Skip to main content
Erschienen in: Journal of NeuroVirology 1/2013

Open Access 01.02.2013

Age-dependent molecular alterations in the autophagy pathway in HIVE patients and in a gp120 tg mouse model: reversal with beclin-1 gene transfer

verfasst von: Jerel Fields, Wilmar Dumaop, Edward Rockenstein, Michael Mante, Brian Spencer, Igor Grant, Ron Ellis, Scott Letendre, Christina Patrick, Anthony Adame, Eliezer Masliah

Erschienen in: Journal of NeuroVirology | Ausgabe 1/2013

Abstract

Aged (>50 years old) human immunodeficiency virus (HIV) patients are the fastest-growing segment of the HIV-infected population in the USA and despite antiretroviral therapy, HIV-associated neurocognitive disorder (HAND) prevalence has increased or remained the same among this group. Autophagy is an intracellular clearance pathway for aggregated proteins and aged organelles; dysregulation of autophagy is implicated in the pathogenesis of Parkinson’s disease, Alzheimer’s disease, and HAND. Here, we hypothesized that dysregulated autophagy may contribute to aging-related neuropathology in HIV-infected individuals. To explore this possibility, we surveyed autophagy marker levels in postmortem brain samples from a cohort of well-characterized <50 years old (young) and >50 years old (aged) HIV+ and HIV encephalitis (HIVE) patients. Detailed clinical and neuropathological data showed the young and aged HIVE patients had higher viral load, increased neuroinflammation and elevated neurodegeneration; however, aged HIVE postmortem brain tissues showed the most severe neurodegenerative pathology. Interestingly, young HIVE patients displayed an increase in beclin-1, cathepsin-D and light chain (LC)3, but these autophagy markers were reduced in aged HIVE cases compared to age-matched HIV+ donors. Similar alterations in autophagy markers were observed in aged gp120 transgenic (tg) mice; beclin-1 and LC3 were decreased in aged gp120 tg mice while mTor levels were increased. Lentivirus-mediated beclin-1 gene transfer, that is known to activate autophagy pathways, increased beclin-1, LC3, and microtubule-associated protein 2 expression while reducing glial fibrillary acidic protein and Iba1 expression in aged gp120 tg mice. These data indicate differential alterations in the autophagy pathway in young versus aged HIVE patients and that autophagy reactivation may ameliorate the neurodegenerative phenotype in these patients.

Introduction

Currently, over 30 million people live with human immunodeficiency virus (HIV) worldwide. In the USA, the aging population represents one of the fastest-growing groups with HIV (Scott et al. 2011). The Center for Disease Control (CDC) estimates that by the year 2015, half of all Americans living with HIV will be over the age of 50 (Smith 2005). In the central nervous system (CNS), microglial cells have been identified as a primary reservoir for HIV infection (Gendelman et al. 1997; Gonzalez-Scarano and Martin-Garcia 2005; Haas et al. 2000; Wiley et al. 1996) with productive infection also detected in astrocytes (Carroll-Anzinger and Al-Harthi 2006). Modern treatments with highly active antiretroviral therapy regimens result in HIV suppression and immune recovery, however the prevalence of HIV-associated neurocognitive disorders (HAND) and neurodegeneration (Budka et al. 1987; Cherner et al. 2007; Gendelman et al. 1997; Heaton et al. 2010, 2011; Wiley and Achim 1994) has remained the same or increased (Joska et al. 2010) in particular among people over the age of 50.
The mechanisms of neurodegeneration in aged individuals with HAND are not completely understood; however, HIV activates apoptotic pathways (Kaul et al. 2001), dysregulates calcium homeostasis (Lipton 1994; Nath et al. 2000, 2002) and promotes oxidative stress (Norman et al. 2008). Moreover, recent studies have shown that HIV proteins might interfere with clearance pathways such as autophagy (Alirezaei et al. 2008a, b; Zhou et al. 2011), a pathway necessary for protein quality control and elimination of defective older intracellular organelles (Cuervo 2004). Autophagy is a complex process that involves nucleation, initiation, elongation, and termination proteins. Initially, a phagophore forms and develops into the autophagosome, a double-membrane sac that delivers cytoplasmic material to the lysosomal compartment for degradation (Codogno et al. 2012). During the aging process, deficits in autophagy have been described in Alzheimer’s disease (AD; Nixon et al. 2005; Pickford et al. 2008), Parkinson’s disease (PD; Crews et al. 2010; Cuervo et al. 2004), and other aging-related disorders (Cuervo 2004). Specifically, the autophagy nucleation protein beclin-1 and closure protein light chain (LC)3 have been implicated in human disease (Crews et al. 2010; Gozuacik and Kimchi 2004; Jaeger and Wyss-Coray 2010). Similarly, neurodegeneration has been linked to defects in autophagy in patients with HIV (Alirezaei et al. 2008a, b; Zhou et al. 2011).
We have recently shown that in the CNS of young HIV human cases and in young transgenic (tg) mice expressing HIV-gp120 protein (gp120 tg; Toggas et al. 1994), abnormal functioning of the autophagy pathway (Zhou et al. 2011) is associated with progressive accumulation of amyloid-beta (Aβ; Achim et al. 2009), α-synuclein (α-syn; Ebrahimi-Fakhari et al. 2011; Everall et al. 2009; Khanlou et al. 2009), and tau (Patrick et al. 2011). In contrast, activation of autophagy by gene therapy recovers the deficits in autophagy observed in α-syn tg mice (Spencer et al. 2009). However, the mechanisms by which HIV proteins might interfere with autophagy during aging are unknown. Here, we postulate that dysfunctional autophagy activity may contribute to HAND progression in aged individuals. In this report, we extend our past studies by characterizing differences in viral load (VL), CNS immune activation and expression of autophagy-related proteins among <50 years old (young) and >50 years old (aged) HIV-infected individuals. Our results indicate that while autophagy was upregulated in the brains of young HIVE patients, it was downregulated in the brains of aged HIVE patients. Similarly, autophagy was downregulated in aged gp120 tg mice and activation with beclin-1 gene transfer ameliorated the neurodegenerative phenotype. These findings provide novel insight and potentially important targets to combat HAND in the aging population.

Methods

Study population

For the present study, we included a total of 83 HIV+ cases, of which 50 are below 50 years and 33 are above 50 years (Table 1) from the HIV Neurobehavioral Research Center and California Neuro-Acquired Immunodeficiency Syndrome (AIDS) Tissue Network at the University of California, San Diego. Cases had neuromedical and neuropsychological examinations within a median of 12 months before death. Most cases died as a result of acute bronchopneumonia or septicemia and autopsy was performed within 24 h of death. Autopsy findings were consistent with AIDS and the associated pathology was most frequently due to systemic cytomegalovirus (CMV), Kaposi sarcoma, and liver disease. Subjects were excluded if they had a history of CNS opportunistic infections or non-HIV-related developmental, neurologic, psychiatric, or metabolic conditions that might affect CNS functioning (e.g., loss of consciousness exceeding 30 min, psychosis, substance dependence). The diagnosis of HIV encephalitis was based on the presence of microglial nodules, astrogliosis, HIV p24-positive cells, and myelin pallor.
Table 1
Summary of subject demographic and brain pathology information
Group
n
Gender F/M
Age
PMI (h)
Brain weight (g)
Brain pathology
Under 50 (young)
50
5/45
39.2 ± 6.04
14.0 ± 12.13
1317 ± 131.3
20 Normal, 1 Alzheimer type II gliosis, 6 microglial nodule encephalitis, 17 HIV encephalitis, 6 other
50 and older (aged)
33
5/28
55.2 ± 5.51
23.6 ± 24.49
1336 ± 176.5
13 Normal, 6 Alzheimer type II gliosis, 1 microglial nodule encephalitis, 8 HIV encephalitis, 5 other

Determination of HIV p24 levels in postmortem samples

HIV-1 p24 levels in postmortem tissues were determined using a commercially available p24 enzyme-linked immunosorbent assay (ELISA; NEK050001KT, PerkinElmer, Waltham, MA, USA). In brief, as previously described (Hashimoto et al. 2002), tissues from human brain samples (0.1 g) were homogenized in 0.7 mL of fractionation buffer containing phosphatase and protease inhibitor cocktails (Calbiochem, San Diego, CA, USA). Samples were precleared by centrifugation at 5,000×g for 5 min at room temperature. Homogenate was analyzed for protein quantity by BCA assay (Thermo Scientific) and then 100 μg of protein from each sample was assayed for p24 using the manufacturer’s protocol.

