30930904

ABSTRACT

Inflammation is a common process involved in aging, multiple sclerosis (MS), and age-related neurodegenerative disorders such as Alzheimer's disease (AD) and Parkinson's disease (PD), but there is limited evidence for the effects of aging on inflammation in the central nervous system. We collected cerebrospinal fluid (CSF) from 105 healthy control subjects representing a wide age range (23-86), and analyzed levels of cytokines associated innate immunity (TNF-α) and different T-helper subtypes: interferon-gamma induced protein 10 (IP-10) for Th1, interleukin-10 (IL-10) for Th2, and interleukin 8 (IL-8/CXCL8) for Th17. We show that CSF levels of TNF-α, IP-10, and IL-8 all increased linearly with age, but levels of IL-10 demonstrated a U-shaped relationship with age. We further found greater age-related increases in TNF-α, IL-10, and IL-8 relative to increases in IP-10 levels, consistent with a shift from Th1 to other inflammatory phenotypes. Finally, when we analyzed the same four cytokines in people with neurological disorders, we found that MS and AD, but not PD or dementia with Lewy bodies, further accentuated the age-related shift from Th1- to non-Th1-related cytokines. We propose that CSF cytokine levels represent powerful surrogates of brain inflammation and aging, and some, but not all, neurological disorders accelerate the shift away from Th1 phenotypes.


Subject(s)
Aging/cerebrospinal fluid , Alzheimer Disease/cerebrospinal fluid , Cytokines/cerebrospinal fluid , Multiple Sclerosis/cerebrospinal fluid , Parkinson Disease/cerebrospinal fluid , Adult , Aged , Aged, 80 and over , Aging/immunology , Alzheimer Disease/immunology , Cytokines/immunology , Female , Humans , Male , Middle Aged , Multiple Sclerosis/immunology , Parkinson Disease/immunology
7.
AIDS ; 32(11): 1403-1411, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29683852

ABSTRACT

OBJECTIVE: Increased brain interferon-alpha (IFNα) is associated with neurodegenerative disorders, including HIV-associated neurocognitive disorders (HAND). HAND occurs in approximately 50% of individuals with HIV despite combined antiretroviral therapy (cART). Therefore, adjunctive therapies must be developed that prevent progression of mild forms of HAND to HIV-associated dementia. Increased IFNα in the CNS has been associated with HAND in patients and in a HAND mouse model. DESIGN AND METHODS: B18R binds IFNα and ameliorates HAND mouse brain histopathology (HIV encephalitis). The HAND model was used to determine if B18R with cART is superior to cART. Behavioral testing [Object recognition Test (ORT)] was used to show that B18R can reverse behavioral deficits. Rat neuronal cultures were used to investigate mechanisms of IFNα neurotoxicity. RESULTS: Mouse brain immunohistochemistry and densitometry suggests that B18R with a common cART regimen improve histopathological markers better than cART alone. B18R reverses ORT behavioral abnormalities in HAND mice. IFNα-treated rat neurons show decreases in PSD-95, suggesting that dendritic spine architecture is disrupted. Decreases in Arf1, a GTP-binding protein, and AMPA receptors on the surface of rat neurons exposed to IFNα suggest the mechanism of IFNα neurotoxicity may relate to decreased Arf1 resulting in destabilization of dendritic spines, decreased PSD-95 expression, and internalization of AMPA receptors. CONCLUSION: B18R reversal of HAND in the mouse model is further evidence that the treatment of IFNα in individuals with HAND could be a viable adjunctive treatment. Investigating pathways of IFNα neurotoxicity may lead to more specific treatments.


Subject(s)
AIDS Dementia Complex/drug therapy , Anti-Retroviral Agents/administration & dosage , Immunologic Factors/administration & dosage , Interferon-alpha/toxicity , Neurons/pathology , Viral Proteins/administration & dosage , AIDS Dementia Complex/pathology , Animals , Immunohistochemistry , Interferon-alpha/antagonists & inhibitors , Male , Mice , Rats, Sprague-Dawley , Treatment Outcome
8.
Clin Infect Dis ; 67(1): 137-140, 2018 06 18.
Article in English | MEDLINE | ID: mdl-29385540

ABSTRACT

New tools are needed to understand human immunodeficiency virus central nervous system involvement. Testing 15 cerebrospinal fluid (CSF) samples for p24 antigen, using a high-sensitivity assay, we found a strong correlation trend between CSF p24 concentration and worse neuropsychological performance.


