Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Neurovirol ; 18(4): 291-302, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22653528

ABSTRACT

Highly active antiretroviral therapy (HAART) has increased life expectancy among HIV-infected individuals, and by 2015, at least half of all HIV-infected individuals will be over 50 years of age. Neurodegenerative processes associated with aging may be facilitated by HIV-1 infection, resulting in premature brain aging. This review will highlight brain abnormalities in HIV patients in the setting of aging, focusing on recent neuroimaging studies of the structural, physiological, functional and neurochemical changes. Magnetic resonance imaging (MRI) and magnetic resonance spectroscopy studies performed during the pre-HAART era or on antiretroviral-naive subjects suggest an accelerated aging process, while those on HAART-treated subjects suggest premature brain atrophy. Diffusion tensor imaging studies yielded conflicting findings on the relationship between HIV and age in neuroasymptomatic individuals. Functional MRI studies found evidence of premature or accelerated aging processes in the brains of HIV subjects. Lastly, many age-related illnesses such as diabetes, stroke, and depression, as well as comorbid substance abuse, may further exacerbate the aging process in the HIV-infected brain, leading to premature or accelerated age-related brain changes. Given the different pathologic or physiologic changes in the brain assessed by the different neuroimaging techniques, using a multimodal approach in longitudinal follow-up studies is recommended for future studies.


Subject(s)
Aging , Brain/pathology , Cognition Disorders/pathology , HIV Infections/pathology , Anti-Retroviral Agents/administration & dosage , Anti-Retroviral Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Brain/diagnostic imaging , Cognition Disorders/complications , Cognition Disorders/diagnostic imaging , Cognition Disorders/drug therapy , HIV Infections/complications , HIV Infections/diagnostic imaging , HIV Infections/drug therapy , Humans , Life Expectancy , Magnetic Resonance Imaging , Neuroimaging , Positron-Emission Tomography
2.
Cell Immunol ; 271(2): 239-55, 2011.
Article in English | MEDLINE | ID: mdl-21788015

ABSTRACT

We used pulsed stable isotope labeling of amino acids in cell culture (pSILAC) to assess protein dynamics during monocyte-macrophage differentiation. pSILAC allows metabolic labeling of newly synthesized proteins. Such de novo protein production was evaluated from 3 to 7 days in culture. Proteins were identified by liquid chromatography-tandem mass spectrometry then quantified by MaxQuant. Protein-protein linkages were then assessed by Ingenuity Pathway Analysis. Proteins identified were linked to cell homeostasis, free radical scavenging, molecular protein transport, carbohydrate metabolism, small molecule chemistry, and cell morphology. The data demonstrates specific biologic events that are linked to monocyte transformation in a defined biologic system.


Subject(s)
Macrophages/chemistry , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , Proteome/metabolism , Cell Differentiation/immunology , Cell Differentiation/physiology , Cells, Cultured , Free Radical Scavengers/metabolism , Hemoglobins/biosynthesis , Humans , Macrophages/immunology , Models, Biological , Monocytes/immunology , NF-kappa B/metabolism , Phagocytosis , Protein Interaction Maps , Tandem Mass Spectrometry
3.
J Proteome Res ; 10(6): 2852-62, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21500866

ABSTRACT

Dynamic interactions between human immunodeficiency virus-1 (HIV-1) and the macrophage govern the tempo of viral dissemination and replication in its human host. HIV-1 affects macrophage phenotype, and the macrophage, in turn, can modulate the viral life cycle. While these processes are linked to host-cell function and survival, the precise intracellular pathways involved are incompletely understood. To elucidate such dynamic virus-cell events, we employed pulsed stable isotope labeling of amino acids in cell culture. Alterations in de novo protein synthesis of HIV-1 infected human monocyte-derived macrophages (MDM) were examined after 3, 5, and 7 days of viral infection. Synthesis rates of cellular metabolic, regulatory, and DNA packaging activities were decreased, whereas, those affecting antigen presentation (major histocompatibility complex I and II) and interferon-induced antiviral activities were increased. Interestingly, enrichment of proteins linked to chromatin assembly or disassembly, DNA packaging, and nucleosome assembly were identified that paralleled virus-induced cytopathology and replication. We conclude that HIV-1 regulates a range of host MDM proteins that affect its survival and abilities to contain infection.


Subject(s)
Amino Acids/metabolism , HIV-1/physiology , Macrophages/virology , Monocytes/cytology , Cell Culture Techniques , Cells, Cultured , Gene Expression Regulation , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV-1/genetics , Humans , Interferons/metabolism , Isotope Labeling , Macrophages/metabolism , Proteome/genetics , Proteome/metabolism , Signal Transduction , Time Factors
4.
Neurobiol Dis ; 37(3): 542-8, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20044002

ABSTRACT

Human immunodeficiency virus type 1 (HIV-1)-associated neurocognitive disorders (HAND) can affect up to 50% of infected people during the disease course. While antiretroviral therapies have substantively increased the quality of life and reduced HIV-1-associated dementia, less severe minor cognitive and motor deficits continue. Trafficking of HIV-1 into the central nervous system (CNS), peripheral immune activation, dysregulated glial immunity, and diminished homeostatic responses are the disease-linked pathobiologic events. Monocyte-macrophage passage into the CNS remains an underlying force for disease severity. Monocyte phenotypes may change at an early stage of cell maturation and immune activation of hematopoietic stem cells. Activated monocytes are pulled into the brain in response to chemokines made as a result of glial inflammatory processes, which in turn, cause secondary functional deficits in neurons. Current therapeutic approaches are focused on adjunctive and brain-penetrating antiretroviral therapies. These may attenuate virus-associated neuroinflammatory activities thereby decreasing the severity and frequency of HAND.