Generation of gp120 tg mice

For studies of autophagy function, an animal model of HIV protein-mediated neurotoxicity, aged (12 months) tg mice expressing high levels of gp120 under the control of the glial fibrillary acidic protein (GFAP) promoter were used (Toggas et al. 1994). These mice develop neurodegeneration accompanied by astrogliosis, microgliosis, and memory deficits in the water maze test (Toggas et al. 1994). The mice were sacrificed within 1 week of behavioral testing and brains were removed for biochemical analyses of frozen or fixed brain tissues.

Construction of lentivirus vectors

The mouse beclin-1 cDNA (Open Biosystems) was PCR amplified and cloned into the third-generation self-inactivating lentivirus vector (Naldini et al. 1996a, b) with the CMV promoter driving expression producing the vector LV-beclin-1. Lentiviruses expressing beclin-1, luciferase or empty vector (as controls) was prepared by transient transfection in 293 T cells (Naldini et al. 1996a, b; Tiscornia et al. 2006; Spencer et al. 2009).

Mouse lines and intracerebral injections of lentiviral vectors

A cohort of aged (12 months) mice (n = 20), gp120 tg (n = 10), and control mice (n = 10) were injected with 3 μl of the lentiviral preparations (2.5 × 107 TU) into the temporal cortex (using a 5 μl Hamilton syringe). Briefly, as previously described (Marr et al. 2003), mice were placed under anesthesia on a Koft stereotaxic apparatus and coordinates (hippocampus: AP, 2.0 mm; lateral, 1.5 mm; depth, 1.3 mm and cortex: AP, 0.5 mm; lateral, 1.5 mm; depth, 1.0 mm) were determined as per the Franklin and Paxinos (1997) atlas. The lentiviral vectors were delivered using a Hamilton syringe connected to a hydraulic system to inject the solution at a rate of 1 μl every 2 min. To allow diffusion of the solution into the brain tissue, the needle was left for an additional 5 min after the completion of the injection. Mice received unilateral injections (right side) to allow comparisons against the contralateral side, with either LV-beclin-1 (n = 5) or LV-control (n = 5). Additional controls were performed by injecting non-tg littermates with either LV-beclin-1 (n = 5) or LV-control (n = 5). Mice survived for 3 months after the lentiviral injection. As an additional control for LV injection, age-matched littermates were injected with LV-luciferase.
Since no differences were observed between the LV-control and the LV-luciferase, all data presented in this paper are shown with the LV-control vector. Following National Institutes of Health (NIH) guidelines for the humane treatment of animals, mice were anesthetized with chloral hydrate and flush-perfused transcardially with 0.9 % saline. Brains and peripheral tissues were removed and divided in sagittal sections. The right hemibrain was post-fixed in phosphate-buffered 4 % PFA (pH 7.4) at 4 °C for 48 h for neuropathological analysis, while the left hemibrain was snap-frozen and stored at −70 °C for subsequent RNA and protein analysis.

Antibodies

For western blot and immunohistochemical analysis of the autophagy pathway, polyclonal antibodies against GFAP (MAB3402, dilution 1:500, Millipore), cathepsin-D (1:500, Calbiochem, San Diego, CA, USA), Iba-1 (1:1,000, Wako Corp., Japan), microtubule-associated protein (MAP) 2 (MAP378, 1:250, Millipore), LC3 (1:1,000, Abcam), mTor (1:1,000, Sigma), beclin-1 (1:1,000, Novus Biologicals, Littleton, CO, USA).

Immunoblot analysis

Frontal cortex tissues from human and mouse brains were homogenized and fractionated using a buffer that facilitates separation of the membrane and cytosolic fractions (1.0 mmol/L HEPES, 5.0 mmol/L benzamidine, 2.0 mmol/L 2-mercaptoethanol, 3.0 mmol/L EDTA, 0.5 mmol/L magnesium sulfate, 0.05 % sodium azide; final pH, 8.8). In brief, as previously described (Hashimoto et al. 2002), tissues from human and mouse brain samples (0.1 g) were homogenized in 0.7 mL of fractionation buffer containing phosphatase and protease inhibitor cocktails (Calbiochem, San Diego, CA, USA). Samples were precleared by centrifugation at 5,000×g for 5 min at room temperature. Supernatants were retained and placed into appropriate ultracentrifuge tubes and were centrifuged at 436,000×g for 1 h at 4 °C in a TL-100 rotor (Beckman Coulter, Brea, CA, USA). This supernatant was collected as representing the cytosolic fraction, and the pellets were resuspended in 0.2 mL of buffer and rehomogenized for the membrane fraction.
After determination of the protein content of all samples by BCA protein assay (Thermo Fisher Scientific, Rockford, IL, USA), homogenates were loaded (20 μg total protein/lane), separated on 4–12 % Bis-Tris gels and electrophoresed in 5 % HEPES running buffer, and blotted onto Immobilon-P 0.45 μm membrane using NuPage transfer buffer. The membranes were blocked in either 5 % nonfat milk/1 % BSA in phosphate-buffered saline (PBS) + 0.05 % Tween-20 (PBST) or in 5 % BSA in PBST for 1 h. Membranes were incubated overnight at 4 °C with primary antibodies. Following visualization, blots were stripped and probed with a mouse monoclonal antibody against Actin (1:2,000, mab1501, Millipore, Billerica, MA, USA) as a loading control. All blots were then washed in PBS, 0.05 % tween-20 and then incubated with secondary species-specific antibodies (American Qualex, 1:5,000 in BSA-PBST) and visualized with enhanced chemiluminescence reagent (Perkin-Elmer). Images were obtained and semiquantitative analysis was performed with the VersaDoc gel imaging system and Quantity One software (Bio-Rad).

immunohistochemistry, image analysis, and laser scanning confocal microscopy

Briefly, as previously described (Masliah et al. 2003), free-floating 40 μm thick vibratome sections were washed with Tris-buffered saline (TBS, pH 7.4), pretreated in 3 % H2O2, and blocked with 10 % serum (Vector Laboratories, Burlingame, CA, USA), 3 % bovine serum albumin (Sigma), and 0.2 % gelatin in TBS-Tween. For human brains, sections from the midfronal cortex were used; for the mice, sagittal sections from the complete brain were studied. Sections were incubated at 4 °C overnight with the primary antibodies. Sections were then incubated in secondary antibody (1:75, Vector), followed by Avidin d-horseradish peroxidase (HRP, ABC Elite, Vector) and reacted with diaminobenzidine (0.2 mg/ml) in 50 mM Tris (pH 7.4) with 0.001 % H2O2. Control experiments consisted of incubation with pre-immune rabbit serum. To investigate the effects of postmortem delay and fixation on the levels of mTor immunoreactivity, preliminary studies were performed in a subset of cases (n = 5) with postmortem delay ranging from 4 to 48 h. In addition, double immunolabeling studies were performed as previously described (Spencer et al. 2009) to determine the cellular localization of the autophagy markers. For this purpose, vibratome sections from the young and aged HIV+ and HIVE+ cases as well as of the non-tg and gp120 tg mice were immunostained with antibodies against the neuronal marker MAP2 (mouse monoclonal) and antibodies against either LC3, mTor, or Cathepsin-D (rabbit polyclonal). Sections were then reacted with secondary antibodies tagged with FITC to detect MAP2 and mTor and with the tyramide Red amplification system (Perkin-Elmer) to detect Cathepsin-D. Conversely for the experiments analyzing LC3, this autophagy marker was detected with FITC while MAP2 was detected with the tyramide Red amplification system (Perkin-Elmer). Sections were mounted on superfrost slides (Fisher) and coverslipped with media containing DAPI. Double-immunolabeled sections were imaged with the laser scanning confocal microscope as described below.
Immunostained sections were imaged with a digital Olympus microscope and assessment of levels of GFAP, Iba1, beclin-1, Cathepsin-D, and mTor immunoreactivity was performed utilizing the Image-Pro Plus program (Media Cybernetics, Silver Spring, MD, USA). For each case, a total of three sections (10 images per section) were analyzed in order to estimate the average number of immunolabeled cells per unit area (square millimeter) and the average intensity of the immunostaining (corrected optical density).
For confocal images (LC3 and MAP2), all sections were processed simultaneously under the same conditions and experiments were performed twice for reproducibility. Sections were imaged with a Zeiss 63× (N.A. 1.4) objective on an Axiovert 35 microscope (Zeiss, Germany) with an attached MRC1024 laser scanning confocal microscope system (BioRad, Hercules, CA, USA).