Subject(s)
HIV Core Protein p24/cerebrospinal fluid , HIV Infections/cerebrospinal fluid , HIV Infections/complications , HIV-1/isolation & purification , Neurocognitive Disorders/etiology , Adult , Enzyme-Linked Immunosorbent Assay , Female , HIV Infections/diagnosis , Humans , Male , Middle Aged , Neurocognitive Disorders/diagnosis , Neuropsychological Tests , RNA, Viral/blood , RNA, Viral/cerebrospinal fluid
9.
J Neurovirol ; 24(2): 180-184, 2018 04.
Article in English | MEDLINE | ID: mdl-28895064

ABSTRACT

HIV-associated neurocognitive disorders (HAND) remain highly prevalent despite combined antiretroviral therapy (cART). Although the most common forms of HAND are mild and identified through neuropsychological testing, there is evidence that with aging these mild forms become more prevalent and may advance to the most severe form of HAND, HIV-associated dementia. Therefore, novel therapies must be developed that can be used adjunctively with cART to prevent deterioration or restore normal cognitive function. In order to develop innovative treatments, animal models are used for preclinical testing. Ideally, a HAND animal model should portray similar mild cognitive deficits that are found in humans. A mouse model of HAND is discussed, which demonstrates mild behavioral deficits and has been used to investigate cART and novel treatments for HAND. This model also shows correlations between abnormal mouse behavior due to HIV in the brain and pathological parameters such as gliosis and neuronal abnormalities. A recent advancement utilizes the object recognition test to monitor mouse behavior before and after treatment. It is postulated that this model is well suited for preclinical testing of novel therapies and provides correlations of mild cognitive impairment with pathological markers that can give further insight into the pathophysiology of HAND.


Subject(s)
AIDS Dementia Complex/physiopathology , Cognitive Dysfunction/physiopathology , Disease Models, Animal , HIV-1/pathogenicity , Macrophages/virology , Pattern Recognition, Visual , AIDS Dementia Complex/virology , Animals , Astrocytes/pathology , Astrocytes/virology , Behavior, Animal , Brain/physiopathology , Brain/virology , Cognitive Dysfunction/virology , HIV-1/physiology , Humans , Injections, Intraventricular , Macrophages/transplantation , Mice , Neuroglia/pathology , Neuroglia/virology , Neurons/pathology , Neurons/virology , Transplantation, Heterologous
10.
Neurochem Res ; 42(11): 3220-3232, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28770436

ABSTRACT

Although antiretroviral (ARV) therapy has reduced the incidence of severe dementia associated with HIV infection, there has been a rise in milder neurocognitive complaints. Data from HIV patients taking ARVs have shown measurable neurocognitive improvements during drug cessation, suggesting a neurotoxic role of the therapy itself. Mechanisms underlying potential ARV neurotoxicity have not been thoroughly investigated, however pathologic oxidative stress and mitochondrial dysfunction have been suspected. Using DIV 16 primary rat cortical neuron culture, we tested eight ARVs from the three most commonly prescribed ARV classes: nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs/NtRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and protease inhibitors (PIs) for effects on neuron viability and morphology after 24 h of drug exposure. Of the tested NRTIs, only stavudine at nearly 100 times the target plasma concentration affected neuron viability with no appreciable change in morphology. Dideoxyinosine induced dendritic simplification at 100 times target plasma concentrations, but did not adversely affect viability. The sole NtRTI, tenofovir, induced dendritic simplification at approximately 3-4 times target plasma concentration, but did not affect viability. Of the tested PIs, only amprenavir decreased neuron viability at nearly 100 times the target plasma concentration. The non-nucleoside reverse transcriptase inhibitor, efavirenz, consistently reduced viability (at 50 µM) and induced dendritic simplification (at 20 µM) nearest the target plasma concentration. Probing mitochondrial energetics of DIV16 cortical neurons after exposure to 20 µM efavirenz showed rapid diminution of mitochondrial-dependent oxygen consumption. Further, 20 µM efavirenz decreased excitability in ex vivo slice culture whereas 2 µM had no effect.