Subject(s)
AIDS Dementia Complex/immunology , AIDS Dementia Complex/physiopathology , Encephalitis/immunology , Encephalitis/physiopathology , HIV-1/immunology , Neuroimmunomodulation/physiology , AIDS Dementia Complex/drug therapy , Anti-Retroviral Agents/pharmacology , Chemokines/metabolism , Chemotaxis, Leukocyte/immunology , Encephalitis/drug therapy , Humans , Monocytes/immunology , Neuroglia/immunology
5.
Neuron ; 64(1): 133-45, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19840555

ABSTRACT

The use of antiretroviral therapy has reduced mortality and increased the quality of life of HIV-1-infected people, particularly in more developed countries where access to treatment is more widespread. However, morbidities continue, which include HIV-1-associated neurocognitive disorders (HAND). Subtle cognitive abnormalities and low-level viral replication underlie disease. The balance between robust antiviral adaptive immunity, neuronal homeostatic mechanisms, and neuroprotective factors on one hand and toxicities afforded by dysregulated immune activities on the other govern disease. New insights into the pathobiological processes for neuroimmune-linked disease and ways to modulate such activities for therapeutic gain are discussed. Better understanding of the complexities of immune regulation during HAND can improve diagnosis and disease outcomes but is also relevant for the pathogenesis of a broad range of neurodegenerative disorders.


Subject(s)
HIV Infections/immunology , HIV Infections/physiopathology , Neuroimmunomodulation/physiology , AIDS Dementia Complex/immunology , AIDS Dementia Complex/physiopathology , AIDS Dementia Complex/therapy , Animals , Brain/immunology , Brain/physiopathology , Brain/virology , HIV Infections/therapy , Humans , Models, Neurological , Neuroglia/immunology , Neuroglia/physiology , Neuroglia/virology , Neurons/immunology , Neurons/physiology , Neurons/virology
6.
J Neuroimmune Pharmacol ; 3(3): 173-86, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18587649

ABSTRACT

The human immunodeficiency virus (HIV) invades the central nervous system early after viral exposure but causes progressive cognitive, behavior, and motor impairments years later with the onset of immune deficiency. Although in the brain, HIV preferentially replicates productively in cells of mononuclear phagocyte (MP; blood borne macrophage and microglia), astrocytes also can be infected, at low and variable frequency, particularly in patients with encephalitis. Among their many functions, astrocytes network with microglia to provide the first line of defense against microbial infection; however, very little is known about astrocytes' consequences on MP. Here, we addressed this question using co-culture systems of HIV-infected mouse astrocytes and microglia. Pseudotyped vesicular stomatis virus/HIV was used to circumvent the absence of viral receptors and ensure cell genotypic uniformity for studies of intercellular communication. The study demonstrated that infected astrocytes show modest changes in protein elements compared to uninfected cells. In contrast, infected astrocytes induce robust changes in the proteome of HIV-1-infected microglia. Accelerated cell death and redox proteins, among others, were produced in abundance. The observations confirmed the potential of astrocytes to influence the neuropathogenesis of HIV-1 infection by specifically altering the neurotoxic potential of infected microglia and regulating viral maturation.


Subject(s)
Astrocytes/pathology , Astrocytes/virology , Gene Expression Regulation, Viral/physiology , Microglia/pathology , Microglia/virology , Proteome/biosynthesis , AIDS Dementia Complex/metabolism , AIDS Dementia Complex/pathology , AIDS Dementia Complex/virology , Animals , Astrocytes/chemistry , Cell Communication/physiology , Cells, Cultured , Coculture Techniques , Female , HIV-1/pathogenicity , HIV-1/physiology , Humans , Mice , Mice, Inbred C57BL , Microglia/chemistry , Pregnancy , Proteome/genetics
7.
PLoS One ; 3(6): e2507, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18575609

ABSTRACT

BACKGROUND: HIV-1-infected and immune competent brain mononuclear phagocytes (MP; macrophages and microglia) secrete cellular and viral toxins that affect neuronal damage during advanced disease. In contrast, astrocytes can affect disease by modulating the nervous system's microenvironment. Interestingly, little is known how astrocytes communicate with MP to influence disease. METHODS AND FINDINGS: MP-astrocyte crosstalk was investigated by a proteomic platform analysis using vesicular stomatitis virus pseudotyped HIV infected murine microglia. The microglial-astrocyte dialogue was significant and affected microglial cytoskeleton by modulation of cell death and migratory pathways. These were mediated, in part, through F-actin polymerization and filament formation. Astrocyte secretions attenuated HIV-1 infected microglia neurotoxicity and viral growth linked to the regulation of reactive oxygen species. CONCLUSIONS: These observations provide unique insights into glial crosstalk during disease by supporting astrocyte-mediated regulation of microglial function and its influence on the onset and progression of neuroAIDS. The results open new insights into previously undisclosed pathogenic mechanisms and open the potential for biomarker discovery and therapeutics that may influence the course of HIV-1-mediated neurodegeneration.


Subject(s)
Astrocytes/cytology , HIV-1/physiology , Microglia/cytology , Proteomics , Animals , Astrocytes/virology , Cytoskeleton , Female , Mice , Mice, Inbred C57BL , Microglia/virology , Pregnancy , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...