Analysis of neurodegeneration

Neuronal structural integrity was evaluated as previously described (Rockenstein et al. 2005a, b). In brief, blind-coded, 40 μm thick microtome sections from human and mouse brains fixed in 4 % paraformaldehyde were immunolabeled with mouse monoclonal antibodies against MAP2 (1:200). After overnight incubation, sections were incubated with fluorescein isothiocyanate-conjugated secondary antibodies (1:75; Vector Laboratories, Burlingame, CA, USA), transferred to SuperFrost slides (Fisher Scientific, Tustin, CA, USA), and mounted under glass coverslips with antifading medium (Vector Laboratories). All sections were processed under the same standardized conditions. The immunolabeled blind-coded sections were serially imaged with a laser scanning confocal microscope (MRC-1024; Bio-Rad) and analyzed with ImageJ v1.43 software (NIH, Bethesda, MD, USA), as previously described (Crews et al. 2010). For each mouse, a total of three sections were analyzed; for each section, four fields in the frontal cortex and hippocampus were examined. Results were expressed as percent area of the neuropil occupied by immunoreactive signal. All sections were processed under the same standardized conditions. Immunostained sections were imaged with a digital Olympus microscope and the Image-Pro Plus program (version 4.5.1, Media Cybernetics).

Statistical analysis

All the analyses were conducted on blind-coded samples. After the results were obtained, the code was broken and data were analyzed with the StatView program (SAS Institute, Inc., Cary, NC, USA). Comparisons among groups were performed with one-way ANOVA, unpaired Student’s t test and Chi-square analysis. All results were expressed as mean ± SEM. The differences were considered to be significant if p values were <0.05.

Role of the funding source

The funding source had no role in the study design, data collection, data analysis, data interpretation or writing of this report. The corresponding author had full access to all the data and had final responsibility for the decision to submit the paper for publication.

Results

Clinical and neuropathological characteristics of young and aged donors with and without HIVE

Before analyzing autophagy protein levels, we first meticulously collected and characterized clinical and neuropathological aspects of the young and aged HIV+ cohort. For this purpose, at first, a total of 83 autopsy cases were analyzed (n = 50 young and n = 33 aged). In order to assess relevant differences between postmortem brain samples from young and aged HIV patients, we characterized the cohorts by brain pathology, CD4+ cell count, viral load, and neuropsychological impairment status. Table 1 demonstrates both young and aged cohorts were predominantly male and 40 % of each cohort had normal brain pathology (Table 1). Only one (2 %) of the young cases showed Alzheimer type II gliosis, whereas six (18 %) of the 33 aged patients showed Alzheimer type II gliosis (Table 1). Conversely, one (3 %) of the aged patients presented microglial nodule encephalitis, while microglial nodule encephalitis was present in six (12 %) of the young patients (Table 1). Seventeen (33 %) of young cases showed HIVE versus eight (24 %) among the aged patients (Table 1). Of the 83 autopsy cases, complete neurovirological and neurological analysis was available in 55 cases (n = 31 younger and n = 24 aged). Further analyses revealed that young cases had significantly (p < 0.05) lower CD4+ cell counts compared to aged patients, and significantly (p < 0.05) higher plasma VL (Table 2). CSF VL was equivalent between young and aged groups (Table 2). Assessment of neuropsychological impairment showed that nine (18 %) of the young group suffered HAD, and another nine (18 %) suffered minor cognitive motor deficits, whereas only one (3 %) and two (6 %) donors of the aged group suffered from these afflictions, respectively (Table 2). Twenty percent of cases from each group presented NPI-other (O), and the aged group had two NPI-unknown (U) cases (Table 2). Five of the young patients were unable to complete assessment and two were normal, while four of the aged patients were unable to complete assessment and six were normal (Table 2). These data indicate stark differences in the pathology and neuropsychological state between young and aged cases in this HIV+ cohort.
Table 2
CD4+ cell count, plasma VL, presence of neuropsychological impairment, and CSF VL in the young and aged HIV+ cohort
Group
CD4*
Log plasma VL*
Log CSF VL
NPI
ARV+/−
Under 50 (young)
70.2 ± 151.9 (n = 29)
4.53 ± 1.39 (n = 29)
3.14 ± 1.35 (n = 30)
9 HAD, 9 MCMD, 3 NPI, 10 NPI-O, 5 unable, 2 normal
16/15
50 and older (aged)
241.2 ± 325.4 (n = 25)
3.64 ± 1.70 (n = 23)
2.64 ± 1.25 (n = 21)
1 HAD, 2 MCMD, 3 NPI, 7 NPI-O, 2 NPI-U, 4 unable, 6 normal
14/10
n number of independent donors, MCMD minor cognitive motor deficits
*p < 0.05

HIV capsid protein, p24, levels are increased in aged and neuropsychological impaired patients

To further characterize neuropathology among the young and aged cohorts, p24 levels in postmortem brain tissue from young and aged HIV patients were measured by ELISA and correlated to neuropathology and neuropsychological impairment. p24 levels were statistically equal between young and aged HIV patients (Fig. 1a). For this analysis, tissues were available for all 83 cases, of them n = 50 young (n = 33 HIV+ and n = 17 HIVE) and n = 33 aged (n = 23 HIV+ and n = 10 HIVE). p24 levels were increased in postmortem tissues from young and aged HIVE patients compared to age-matched HIV controls (Fig. 1b, p < 0.05 and p < 0.001, respectively). p24 levels in postmortem tissues from aged HIVE patients were significantly increased compared to young HIVE patients (Fig. 1b, p < 0.001). Similarly, p24 levels are increased in young and aged patients with any neuropsychological impairment compared to age-matched HIV controls (Fig. 1c). p24 levels were highest in the aged patients with neuropsychological impairment (Fig. 1c). Data represent analysis of the entire cohort of HIV+ and HIVE donor brains.

Neuroinflammation and neuropathology were increased in postmortem brains from aged HIVE patients

Next, we measured levels of inflammatory and neuronal markers by immunohistochemical analysis in a subgroup of n = 29 postmortem cases, n = 15 young (n = 7 HIV+ and n = 8 HIVE) and n = 14 aged (n = 8 HIV+ and n = 6 HIVE). Vibratome sections from paraformaldehyde fixed frontal cortex were used. Inflammatory markers GFAP and Iba-1 were increased, while MAP2 (somato-dendritic marker) immunostaining was decreased in brain tissue from aged HIVE patients compared to young HIVE patients (Fig. 2a–c). Astroglial marker, GFAP immunoreativity, and signal were elevated in young and aged HIVE tissues versus young and aged HIV+ samples; however, GFAP signal was most intense in tissues from aged HIVE patients (Fig. 2a). The microglial marker, Iba-1, signal intensity was increased in brain tissues of young and aged HIVE patients compared to HIV+ patients from both groups, with cells from HIVE tissues having more extended processes in greater numbers (Fig. 2b). MAP2 neuronal marker immunoreactivity was opposite that of GFAP and Iba-1 staining; young and aged HIVE tissues had reduced MAP2 signal compared to young and aged HIVE tissues (Fig. 2c). Furthermore, MAP2 staining in aged HIVE tissues was the most punctate and discontinuous, as indicated by the white arrows (Fig. 2c). GFAP and Iba-1 optical density in HIVE samples was significantly (p < 0.001) increased compared to age-matched HIV+ samples (Fig. 2d–e). GFAP optical density was significantly (p < 0.05) increased in aged HIVE samples compared to young HIVE samples (Fig. 2d). Quantification of MAP2 neuronal marker indicated area of the neuropil stained decreased from 26 % (in young HIV+ patients) to 20 % (area of neuropil in young HIVE patients; p < 0.01). MAP2 levels were decreased from 24 % (area of the neuropil in aged HIV+ patients) to 16 % (area of neuropil in aged HIVE patients; Fig. 2e, p < 0.001). MAP2 (16 % of the neuropil) levels in aged HIVE samples were significantly (p < 0.05) reduced compared to young HIVE samples (21 % of neuropil).