Subject(s)
Anti-HIV Agents/toxicity , Anti-Retroviral Agents/toxicity , Benzoxazines/toxicity , Reverse Transcriptase Inhibitors/toxicity , Alkynes , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cyclopropanes , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Neurons/pathology , Rats , Rats, Sprague-Dawley
11.
FASEB J ; 31(8): 3608-3621, 2017 08.
Article in English | MEDLINE | ID: mdl-28442545

ABSTRACT

Pseudomonas aeruginosa is a significant contributor to recalcitrant multidrug-resistant infections, especially in immunocompromised and hospitalized patients. The pathogenic profile of P. aeruginosa is related to its ability to secrete a variety of virulence factors and to promote biofilm formation. Quorum sensing (QS) is a mechanism wherein P. aeruginosa secretes small diffusible molecules, specifically acyl homo serine lactones, such as N-(3-oxo-dodecanoyl)-l-homoserine lactone (3O-C12-HSL), that promote biofilm formation and virulence via interbacterial communication. Strategies that strengthen the host's ability to inhibit bacterial virulence would enhance host defenses and improve the treatment of resistant infections. We have recently shown that peroxisome proliferator-activated receptor γ (PPARγ) agonists are potent immunostimulators that play a pivotal role in host response to virulent P. aeruginosa Here, we show that QS genes in P. aeruginosa (strain PAO1) and 3O-C12-HSL attenuate PPARγ expression in bronchial epithelial cells. PAO1 and 3O-C12-HSL induce barrier derangements in bronchial epithelial cells by lowering the expression of junctional proteins, such as zonula occludens-1, occludin, and claudin-4. Expression of these proteins was restored in cells that were treated with pioglitazone, a PPARγ agonist, before infection with PAO1 and 3O-C12-HSL. Barrier function and bacterial permeation studies that have been performed in primary human epithelial cells showed that PPARγ agonists are able to restore barrier integrity and function that are disrupted by PAO1 and 3O-C12-HSL. Mechanistically, we show that these effects are dependent on the induction of paraoxonase-2, a QS hydrolyzing enzyme, that mitigates the effects of QS molecules. Importantly, our data show that pioglitazone, a PPARγ agonist, significantly inhibits biofilm formation on epithelial cells by a mechanism that is mediated via paraoxonase-2. These findings elucidate a novel role for PPARγ in host defense against P. aeruginosa Strategies that activate PPARγ can provide a therapeutic complement for treatment of resistant P. aeruginosa infections.-Bedi, B., Maurice, N. M., Ciavatta, V. T., Lynn, K. S., Yuan, Z., Molina, S. A., Joo, M., Tyor, W. R., Goldberg, J. B., Koval, M., Hart, C. M., Sadikot, R. T. Peroxisome proliferator-activated receptor-γ agonists attenuate biofilm formation by Pseudomonas aeruginosa.


Subject(s)
Bacterial Proteins/pharmacology , Biofilms/growth & development , PPAR gamma/agonists , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/physiology , Aryldialkylphosphatase/genetics , Aryldialkylphosphatase/metabolism , Cell Line , Epithelial Cells/microbiology , Gene Expression Regulation/physiology , Humans , Mutation , Pseudomonas aeruginosa/genetics , Quorum Sensing
12.
J Investig Med ; 65(5): 883-891, 2017 06.
Article in English | MEDLINE | ID: mdl-28130412

ABSTRACT

Multiple sclerosis (MS) is an autoimmune, demyelinating disease of the central nervous system (CNS). It predominantly affects young women and is one of the most common causes of disability in young adults. MS is characterized by formation of white matter lesions in the CNS as a result of inflammation, demyelination, and axonal loss. Treatment has been a focus of neurological research for over 60 years. A number of disease-modifying therapies (DMTs) have become available making MS a treatable disease. These compounds target the inflammatory response in MS. They work by decreasing the chances of relapse, decreasing the chances of new lesion formation seen on MRI of the CNS and slowing the accumulation of disability. The first drugs for MS to be available were interferon-ß and glatiramer acetate. These work by modulating the inflammatory response via different mechanisms that are briefly discussed. Newer agents have since become available and have significantly changed the dynamics of MS treatment. These include fingolimod, dimethyl fumarate and teriflunomide, which are oral agents. Other second-line and third-line Food and Drug Administration (FDA) approved medications include natalizumab and alemtuzumab. Natalizumab is considered one of the most potent treatments for relapse prevention. However, the high risk of progressive multifocal leukoencephalopathy (PML), which is caused by JC virus infection in the brain, tempers the more widespread use of this agent; nevertheless, JC virus antibody tests have helped to stratify the risk of PML. Alemtuzumab, which also has a considerable side effect profile, is likewise highly efficacious. Ocrelizumab, a monoclonal antibody to CD20 on B cells, is a highly effective agent for MS that is likely to be approved soon by the FDA. MS is a major contributor to healthcare costs and it is critical that healthcare providers be aware of the availability and benefits of DMTs. It is imperative that prompt and adequate treatment be established on diagnosis. Changes in therapy should be considered when there is evidence of disease activity as well as accumulation of disability or safety or tolerability concerns.