Autophagy markers are reduced in aged HIVE patients compared to young HIVE patients

To analyze autophagy pathway activity, aged and young HIV+ and HIVE brain beclin-1, cathepsin-D, LC3, and mTor were analyzed immunohistochemistry (Fig. 3a–h). Immunohistochemical analysis of pyramidal neurons in layers II–III and V of the midfrontal cortex from young and aged HIV+ and HIVE patients showed beclin-1, cathepsin-D, and LC-3 levels were increased in young HIVE versus young HIV+ samples; however, levels of all three autophagy markers were decreased in aged HIVE versus aged HIV+ brains (Fig. 3a–c). For each case analysis of beclin-1, cathepsin-D and LC-3 immunoreactivity was performed in comparable layers of the neocortex to assure consistent comparisons. The black box inset denotes area of high cathepsin-D immunoreactivity in young HIV+ patients (Fig. 3b). White arrows indicate LC3 staining of autophagosomes in neurons (Fig. 3c). Levels of mTor were moderate in young HIV+ and HIVE brains, but were increased in aged HIV+ and HIVE brains (Fig. 3d). Beclin-1 optical density is significantly (p < 0.001) increased in young HIVE compared to young HIV+ samples; however, beclin-1 optical density in aged HIVE samples is significantly (p < 0.001) reduced compared to levels in young HIVE samples (Fig. 3e). Cathepsin-D optical density levels were significantly increased in young HIVE compared to levels in young HIV+ brains and similar to beclin-1 levels, cathepsin-D levels in aged brains were significantly (p < 0.01) reduced compared to young HIVE samples (Fig. 3f). Quantification of LC3 confocal image pixel intensity revealed LC3 levels in young HIVE brains are significantly (p < 0.001) reduced compared to levels young HIV+ brains (Fig. 3g). Aged HIV+ and HIVE brain LC3 levels are equivalent but significantly (p < 0.001) reduced compared to young HIVE brain LC3 levels. Quantification of mTor optical density reveals young HIV+ and HIVE brains have similar mTor expression; however, mTor levels in aged HIV+ and HIVE were increased compared to young samples (Fig. 3h). Furthermore, mTor levels in aged HIVE samples were significantly (p < 0.01) higher than levels in aged HIV+ samples (Fig. 3h). To confirm that the alterations in markers of autophagy were in pyramidal neurons of layers II–III and V, double labeling and confocal microscopy studies were performed. Both in the young and aged HIV+ cases, most (approximately 90 %) of the cathepsin-D, LC3, and mTor immunoreactive granules in the cytoplasm of cells were observed in MAP2 immunoreactive neurons (Fig. 4a).
To corroborate immunohistochemical data beclin-1, LC3, mTor, and actin were measured by immunoblot in postmortem brain samples from aged and young HIV+ and HIVE patients (Fig. 5a–c). Beclin-1 and LC3 signals were increased in young HIVE compared to young HIV+ samples; however, beclin-1 and LC3 signals were decreased in aged HIVE compared to aged HIV+ samples (Fig. 5a). Analysis of band density revealed that young HIVE beclin-1 levels were increased 2.5-fold compared to young HIV+ samples (p < 0.05); however, aged HIVE sample beclin-1 levels were decreased by 60 % compared to aged HIV+ samples (Fig. 5b, p < 0.05). Similarly, LC3 band density analysis revealed that levels were increased in young HIVE compared to young HIV+ patients (p < 0.05), yet aged HIVE LC3 levels were significantly reduced (p < 0.05) compared to aged HIV+ LC3 levels (Fig. 5c).

Autophagy markers are reduced in aged gp120-tg mice; lentivirus delivery of beclin-1 restores autophagy marker expression

To investigate alterations in the autophagy pathway in an experimental model of HIV protein neurotoxicity, immunochemical analysis was performed in aged (12 months) gp120 tg and non-tg mice. Immunohistochemical analysis showed that compared to aged non-tg mice, levels of beclin-1, cathepsin-D, and LC3 are reduced and mTor is increased in brains of aged gp120 tg mice. Lentiviral-mediated delivery of beclin-1 via stereotaxic injection to the temporal cortex of gp120 tg mice increased beclin-1, cathepsin-D, and LC3 levels in aged non-tg and aged gp120 tg mice (Fig. 6a–c). Beclin-1 delivery significantly (p < 0.05) reduced mTor levels in aged gp120 tg mice (Fig. 6d). Quantification of autophagy marker optical density or pixel intensity showed beclin-1, cathepsin-D, and LC3 levels were significantly (p < 0.05) reduced in aged gp120 tg brains compared to levels in aged non-tg control mice brains (Fig. 6e–g). Levels of mTor immunoreactivity were increased in aged gp120 tg mice compared to aged non-tg mice, and this was reversed by LV-mediated beclin-1 delivery (Fig. 6h). To confirm that the effects of the lentivirus on markers of autophagy are in pyramidal neurons, double labeling and confocal microscopy studies were performed. Both in the non-tg and gp120 tg mice treated with the LV-beclin1 the majority (approximately 90 %) of the cathepsin-D, LC3, and mTor immunoreactive cells were localized to MAP2 immunostained cells (Fig. 4a).

LV-mediated beclin-1 delivery reduces neuroinflammation and neurodegeneration in aged gp120 tg mice brains

Overexpression of beclin-1 from the lentivirus reduced astrogliosis and microgliosis markers and neurodegeneration in gp120 tg mice. Immunohistochemical analyses of gp120 tg mice brains show GFAP and Iba1 staining was increased while MAP2 staining was decreased compared to control non-tg mice (Fig. 7a–c). GFAP positive cells were more numerous with extended processes and displayed more intense GFAP signal (Fig. 7a). Similarly, Iba1 signal was increased in gp120 tg mouse brains compared to non-tg control mice (Fig. 7b). gp120 tg mouse brains expressed reduced levels of MAP2 in a more punctate and discontinuous pattern (Fig. 7c). Compared with the LV-control, LV-beclin-1 injection reduced GFAP and Iba1 signal increased and normalized MAP2 staining in gp120 tg mouse brains (Fig. 7a–c). Quantification of GFAP, Iba1, and MAP2 immunoreactivity corroborated evidence that LV-beclin-1 reduced astrogliosis, microgliosis, and neurodegeneration in GFAP gp120 tg mice (Fig. 7d–f). GFAP optical density was equivalent between LV-control and LV-beclin-1 treated non-tg mice; however, GFAP optical density was significantly (p < 0.05, twofold) elevated in gp120 tg mice brains compared to non-tg mice (Fig. 7d). LV-beclin-1 significantly (p < 0.05, −30 %) reduced GFAP optical density in gp120 tg mice brains (Fig. 7d). Iba1 staining was significantly (p < 0.05) elevated in gp120 tg mice compared to non-tg mice; however, LV-beclin-1 significantly (p < 0.05) reduced Iba1 staining in fp120 tg mouse brains (Fig. 7e). The area of the neuropil stained by MAP2 was significantly (p < 0.05, −25 %) reduced in gp120 tg mouse brains compared to non-tg mice; LV-beclin-1 restored gp120 tg brain MAP2 staining to levels of non-tg mice (Fig. 7f).

Discussion

In the current study, we show for the first time differential levels of autophagy proteins in postmortem brain tissue samples from aged HIVE and young HIVE patients. While the young HIVE group display increased autophagy markers, the aged HIVE group showed reduced autophagy pathway proteins. Viral protein and inflammatory marker levels were increased while autophagy proteins beclin-1, LC3 and the neuronal marker MAP2 were reduced in HIVE brain tissues from aged donors compared to those from young donors. Similarly, analysis of aged gp120 tg mouse brains showed reduced autophagy marker expression and increased neurodegeneration compared to aged non-tg mice. LV-beclin-1 gene transfer restored autophagy markers and reduced neuroinflammation and neurodegeneration in aged gp120 tg mouse brains. Previous studies have shown that HIV can alter the autophagy pathway, but these studies show possible reduced autophagy function in aged HIVE patients and increased inflammation markers accompanied with simplified neuronal synaptic complexity compared to young HIVE donors. Furthermore, LV-mediated beclin-1 gene transfer restores autophagy function and reduces neurodegeneration in aged gp120 tg mice. HIV infection in young patients may differ markedly to that in aged patients with respect to autophagy and neuronal injury; these data corroborate evidence that HIV infection may exacerbate consequences of normal aging and may provide a potential therapy for aged HAND patients.
The present studies are consistent with recent findings that suggest the autophagy pathway is dysregulated during AD, PD, and HIV (Ebrahimi-Fakhari et al. 2011; Pickford et al. 2008; Zhou et al. 2011); however, this is the first report of distinct differences in autophagy marker expression among young and aged HIVE patients. A survey of HIV patients showed overall autophagy markers are increased in postmortem samples from HIV-infected individuals (Zhou et al. 2011). By further categorizing patients according to age and HIVE status, the current studies revealed distinct differences in aged and young HIVE brains. These differences in autophagy function among young and aged HIVE patients may contribute to the growing list of chronic comorbidities among aged HIV patients. The differences observed in brains from deceased young versus aged HIVE patients suggest that two distinct types of infection may be responsible for the differences observed between young and aged HIVE patients: (1) viremia is robust and infiltrates the CNS leading to early neural complications and death (young) or (2) viremia is subdued throughout infection, the CNS is not dramatically affected early on, yet long-term infection gradually increases neuroinflammation and neurodegeneration (aged). HIV patients that live over 50 years may harbor a low but chronic viral burden that can limit normal autophagy function and lead to proteinopathies such as seen in AD and PD. Another alternative is that HIV infection progresses similarly in most patients but some patients escape the battery of opportunistic infections to live a longer life. The compensatory autophagy response seen in young patients may be meant to clear excessive toxic molecule build up until the system is overwhelmed and the molecular recycling system fails. Reduced autophagy marker levels detected in aged HIVE samples may represent less viable cells with reduced capacity to perform normal physiological processes, such as autophagy. Consistent with this notion, Aβ expression correlates with age in postmortem samples from HIVE patients, and in the second scenario this may be a natural occurrence of most chronic HIV infections (Achim et al. 2009; Khanlou et al. 2009). In either case, understanding how HIV affects neuronal autophagy may lead to therapies that slow down neurodegeneration. Importantly, neither of these scenarios considers those individuals who contract HIV infection after turning 50, for which alternative patterns of disease have been reported (Erlandson et al. 2012). Additional studies using samples from large cohorts of HIV patients combined with animal models and cellular-based studies will prove valuable in dissecting the complications of aging with neuroAIDS.
Beclin-1 and LC3 are essential proteins in the autophagy pathway, and interference at either level may result in cellular toxicity (Kragh et al. 2012). Beclin-1 mediates autophagosome nucleation and LC3 is an integral autophagy protein that facilitates autophagosome elongation and closure and may also be involved in disease (Dinkins et al. 2010; Gannage et al. 2010; Kyei et al. 2009). Despite increased autophagy machinery, HIV patients have increased Aβ and α-synuclein accumulation in the brain (Achim et al. 2009; Khanlou et al. 2009). These and other reports suggest that dysfunctional autophagy is associated with neurodegenerative diseases and may exacerbate problems associated with HIV infection of the CNS. In this context, it may be possible to reintroduce functional autophagy by gene transfer to ameliorate HIVE phenotypes.
It was recently reported that HIV-1 infection inhibits autophagy pathways in monocytes, but no direct effect on neurons has been shown (Van Grol et al. 2010). Kyei et al. showed that the HIV-1 protein Nef interacts with autophagy pathways in macrophages (Kyei et al. 2009). In subsequent findings, groups have shown that HIV-1 may highjack the autophagy pathway to promote replication (Gregoire et al. 2011; Killian 2012). Despite these advances, it is not known if HIV directly or indirectly affects neuronal autophagy and ultimately contributes to neurodegeneration. Our current studies show aged HIVE patients express increased neuroinflammatory markers and reduced neuronal markers, indicating progressed neurodegeneration, and loss of synaptic processes may be associated with decreased autophagy-related proteins (beclin-1 and LC3). The gp120 tg mouse serves as a model for HIV-1-protein mediated neurotoxicity and the mice present neuropathology and behavioral deficits similar to those seen in HAND patients (Toggas et al. 1994). Furthermore, in the α-syn tg mouse model for PD, LV-mediated delivery of beclin-1 to the CNS reversed neuropathology and reduced the accumulation of α-syn (Spencer et al. 2009). The current studies further support the hypothesis that proteinopathies may be due to dysfunctional autophagy and that normal molecular recycling may be restored via LV-mediated beclin-1 gene transfer. Indeed, LV-beclin-1 ameliorated astrogliosis, microgliosis, and neurodegeneration in the gp120 tg mouse model and may serve as a therapy for aged HAND patients.
Impetus has been placed on understanding how aging may exacerbate HAND. Healthy individuals >65 are designated elderly by the Centers for Disease Control, but HIV infected individuals are designated “aged” HIV patients at >50, due to progressed signs of aging. It is predicted that by 2015, over 50 % of AIDS cases in the USA, and 15 % of newly diagnosed cases will be >50 (Erlandson et al. 2012; Watkins and Treisman 2012). Reports have demonstrated that aged HIV patients have increased risk of severe depression and cognitive impairment, less time between infection and AIDS diagnosis and reduced T-lymphocyte proliferation (Watkins and Treisman 2012). Further studies are needed to characterize and combat HIV in this particularly vulnerable group. Our data support the hypothesis that HIV exacerbates the effects of aging and implicate dysregulated autophagy as a possible cause of HAND.
Studies on mechanisms of HIV pathogenesis in aged patients are lacking; aging with HIV is a relatively novel problem. Most studies consider neuroAIDS with respect to the HIV-infected population as a whole, yet physicians and the CDC note stark differences in the progression of disease between young and aged groups (Erlandson et al. 2012; Watkins and Treisman 2012). In an effort to understand differences in HIV pathogenesis between young and aged patients, we surveyed 83 HIV patients, 33 of which were >50 years old. Among the young and aged groups, 40 % of each group was classified as normal brain pathology; however, 18 % of the aged group presented with Alzheimer type II gliosis in contrast to only one patient from the young group. Also noteworthy, 34 % of the young group had encephalitis versus 24 % of the aged group. High VL and encephalitis may have contributed to the premature demise of young patients, whereas persistent low-level infection, as seen in aged patients, may contribute to the Alzheimer-like pathology. In contrast to past reports, the current studies indicate that CD4+ cell count was higher in aged brains, and CNS plasma viral burden is reduced in aged HIV patients; yet, p24 and inflammatory markers are increased compared to their younger counterparts. Chronic inflammation may promote long-term deleterious changes in gene expression in aged HIV infected individuals, which may explain how aged HIV patients suffer increased morbidity despite higher CD4+ cell counts. Cohort size and population dynamics all contribute to data variability.
The proportion of HIV-1-infected individuals over the age of 50 is rapidly increasing, yet therapies to prevent or treat HAND remain scant (Erlandson et al. 2012; Watkins and Treisman 2012). Furthermore, the HIV-infected population is not alone as the average age of the general population is increasing along with the prevalence of age-associated neurodegenerative diseases. These studies implicate autophagy as a novel target for preventing and treating HAND, AD, PD, and possibly other neurodegenerative disorders. Importantly, this report suggests that young and aged HIV patients may require unique drug regimens to combat progression in these two distinct stages of disease.

Acknowledgments

We thank the National Institutes of Aging (AG043384) and the National Institutes of Mental Health for (MH062962, MH5974 and MH83506) funding this work.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Neurologie & Psychiatrie