Subject(s)
Immunologic Factors/therapeutic use , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Administration, Oral , Alemtuzumab/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Crotonates/therapeutic use , Daclizumab , Dimethyl Fumarate/therapeutic use , Drug Approval , Fingolimod Hydrochloride/therapeutic use , Glatiramer Acetate/therapeutic use , Humans , Hydroxybutyrates , Immunoglobulin G/therapeutic use , Infusions, Intravenous , JC Virus , Leukoencephalopathy, Progressive Multifocal/complications , Leukoencephalopathy, Progressive Multifocal/virology , Multiple Sclerosis , Natalizumab/therapeutic use , Nitriles , Polyomavirus Infections/complications , Recurrence , Toluidines/therapeutic use , Treatment Outcome , Tumor Virus Infections/complications , United States , United States Food and Drug Administration
13.
J Neurovirol ; 23(1): 106-112, 2017 02.
Article in English | MEDLINE | ID: mdl-27400930

ABSTRACT

HIV-associated neurocognitive disorders (HANDs) continue to be common and are associated with increased morbidity and mortality. However, the underlying mechanisms in the combination antiretroviral therapy (cART) era are not fully understood. Interferon alpha (IFNα) is an antiviral cytokine found to be elevated in the cerebrospinal fluid (CSF) of individuals with advanced HIV-associated dementia in the pre-cART era. In this cross-sectional study, we investigated the association between IFNα and neurocognitive performance in ambulatory HIV-infected individuals with milder impairment. An eight-test neuropsychological battery representing six cognitive domains was administered. Individual scores were adjusted for demographic characteristics, and a composite neuropsychological score (NPT-8) was calculated. IFNα and CSF neurofilament light chain (NFL) levels were measured using enzyme-linked immunosorbent assay (ELISA). There were 15 chronically infected participants with a history of significant immunocompromise (median nadir CD4+ of 49 cells/µl). Most participants were neurocognitively impaired (mean global deficit score of 0.86). CSF IFNα negatively correlated with three individual tests (Trailmaking A, Trailmaking B, and Stroop Color-Word) as well as the composite NPT-8 score (r = -0.67, p = 0.006). These negative correlations persisted in multivariable analyses adjusting for chronic hepatitis B and C. Additionally, CSF IFNα correlated strongly with CSF NFL, a marker of neuronal damage (rho = 0.748, p = 0.0013). These results extend findings from individuals with severe HIV-associated dementia in the pre-cART era and suggest that IFNα may continue to play a role in HAND pathogenesis during the cART era. Further investigation into the role of IFNα is indicated.


Subject(s)
AIDS Dementia Complex/diagnosis , AIDS Dementia Complex/immunology , Antiviral Agents/therapeutic use , Interferon-alpha/cerebrospinal fluid , Neurofilament Proteins/cerebrospinal fluid , AIDS Dementia Complex/cerebrospinal fluid , AIDS Dementia Complex/drug therapy , Adult , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cognition/physiology , Cross-Sectional Studies , Female , Gene Expression , Humans , Immunocompromised Host , Interferon-alpha/genetics , Interferon-alpha/immunology , Male , Middle Aged , Neurofilament Proteins/genetics , Neurofilament Proteins/immunology , Neuropsychological Tests , Outpatients
14.
J Neurovirol ; 22(6): 816-822, 2016 12.
Article in English | MEDLINE | ID: mdl-27245594

ABSTRACT

Given the high prevalence of HIV-associated neurocognitive disorders (HAND), we examined the performance of a novel computerized cognitive assessment device (NCAD) for the evaluation of neurocognitive impairment in the setting of HIV. In addition to a standard 8-test neuropsychological battery, each participant underwent testing with the NCAD, which requires approximately 20 min and has been shown to accurately measure neurocognition in elderly individuals. The NCAD yields seven subtest scores in addition to an overall predictive score that is calculated based on subtest results. Thirty-nine HIV-infected participants were included in this study; the majority of which (71.8 %) had undetectable plasma HIV RNA levels and a history of significant immunocompromise (median nadir CD4+ count 34 cells/µl). The mean composite neuropsychological score (NPT-8) was 46.07, and mean global deficit score (GDS) was 0.59. NCAD total subtest accuracy correlated significantly with NPT-8 (Pearson correlation r = 0.59, p < 0.0001) as well as GDS (Spearman's rho = -0.36, p = 0.02). NCAD predictive score also correlated significantly with NPT-8 (Spearman's rho = -0.5601, p = 0.0016) and GDS (Spearman's rho = 0.45, p = 0.0144). When using the most recent nosology of HAND criteria for neurocognitive impairment, the area under the curve (AUC) for NCAD total subtest accuracy was 0.7562 (p = 0.012), while the AUC for the HIV dementia scale was 0.508 (p = 0.930). While not as comprehensive as a full neuropsychological battery, the NCAD shows promise as a rapid screening tool for HIV-infected individuals, and additional research of this device is indicated.


Subject(s)
Brain-Computer Interfaces , Cognitive Dysfunction/diagnosis , HIV Infections/diagnosis , Neuropsychological Tests , Adult , Area Under Curve , CD4 Lymphocyte Count , Cognitive Dysfunction/complications , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/psychology , Equipment Design , Female , Georgia , HIV Infections/complications , HIV Infections/physiopathology , HIV Infections/psychology , Humans , Male , Middle Aged , RNA, Viral/blood , Sensitivity and Specificity , Severity of Illness Index
16.
Neurobiol Dis ; 92(Pt B): 137-43, 2016 08.
Article in English | MEDLINE | ID: mdl-26851503

ABSTRACT

A hallmark of persistent HIV-1 infection in the central nervous system is increased activation of mononuclear phagocytes and surrounding astrogliosis, conferring persistent HIV-induced inflammation. This inflammation is believed to result in neuronal dysfunction and the clinical manifestations of HIV-associated neurocognitive disorders (HAND). The Jak/STAT pathway is activated in macrophages/myeloid cells upon HIV-1 infection, modulating many pro-inflammatory pathways that result in HAND, thereby representing an attractive cellular target. Thus, the impact of ruxolitinib, a Janus Kinase (Jak) 1/2 inhibitor that is FDA approved for myelofibrosis and polycythemia vera, was assessed for its potential to inhibit HIV-1 replication in macrophages and HIV-induced activation in monocytes/macrophages in culture. In addition, a murine model of HIV encephalitis (HIVE) was used to assess the impact of ruxolitinib on histopathological features of HIVE, brain viral load, as well as its ability to penetrate the blood-brain-barrier (BBB). Ruxolitinib was found to inhibit HIV-1 replication in macrophages, HIV-induced activation of monocytes (CD14/CD16) and macrophages (HLA-DR, CCR5, and CD163) without apparent toxicity. In vivo, systemically administered ruxolitinib was detected in the brain during HIVE in SCID mice and markedly inhibited astrogliosis. Together, these data indicate that ruxolitinib reduces HIV-induced activation and infiltration of monocytes/macrophages in vitro, reduces the replication of HIV in vitro, penetrates the BBB when systemically administered in mice and reduces astrogliosis in the brains of mice with HIVE. These data suggest that ruxolitinib will be useful as a novel therapeutic to treat humans with HAND.