Kombi-Abonnement

Mit e.Med Neurologie & Psychiatrie erhalten Sie Zugang zu CME-Fortbildungen der Fachgebiete, den Premium-Inhalten der dazugehörigen Fachzeitschriften, inklusive einer gedruckten Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Literatur
Zurück zum Zitat Achim CL, Adame A, Dumaop W, Everall IP, Masliah E (2009) Increased accumulation of intraneuronal amyloid beta in HIV-infected patients. J Neuroimmune Pharmacol 4:190–199PubMedCrossRef Achim CL, Adame A, Dumaop W, Everall IP, Masliah E (2009) Increased accumulation of intraneuronal amyloid beta in HIV-infected patients. J Neuroimmune Pharmacol 4:190–199PubMedCrossRef
Zurück zum Zitat Alirezaei M, Kiosses WB, Flynn CT, Brady NR, Fox HS (2008a) Disruption of neuronal autophagy by infected microglia results in neurodegeneration. PLoS One 3:e2906PubMedCrossRef Alirezaei M, Kiosses WB, Flynn CT, Brady NR, Fox HS (2008a) Disruption of neuronal autophagy by infected microglia results in neurodegeneration. PLoS One 3:e2906PubMedCrossRef
Zurück zum Zitat Alirezaei M, Kiosses WB, Fox HS (2008b) Decreased neuronal autophagy in HIV dementia: a mechanism of indirect neurotoxicity. Autophagy 4:963–966PubMed Alirezaei M, Kiosses WB, Fox HS (2008b) Decreased neuronal autophagy in HIV dementia: a mechanism of indirect neurotoxicity. Autophagy 4:963–966PubMed
Zurück zum Zitat Budka H, Costanzi G, Cristina S, Lechi A, Parravicini C, Trabattoni R, Vago L (1987) Brain pathology induced by infection with the human immunodeficiency virus (HIV). A histological, immunocytochemical, and electron microscopical study of 100 autopsy cases. Acta Neuropathol(Berl) 75:185–198CrossRef Budka H, Costanzi G, Cristina S, Lechi A, Parravicini C, Trabattoni R, Vago L (1987) Brain pathology induced by infection with the human immunodeficiency virus (HIV). A histological, immunocytochemical, and electron microscopical study of 100 autopsy cases. Acta Neuropathol(Berl) 75:185–198CrossRef
Zurück zum Zitat Carroll-Anzinger D, Al-Harthi L (2006) Gamma interferon primes productive human immunodeficiency virus infection in astrocytes. J Virol 80:541–544PubMedCrossRef Carroll-Anzinger D, Al-Harthi L (2006) Gamma interferon primes productive human immunodeficiency virus infection in astrocytes. J Virol 80:541–544PubMedCrossRef
Zurück zum Zitat Cherner M, Cysique L, Heaton RK, Marcotte TD, Ellis RJ, Masliah E, Grant I (2007) Neuropathologic confirmation of definitional criteria for human immunodeficiency virus-associated neurocognitive disorders. J Neurovirol 13:23–28PubMedCrossRef Cherner M, Cysique L, Heaton RK, Marcotte TD, Ellis RJ, Masliah E, Grant I (2007) Neuropathologic confirmation of definitional criteria for human immunodeficiency virus-associated neurocognitive disorders. J Neurovirol 13:23–28PubMedCrossRef
Zurück zum Zitat Codogno P, Mehrpour M, Proikas-Cezanne T (2012) Canonical and non-canonical autophagy: variations on a common theme of self-eating? Nat Rev Mol Cell Biol 13:7–12 Codogno P, Mehrpour M, Proikas-Cezanne T (2012) Canonical and non-canonical autophagy: variations on a common theme of self-eating? Nat Rev Mol Cell Biol 13:7–12
Zurück zum Zitat Crews L, Spencer B, Desplats P, Patrick C, Paulino A, Rockenstein E, Hansen L, Adame A, Galasko D, Masliah E (2010) Selective molecular alterations in the autophagy pathway in patients with Lewy body disease and in models of alpha-synucleinopathy. PLoS One 5:e9313PubMedCrossRef Crews L, Spencer B, Desplats P, Patrick C, Paulino A, Rockenstein E, Hansen L, Adame A, Galasko D, Masliah E (2010) Selective molecular alterations in the autophagy pathway in patients with Lewy body disease and in models of alpha-synucleinopathy. PLoS One 5:e9313PubMedCrossRef
Zurück zum Zitat Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305:1292–1295PubMedCrossRef Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305:1292–1295PubMedCrossRef
Zurück zum Zitat Dinkins C, Arko-Mensah J, Deretic V (2010) Autophagy and HIV. Semin Cell Dev Biol 21:712–718PubMedCrossRef Dinkins C, Arko-Mensah J, Deretic V (2010) Autophagy and HIV. Semin Cell Dev Biol 21:712–718PubMedCrossRef
Zurück zum Zitat Ebrahimi-Fakhari D, Cantuti-Castelvetri I, Fan Z, Rockenstein E, Masliah E, Hyman BT, McLean PJ, Unni VK (2011) Distinct roles in vivo for the ubiquitin-proteasome system and the autophagy-lysosomal pathway in the degradation of alpha-synuclein. The J of neurosci: the official j of the Soc for Neurosci 31:14508–14520CrossRef Ebrahimi-Fakhari D, Cantuti-Castelvetri I, Fan Z, Rockenstein E, Masliah E, Hyman BT, McLean PJ, Unni VK (2011) Distinct roles in vivo for the ubiquitin-proteasome system and the autophagy-lysosomal pathway in the degradation of alpha-synuclein. The J of neurosci: the official j of the Soc for Neurosci 31:14508–14520CrossRef
Zurück zum Zitat Erlandson KM, Allshouse AA, Duong S, Mawhinney S, Kohrt WM, Campbell TB (2012) HIV, aging, and advance care planning: are we successfully planning for the future? J Palliat Med 15(10):1124–1129PubMedCrossRef Erlandson KM, Allshouse AA, Duong S, Mawhinney S, Kohrt WM, Campbell TB (2012) HIV, aging, and advance care planning: are we successfully planning for the future? J Palliat Med 15(10):1124–1129PubMedCrossRef
Zurück zum Zitat Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Letendre S, Moore D, Ellis R, Cherne M, Gelman B, Morgello S, Singer E, Grant I, Masliah E (2009) Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J Neurovirol 15(5–6):360–370PubMedCrossRef Everall I, Vaida F, Khanlou N, Lazzaretto D, Achim C, Letendre S, Moore D, Ellis R, Cherne M, Gelman B, Morgello S, Singer E, Grant I, Masliah E (2009) Cliniconeuropathologic correlates of human immunodeficiency virus in the era of antiretroviral therapy. J Neurovirol 15(5–6):360–370PubMedCrossRef
Zurück zum Zitat Franklin KBJ, Paxinos G (1997) The mouse brain in stereotaxic coordinates. Academic Press, San Diego Franklin KBJ, Paxinos G (1997) The mouse brain in stereotaxic coordinates. Academic Press, San Diego
Zurück zum Zitat Gannage M, Ramer PC, Munz C (2010) Targeting Beclin 1 for viral subversion of macroautophagy. Autophagy 6:166–167PubMedCrossRef Gannage M, Ramer PC, Munz C (2010) Targeting Beclin 1 for viral subversion of macroautophagy. Autophagy 6:166–167PubMedCrossRef
Zurück zum Zitat Gendelman HE, Persidsky Y, Ghorpade A, Limoges J, Stins M, Fiala M, Morrisett R (1997) The neuropathogenesis of the AIDS dementia complex. Aids 11(A):S35–S45PubMed Gendelman HE, Persidsky Y, Ghorpade A, Limoges J, Stins M, Fiala M, Morrisett R (1997) The neuropathogenesis of the AIDS dementia complex. Aids 11(A):S35–S45PubMed
Zurück zum Zitat Gonzalez-Scarano F, Martin-Garcia J (2005) The neuropathogenesis of AIDS. Nat Rev Immunol 5:69–81PubMedCrossRef Gonzalez-Scarano F, Martin-Garcia J (2005) The neuropathogenesis of AIDS. Nat Rev Immunol 5:69–81PubMedCrossRef
Zurück zum Zitat Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906PubMedCrossRef Gozuacik D, Kimchi A (2004) Autophagy as a cell death and tumor suppressor mechanism. Oncogene 23:2891–2906PubMedCrossRef
Zurück zum Zitat Gregoire IP, Richetta C, Meyniel-Schicklin L, Borel S, Pradezynski F, Diaz O, Deloire A, Azocar O, Baguet J, Le Breton M, Mangeot PE, Navratil V, Joubert PE, Flacher M, Vidalain PO, Andre P, Lotteau V, Biard-Piechaczyk M, Rabourdin-Combe C, Faure M (2011) IRGM is a common target of RNA viruses that subvert the autophagy network. PLoS Pathog 7:e1002422PubMedCrossRef Gregoire IP, Richetta C, Meyniel-Schicklin L, Borel S, Pradezynski F, Diaz O, Deloire A, Azocar O, Baguet J, Le Breton M, Mangeot PE, Navratil V, Joubert PE, Flacher M, Vidalain PO, Andre P, Lotteau V, Biard-Piechaczyk M, Rabourdin-Combe C, Faure M (2011) IRGM is a common target of RNA viruses that subvert the autophagy network. PLoS Pathog 7:e1002422PubMedCrossRef
Zurück zum Zitat Haas DW, Clough LA, Johnson BW, Harris VL, Spearman P, Wilkinson GR, Fletcher CV, Fiscus S, Raffanti S, Donlon R, McKinsey J, Nicotera J, Schmidt D, Shoup RE, Kates RE, Lloyd RM Jr, Larder B (2000) Evidence of a source of HIV type 1 within the central nervous system by ultraintensive sampling of cerebrospinal fluid and plasma. AIDS Res Hum Retrovir 16:1491–1502PubMedCrossRef Haas DW, Clough LA, Johnson BW, Harris VL, Spearman P, Wilkinson GR, Fletcher CV, Fiscus S, Raffanti S, Donlon R, McKinsey J, Nicotera J, Schmidt D, Shoup RE, Kates RE, Lloyd RM Jr, Larder B (2000) Evidence of a source of HIV type 1 within the central nervous system by ultraintensive sampling of cerebrospinal fluid and plasma. AIDS Res Hum Retrovir 16:1491–1502PubMedCrossRef
Zurück zum Zitat Hashimoto M, Sagara Y, Everall IP, Mallory M, Everson A, Langford D, Masliah E (2002) Fibroblast growth factor 1 regulates signaling via the GSK3{beta} pathway: implications for neuroprotection. J Biol Chem 277:32985–32991PubMedCrossRef Hashimoto M, Sagara Y, Everall IP, Mallory M, Everson A, Langford D, Masliah E (2002) Fibroblast growth factor 1 regulates signaling via the GSK3{beta} pathway: implications for neuroprotection. J Biol Chem 277:32985–32991PubMedCrossRef
Zurück zum Zitat Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, Rivera-Mindt M, Vigil OR, Taylor MJ, Collier AC, Marra CM, Gelman BB, McArthur JC, Morgello S, Simpson DM, McCutchan JA, Abramson I, Gamst A, Fennema-Notestine C, Jernigan TL, Wong J, Grant I (2010) HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology 75:2087–2096PubMedCrossRef Heaton RK, Clifford DB, Franklin DR Jr, Woods SP, Ake C, Vaida F, Ellis RJ, Letendre SL, Marcotte TD, Atkinson JH, Rivera-Mindt M, Vigil OR, Taylor MJ, Collier AC, Marra CM, Gelman BB, McArthur JC, Morgello S, Simpson DM, McCutchan JA, Abramson I, Gamst A, Fennema-Notestine C, Jernigan TL, Wong J, Grant I (2010) HIV-associated neurocognitive disorders persist in the era of potent antiretroviral therapy: CHARTER Study. Neurology 75:2087–2096PubMedCrossRef
Zurück zum Zitat Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, Leblanc S, Corkran SH, Duarte NA, Clifford DB, Woods SP, Collier AC, Marra CM, Morgello S, Mindt MR, Taylor MJ, Marcotte TD, Atkinson JH, Wolfson T, Gelman BB, McArthur JC, Simpson DM, Abramson I, Gamst A, Fennema-Notestine C, Jernigan TL, Wong J, Grant I (2011) HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J Neurovirol 17:3–16PubMedCrossRef Heaton RK, Franklin DR, Ellis RJ, McCutchan JA, Letendre SL, Leblanc S, Corkran SH, Duarte NA, Clifford DB, Woods SP, Collier AC, Marra CM, Morgello S, Mindt MR, Taylor MJ, Marcotte TD, Atkinson JH, Wolfson T, Gelman BB, McArthur JC, Simpson DM, Abramson I, Gamst A, Fennema-Notestine C, Jernigan TL, Wong J, Grant I (2011) HIV-associated neurocognitive disorders before and during the era of combination antiretroviral therapy: differences in rates, nature, and predictors. J Neurovirol 17:3–16PubMedCrossRef
Zurück zum Zitat Jaeger PA, Wyss-Coray T (2010) Beclin 1 complex in autophagy and Alzheimer disease. Arch Neurol 67:1181–1184PubMedCrossRef Jaeger PA, Wyss-Coray T (2010) Beclin 1 complex in autophagy and Alzheimer disease. Arch Neurol 67:1181–1184PubMedCrossRef
Zurück zum Zitat Joska JA, Gouse H, Paul RH, Stein DJ, Flisher AJ (2010) Does highly active antiretroviral therapy improve neurocognitive function? A systematic review. J Neurovirol 16:101–114PubMedCrossRef Joska JA, Gouse H, Paul RH, Stein DJ, Flisher AJ (2010) Does highly active antiretroviral therapy improve neurocognitive function? A systematic review. J Neurovirol 16:101–114PubMedCrossRef
Zurück zum Zitat Kaul M, Garden GA, Lipton SA (2001) Pathways to neuronal injury and apoptosis in HIV-associated dementia. Nature 410:988–994PubMedCrossRef Kaul M, Garden GA, Lipton SA (2001) Pathways to neuronal injury and apoptosis in HIV-associated dementia. Nature 410:988–994PubMedCrossRef
Zurück zum Zitat Khanlou N, Moore DJ, Chana G, Cherner M, Lazzaretto D, Dawes S, Grant I, Masliah E, Everall IP (2009) Increased frequency of alpha-synuclein in the substantia nigra in human immunodeficiency virus infection. J Neurovirol 15:131–138PubMedCrossRef Khanlou N, Moore DJ, Chana G, Cherner M, Lazzaretto D, Dawes S, Grant I, Masliah E, Everall IP (2009) Increased frequency of alpha-synuclein in the substantia nigra in human immunodeficiency virus infection. J Neurovirol 15:131–138PubMedCrossRef
Zurück zum Zitat Killian MS (2012) Dual role of autophagy in HIV-1 replication and pathogenesis. AIDS Res Ther 9:16PubMedCrossRef Killian MS (2012) Dual role of autophagy in HIV-1 replication and pathogenesis. AIDS Res Ther 9:16PubMedCrossRef
Zurück zum Zitat Kragh CL, Ubhi K, Wyss-Corey T, Masliah E (2012) Autophagy in dementias. Brain pathol 22:99–109PubMedCrossRef Kragh CL, Ubhi K, Wyss-Corey T, Masliah E (2012) Autophagy in dementias. Brain pathol 22:99–109PubMedCrossRef
Zurück zum Zitat Kyei GB, Dinkins C, Davis AS, Roberts E, Singh SB, Dong C, Wu L, Kominami E, Ueno T, Yamamoto A, Federico M, Panganiban A, Vergne I, Deretic V (2009) Autophagy pathway intersects with HIV-1 biosynthesis and regulates viral yields in macrophages. J Cell Biol 186:255–268PubMedCrossRef Kyei GB, Dinkins C, Davis AS, Roberts E, Singh SB, Dong C, Wu L, Kominami E, Ueno T, Yamamoto A, Federico M, Panganiban A, Vergne I, Deretic V (2009) Autophagy pathway intersects with HIV-1 biosynthesis and regulates viral yields in macrophages. J Cell Biol 186:255–268PubMedCrossRef
Zurück zum Zitat Lipton SA (1994) AIDS-related dementia and calcium homeostasis. Ann N Y Acad Sci 747:205–224PubMedCrossRef Lipton SA (1994) AIDS-related dementia and calcium homeostasis. Ann N Y Acad Sci 747:205–224PubMedCrossRef
Zurück zum Zitat Marr RA, Rockenstein E, Mukherjee A, Kindy MS, Hersh LB, Gage FH, Verma IM, Masliah E (2003) Neprilysin gene transfer reduces human amyloid pathology in transgenic mice. J Neurosci 23:1992–1996 Marr RA, Rockenstein E, Mukherjee A, Kindy MS, Hersh LB, Gage FH, Verma IM, Masliah E (2003) Neprilysin gene transfer reduces human amyloid pathology in transgenic mice. J Neurosci 23:1992–1996
Zurück zum Zitat Masliah E, Alford M, Adame A, Rockenstein E, Galasko D, Salmon D, Hansen LA, Thal LJ (2003) Abeta1-42 promotes cholinergic sprouting in patients with AD and Lewy body variant of AD. Neurology 61:206–211PubMedCrossRef Masliah E, Alford M, Adame A, Rockenstein E, Galasko D, Salmon D, Hansen LA, Thal LJ (2003) Abeta1-42 promotes cholinergic sprouting in patients with AD and Lewy body variant of AD. Neurology 61:206–211PubMedCrossRef
Zurück zum Zitat Naldini L, Blomer U, Gage FH, Trono D, Verma IM (1996a) Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci U S A 93:11382–11388PubMedCrossRef Naldini L, Blomer U, Gage FH, Trono D, Verma IM (1996a) Efficient transfer, integration, and sustained long-term expression of the transgene in adult rat brains injected with a lentiviral vector. Proc Natl Acad Sci U S A 93:11382–11388PubMedCrossRef
Zurück zum Zitat Naldini L, Blomer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM, Trono D (1996b) In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 272:263–267PubMedCrossRef Naldini L, Blomer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM, Trono D (1996b) In vivo gene delivery and stable transduction of nondividing cells by a lentiviral vector. Science 272:263–267PubMedCrossRef
Zurück zum Zitat Nath A (2002) Human immunodeficiency virus (HIV) proteins in neuropathogenesis of HIV dementia. J Infect Dis 186(Suppl 2):S193–S198PubMedCrossRef Nath A (2002) Human immunodeficiency virus (HIV) proteins in neuropathogenesis of HIV dementia. J Infect Dis 186(Suppl 2):S193–S198PubMedCrossRef