Subject(s)
Antiviral Agents/pharmacology , Encephalitis, Viral/drug therapy , HIV Infections/drug therapy , Janus Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Virus Replication/drug effects , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Disease Models, Animal , Encephalitis, Viral/metabolism , Encephalitis, Viral/pathology , Gliosis/drug therapy , Gliosis/metabolism , Gliosis/pathology , Gliosis/virology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/drug effects , HIV-1/physiology , Humans , Macrophages/drug effects , Macrophages/virology , Male , Mice , Monocytes/drug effects , Monocytes/virology , Nitriles , Pyrimidines , Tenofovir/pharmacology
17.
Neurochem Res ; 40(11): 2220-9, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26342829

ABSTRACT

Thrombin's role in the nervous system is not well understood. Under conditions of blood-brain barrier compromise (e.g., neurosurgery or stroke), thrombin can result in neuroapoptosis and the formation of glial scars. Despite this, preconditioning with thrombin has been found to be neuroprotective in models of cerebral ischemia and intracerebral hemorrhage. We investigated the effects of physiologically relevant concentrations of thrombin on cortical neurons using two culture-based assays. We examined thrombin's effect on neurites by quantitative analysis of fluorescently labeled neurons. To characterize thrombin's effects on neuron survival, we spectrophotometrically measured changes in enzymatic activity. Using receptor agonists and thrombin inhibitors, we separately examined the role of thrombin and its receptor in neuroprotection. We found that low concentrations of thrombin (1 nM) enhances neurite growth and branching, neuron viability, and protects against excitotoxic damage. In contrast, higher concentrations of thrombin (100 nM) are potentially detrimental to neuronal health as evidenced by inhibition of neurite growth. Lower concentrations of thrombin resulted in equivalent neuroprotection as the antifibrinolytic, aprotinin, and the direct thrombin inhibitor, argatroban. Interestingly, exogenous application of the species-specific thrombin inhibitor, antithrombin III, was detrimental to neuronal health; suggesting that some endogenous thrombin is necessary for optimal neuron health in our culture system. Activation of the thrombin receptor, protease-activated receptor-1 (PAR-1), via micromolar concentrations of the thrombin receptor agonist peptide, TRAP, did not adversely affect neuronal viability. An optimal concentration of thrombin exists to enhance neuronal health. Neurotoxic effects of thrombin do not involve activation of PAR receptors and thus separate pharmacologic manipulation of thrombin's receptor in the setting of direct thrombin inhibitors could be a potential neuroprotective strategy.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Thrombin/pharmacology , Animals , Antithrombin III/pharmacology , Aprotinin/pharmacology , Arginine/analogs & derivatives , Cell Survival/drug effects , Cells, Cultured , Neurites/drug effects , Peptide Fragments/pharmacology , Pipecolic Acids/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, PAR-1/agonists , Receptors, Thrombin/drug effects , Sulfonamides , Thrombin/antagonists & inhibitors
18.
J Interferon Cytokine Res ; 35(4): 317-24, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25517826

ABSTRACT

Elevated levels of interferon-alpha (IFNα) in the central nervous system (CNS) are linked to cognitive dysfunction in patients with inflammatory CNS diseases such as HIV-associated neurocognitive disorders (HAND). Increased CNS IFNα has also been found to be associated with cognitive dysfunction in a HAND mouse model. Here, we corroborate previous studies showing a dose-dependent decrease in dendritic branching and length caused by IFNα treatment and extend those studies. Because both direct and indirect mechanisms of IFNα-induced neurotoxicity are likely involved, the cell signaling pathway involving the IFNα receptor (IFNAR) was initially evaluated. Rat neuronal cultures exposed to IFNα demonstrate increased phosphorylation of STAT1 and increased interferon stimulating gene 15 (ISG15) expression, indicators of IFNAR engagement. However, specific blocking antibodies to the IFNAR were found to only partially protect neurons from IFNα-induced neurotoxicity. Additionally, inhibiting the GluN2A subunit of N-methyl-D-asparate receptor (NMDAR) was also found to be partially protective against IFNα-induced neurotoxicity compared with the GluN2B subunit. Neurotoxicity is evident in neurons extracted from IFNAR KO mice treated with IFNα as well, further indicating that IFNAR signaling is not required for IFNα neurotoxicity. The neurotoxic actions of IFNα are mediated through both the IFNAR as well as the GluN2A subunit of the NMDAR to reduce dendritic arborization in neurons. Complete protection from IFNα-induced neurotoxicity was demonstrated when both pathways were blocked. Blocking these pathways could lead to potential therapies for cognitive dysfunction during neuroinflammation and specifically lead to better treatments for HAND.


Subject(s)
Interferon-alpha/pharmacology , Neurons/drug effects , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Blotting, Western , Cells, Cultured , Cytokines/genetics , Gene Expression Regulation/drug effects , Interferon-alpha/metabolism , Mice , Phosphorylation/drug effects , Polymerase Chain Reaction , Rats , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects
19.
J Interferon Cytokine Res ; 34(7): 510-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24564363

ABSTRACT

Interferon-alpha (IFN-α) has been identified as a neurotoxin that plays a prominent role in human immunodeficiency virus (HIV)-associated neurocognitive disorders and HIV encephalitis (HIVE) pathology. IFN-α is associated with cognitive dysfunction in other inflammatory diseases where IFN-α is upregulated. Trials of monoclonal anti-IFN-α antibodies have been generally disappointing possibly due to high specificity to limited IFN-α subtypes and low affinity. We investigated a novel IFN-α inhibitor, B18R, in an HIVE/severe combined immunodeficiency (SCID) mouse model. Immunostaining for B18R in systemically treated HIVE/SCID mice suggested the ability of B18R to cross the blood-brain barrier (BBB). Real-time PCR indicated that B18R treatment resulted in a decrease in gene expression associated with IFN-α signaling in the brain. Mice treated with B18R were found to have decreased mouse mononuclear phagocytes and significant retention of neuronal arborization compared to untreated HIVE/SCID mice. Increased mononuclear phagocytes and decreased neuronal arborization are key features of HIVE. These results suggest that B18R crosses the BBB, blocks IFN-α signaling, and it prevents key features of HIVE pathology. These data suggest that the high affinity and broad IFN-α subtype specificity of B18R make it a viable alternative to monoclonal antibodies for the inhibition of IFN-α in the immune-suppressed environment.


Subject(s)
Encephalitis , HIV Infections , Interferon-alpha/genetics , Interferon-alpha/metabolism , Recombinant Proteins/therapeutic use , Viral Proteins/therapeutic use , Animals , Disease Models, Animal , Encephalitis/drug therapy , Encephalitis/etiology , Gene Expression Regulation/drug effects , HIV Infections/complications , HIV Infections/physiopathology , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Vaccinia virus/genetics , Viral Proteins/genetics , Viral Proteins/pharmacology
20.
J Neurovirol ; 20(1): 9-17, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24415129

ABSTRACT

The role of brain HIV load in the pathogenesis of HIV-associated neurocognitive disorders (HAND) is unclear. To try and determine if the amount of HIV drives the severity of pathology, a severe combined immunodeficient (SCID) mouse model of HIV encephalitis (HIVE) was utilized to determine the effectiveness of a systemically administered combined antiretroviral (cART) regimen. SCID mice were inoculated intracerebrally with HIV-infected or uninfected (control) human macrophages and treated subcutaneously with cART or saline for 10 days. Immunohistochemistry was then used to examine gliosis and neuronal damage. Drug levels were measured in brain and plasma using high-performance liquid chromatography. Peak plasma and brain levels of atazanavir, tenofovir, and emtricitabine were determined to be 1 h post-injection of cART therapy. cART significantly reduced neuropathological features of HIVE, including astrogliosis and the presence of mononuclear phagocytes, and ameliorated reduced MAP2 (neuronal integrity) staining. However, cART did not eradicate HIV from the brain. Using this animal model of HIVE, these data indicate effective penetration of cART reduces brain viral loads and HIV pathology, possibly by eliminating the production of HIV proteins, virus infected cells, or both. Importantly, these data suggest that viral load directly affects the extent of pathology seen in the brain, particularly neuronal damage, which implies that more effective suppression of HIV in the CNS could reduce currently highly prevalent forms of HAND. However, these data also strongly suggest that cART will not eliminate HIV from the brain and that adjunctive therapies must be developed.


Subject(s)
AIDS Dementia Complex/virology , Anti-Retroviral Agents/pharmacology , Brain/virology , Viral Load/drug effects , AIDS Dementia Complex/pathology , Animals , Brain/pathology , Chromatography, High Pressure Liquid , Disease Models, Animal , HIV-1 , Humans , Immunohistochemistry , Male , Mice , Mice, SCID
SELECTION OF CITATIONS
SEARCH DETAIL
...