Zurück zum Zitat Nath A, Haughey NJ, Jones M, Anderson C, Bell JE, Geiger JD (2000) Synergistic neurotoxicity by human immunodeficiency virus proteins Tat and gp120: protection by memantine. Ann Neurol 47:186–194PubMedCrossRef Nath A, Haughey NJ, Jones M, Anderson C, Bell JE, Geiger JD (2000) Synergistic neurotoxicity by human immunodeficiency virus proteins Tat and gp120: protection by memantine. Ann Neurol 47:186–194PubMedCrossRef
Zurück zum Zitat Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, Cuervo AM (2005) Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol 64:113–122PubMed Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, Cuervo AM (2005) Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol 64:113–122PubMed
Zurück zum Zitat Norman JP, Perry SW, Reynolds HM, Kiebala M, De Mesy Bentley KL, Trejo M, Volsky DJ, Maggirwar SB, Dewhurst S, Masliah E, Gelbard HA (2008) HIV-1 Tat activates neuronal ryanodine receptors with rapid induction of the unfolded protein response and mitochondrial hyperpolarization. PLoS One 3:e3731PubMedCrossRef Norman JP, Perry SW, Reynolds HM, Kiebala M, De Mesy Bentley KL, Trejo M, Volsky DJ, Maggirwar SB, Dewhurst S, Masliah E, Gelbard HA (2008) HIV-1 Tat activates neuronal ryanodine receptors with rapid induction of the unfolded protein response and mitochondrial hyperpolarization. PLoS One 3:e3731PubMedCrossRef
Zurück zum Zitat Patrick C, Crews L, Desplats P, Dumaop W, Rockenstein E, Achim CL, Everall IP, Masliah E (2011) Increased CDK5 expression in HIV encephalitis contributes to neurodegeneration via Tau phosphorylation and is reversed with roscovitine. Am J Pathol 178:1646–1661PubMedCrossRef Patrick C, Crews L, Desplats P, Dumaop W, Rockenstein E, Achim CL, Everall IP, Masliah E (2011) Increased CDK5 expression in HIV encephalitis contributes to neurodegeneration via Tau phosphorylation and is reversed with roscovitine. Am J Pathol 178:1646–1661PubMedCrossRef
Zurück zum Zitat Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, Small S, Spencer B, Rockenstein E, Levine B, Wyss-Coray T (2008) The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 118:2190–2199PubMed Pickford F, Masliah E, Britschgi M, Lucin K, Narasimhan R, Jaeger PA, Small S, Spencer B, Rockenstein E, Levine B, Wyss-Coray T (2008) The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid beta accumulation in mice. J Clin Invest 118:2190–2199PubMed
Zurück zum Zitat Rockenstein E, Mante M, Alford M, Adame A, Crews L, Hashimoto M, Esposito L, Mucke L, Masliah E (2005a) High beta-secretase activity elicits neurodegeneration in transgenic mice despite reductions in amyloid-beta levels: implications for the treatment of Alzheimer disease. J Biol Chem 280:32957–32967PubMedCrossRef Rockenstein E, Mante M, Alford M, Adame A, Crews L, Hashimoto M, Esposito L, Mucke L, Masliah E (2005a) High beta-secretase activity elicits neurodegeneration in transgenic mice despite reductions in amyloid-beta levels: implications for the treatment of Alzheimer disease. J Biol Chem 280:32957–32967PubMedCrossRef
Zurück zum Zitat Rockenstein E, Schwach G, Ingolic E, Adame A, Crews L, Mante M, Pfragner R, Schreiner E, Windisch M, Masliah E (2005b) Lysosomal pathology associated with alpha-synuclein accumulation in transgenic models using an eGFP fusion protein. J Neurosci Res 80:247–259PubMedCrossRef Rockenstein E, Schwach G, Ingolic E, Adame A, Crews L, Mante M, Pfragner R, Schreiner E, Windisch M, Masliah E (2005b) Lysosomal pathology associated with alpha-synuclein accumulation in transgenic models using an eGFP fusion protein. J Neurosci Res 80:247–259PubMedCrossRef
Zurück zum Zitat Scott JC, Woods SP, Carey CL, Weber E, Bondi MW, Grant I (2011) Neurocognitive consequences of HIV infection in older adults: an evaluation of the “cortical” hypothesis. AIDS Behav 15:1187–1196PubMedCrossRef Scott JC, Woods SP, Carey CL, Weber E, Bondi MW, Grant I (2011) Neurocognitive consequences of HIV infection in older adults: an evaluation of the “cortical” hypothesis. AIDS Behav 15:1187–1196PubMedCrossRef
Zurück zum Zitat Smith G (2005). Statement of Senator Gordon H. Smith. Aging hearing: HIV over fifty, exploring the new threat. In: Senate Committee on Aging: Washington, DC Smith G (2005). Statement of Senator Gordon H. Smith. Aging hearing: HIV over fifty, exploring the new threat. In: Senate Committee on Aging: Washington, DC
Zurück zum Zitat Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C, Gindi R, Adame A, Wyss-Coray T, Masliah E (2009) Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson’s and Lewy body diseases. J Neurosci 29:13578–13588PubMedCrossRef Spencer B, Potkar R, Trejo M, Rockenstein E, Patrick C, Gindi R, Adame A, Wyss-Coray T, Masliah E (2009) Beclin 1 gene transfer activates autophagy and ameliorates the neurodegenerative pathology in alpha-synuclein models of Parkinson’s and Lewy body diseases. J Neurosci 29:13578–13588PubMedCrossRef
Zurück zum Zitat Tiscornia G, Singer O, Verma IM (2006) Design and cloning of lentiviral vectors expressing small interfering RNAs. Nat Protoc 1:234–240PubMedCrossRef Tiscornia G, Singer O, Verma IM (2006) Design and cloning of lentiviral vectors expressing small interfering RNAs. Nat Protoc 1:234–240PubMedCrossRef
Zurück zum Zitat Toggas SM, Masliah E, Rockenstein EM, Rall GF, Abraham CR, Mucke L (1994) Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice. Nature 367:188–193PubMedCrossRef Toggas SM, Masliah E, Rockenstein EM, Rall GF, Abraham CR, Mucke L (1994) Central nervous system damage produced by expression of the HIV-1 coat protein gp120 in transgenic mice. Nature 367:188–193PubMedCrossRef
Zurück zum Zitat Van Grol J, Subauste C, Andrade RM, Fujinaga K, Nelson J, Subauste CS (2010) HIV-1 inhibits autophagy in bystander macrophage/monocytic cells through Src-Akt and STAT3. PLoS One 5:e11733PubMedCrossRef Van Grol J, Subauste C, Andrade RM, Fujinaga K, Nelson J, Subauste CS (2010) HIV-1 inhibits autophagy in bystander macrophage/monocytic cells through Src-Akt and STAT3. PLoS One 5:e11733PubMedCrossRef
Zurück zum Zitat Watkins CC, Treisman GJ (2012) Neuropsychiatric complications of aging with HIV. J Neurovirol 18:277–290PubMedCrossRef Watkins CC, Treisman GJ (2012) Neuropsychiatric complications of aging with HIV. J Neurovirol 18:277–290PubMedCrossRef
Zurück zum Zitat Wiley C, Achim C (1994) HIV encephalitis is the pathologic correlate of dementia in AIDS. AnnNeurol 36:673–676 Wiley C, Achim C (1994) HIV encephalitis is the pathologic correlate of dementia in AIDS. AnnNeurol 36:673–676
Zurück zum Zitat Wiley CA, Baldwin M, Achim CL (1996) Expression of HIV regulatory and structural mRNA in the central nervous system. AIDS 10:843–847PubMedCrossRef Wiley CA, Baldwin M, Achim CL (1996) Expression of HIV regulatory and structural mRNA in the central nervous system. AIDS 10:843–847PubMedCrossRef
Zurück zum Zitat Zhou D, Masliah E, Spector SA (2011) Autophagy is increased in postmortem brains of persons with HIV-1-associated encephalitis. J Infect Dis 203:1647–1657PubMedCrossRef Zhou D, Masliah E, Spector SA (2011) Autophagy is increased in postmortem brains of persons with HIV-1-associated encephalitis. J Infect Dis 203:1647–1657PubMedCrossRef
Metadaten
Titel
Age-dependent molecular alterations in the autophagy pathway in HIVE patients and in a gp120 tg mouse model: reversal with beclin-1 gene transfer
verfasst von
Jerel Fields
Wilmar Dumaop
Edward Rockenstein
Michael Mante
Brian Spencer
Igor Grant
Ron Ellis
Scott Letendre
Christina Patrick
Anthony Adame
Eliezer Masliah
Publikationsdatum
01.02.2013
Verlag
Springer US
Erschienen in
Journal of NeuroVirology / Ausgabe 1/2013
Print ISSN: 1355-0284
Elektronische ISSN: 1538-2443
DOI
https://doi.org/10.1007/s13365-012-0145-7

Weitere Artikel der Ausgabe 1/2013

Journal of NeuroVirology 1/2013 Zur Ausgabe

Leitlinien kompakt für die Neurologie

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.

Update Neurologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.