Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
J Neurovirol ; 29(2): 156-166, 2023 04.
Article in English | MEDLINE | ID: mdl-36790601

ABSTRACT

HIV-associated neurocognitive disorders (HAND) persist in the era of antiretroviral therapy (ART). Thus, ART does not completely halt or reverse the pathological processes behind HAND. Adjuvant mitigating treatments are, therefore, prudent. Lithium treatment is known to promote neuronal brain-derived neurotrophic factors (BDNF). Lithium is also an inhibitor of glycogen synthase kinase-3 beta (GSK-3-ß). We analyzed biomarkers obtained from participants in a randomized placebo-controlled trial of lithium in ART-treated individuals with moderate or severe HAND. We assayed markers at baseline and 24 weeks across several pathways hypothesized to be affected by HIV, inflammation, or degeneration. Investigated biomarkers included dopamine, BDNF, neurofilament light chain, and CD8 + lymphocyte activation (CD38 + HLADR +). Alzheimer's Disease (AD) biomarkers included soluble amyloid precursor protein alpha and beta (sAPPα/ß), Aß38, 40, 42, and ten other biomarkers validated as predictors of mild cognitive impairment and progression in previous studies. These include apolipoprotein C3, pre-albumin, α1-acid glycoprotein, α1-antitrypsin, PEDF, CC4, ICAM-1, RANTES, clusterin, and cystatin c. We recruited 61 participants (placebo = 31; lithium = 30). The age baseline mean was 40 (± 8.35) years and the median CD4 + T-cell count was 498 (IQR: 389-651) cells/µL. Biomarker concentrations between groups did not differ at baseline. However, both groups' blood dopamine levels decreased significantly after 24 weeks (adj. p < 002). No other marker was significantly different between groups, and we concluded that lithium did not confer neuroprotection following 24 weeks of treatment. However, the study was limited in duration and sample size.


Subject(s)
HIV Infections , HIV , Humans , Adult , Middle Aged , Lithium/therapeutic use , Brain-Derived Neurotrophic Factor , Dopamine , Glycogen Synthase Kinase 3/therapeutic use , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/psychology , Biomarkers
3.
Sci Rep ; 12(1): 17221, 2022 10 14.
Article in English | MEDLINE | ID: mdl-36241688

ABSTRACT

For SARS-CoV-2, R0 calculations in the range of 2-3 dominate the literature, but much higher estimates have also been published. Because capacity for RT-PCR testing increased greatly in the early phase of the Covid-19 pandemic, R0 determinations based on these incidence values are subject to strong bias. We propose to use Covid-19-induced excess mortality to determine R0 regardless of RT-PCR testing capacity. We used data from the Robert Koch Institute (RKI) on the incidence of Covid cases, Covid-related deaths, number of RT-PCR tests performed, and excess mortality calculated from data from the Federal Statistical Office in Germany. We determined R0 using exponential growth estimates with a serial interval of 4.7 days. We used only datasets that were not yet under the influence of policy measures (e.g., lockdowns or school closures). The uncorrected R0 value for the spread of SARS-CoV-2 based on RT-PCR incidence data was 2.56 (95% CI 2.52-2.60) for Covid-19 cases and 2.03 (95% CI 1.96-2.10) for Covid-19-related deaths. However, because the number of RT-PCR tests increased by a growth factor of 1.381 during the same period, these R0 values must be corrected accordingly (R0corrected = R0uncorrected/1.381), yielding 1.86 for Covid-19 cases and 1.47 for Covid-19 deaths. The R0 value based on excess deaths was calculated to be 1.34 (95% CI 1.32-1.37). A sine-function-based adjustment for seasonal effects of 40% corresponds to a maximum value of R0January = 1.68 and a minimum value of R0July = 1.01. Our calculations show an R0 that is much lower than previously thought. This relatively low range of R0 fits very well with the observed seasonal pattern of infection across Europe in 2020 and 2021, including the emergence of more contagious escape variants such as delta or omicron. In general, our study shows that excess mortality can be used as a reliable surrogate to determine the R0 in pandemic situations.


Subject(s)
Basic Reproduction Number , COVID-19 , COVID-19/epidemiology , COVID-19/mortality , COVID-19 Nucleic Acid Testing , Germany/epidemiology , Humans , Pandemics , Reproducibility of Results , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification
4.
J Neural Transm (Vienna) ; 129(5-6): 545-555, 2022 06.
Article in English | MEDLINE | ID: mdl-35364741

ABSTRACT

Idiopathic Parkinson's disease (PD) is characterized by a progredient degeneration of the brain, starting at deep subcortical areas such as the dorsal motor nucleus of the glossopharyngeal and vagal nerves (DM) (stage 1), followed by the coeruleus-subcoeruleus complex; (stage 2), the substantia nigra (SN) (stage 3), the anteromedial temporal mesocortex (MC) (stage 4), high-order sensory association areas and prefrontal fields (HC) (stage 5) and finally first-order sensory association areas, premotor areas, as well as primary sensory and motor field (FC) (stage 6). Autoimmunity might play a role in PD pathogenesis. Here we analyzed whether anti-brain autoantibodies differentially recognize different human brain areas and identified autoantigens that correlate with the above-described dissemination of PD pathology in the brain. Brain tissue was obtained from deceased individuals with no history of neurological or psychiatric disease and no neuropathological abnormalities. Tissue homogenates from different brain regions (DM, SN, MC, HC, FC) were subjected to SDS-PAGE and Western blot. Blots were incubated with plasma samples from 30 PD patients and 30 control subjects and stained with anti-IgG antibodies to detect anti-brain autoantibodies. Signals were quantified. Prominent autoantigens were identified by 2D-gel-coupled mass spectrometry sequencing. Anti-brain autoantibodies are frequent and occur both in healthy controls and individuals with PD. Glial fibrillary acidic protein (GFAP) was identified as a prominent autoantigen recognized in all plasma samples. GFAP immunoreactivity was highest in DM areas and lowest in FC areas with no significant differences in anti-GFAP autoantibody titers between healthy controls and individuals with PD. The anti-GFAP autoimmunoreactivity of different brain areas correlates with the dissemination of histopathological neurodegeneration in PD. We hypothesize that GFAP autoantibodies are physiological but might be involved as a cofactor in PD pathogenesis secondary to a leakage of the blood-brain barrier.


Subject(s)
Parkinson Disease , Autoantibodies , Autoantigens/metabolism , Brain/metabolism , Glial Fibrillary Acidic Protein/metabolism , Humans , Parkinson Disease/metabolism
5.
Medicine (Baltimore) ; 95(46): e5401, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27861379

ABSTRACT

BACKGROUND: HIV-associated neurocognitive disorder (HAND) remains highly prevalent despite effective anti-retroviral therapy (ART). A number of adjunctive pharmacotherapies for HAND have been studied with disappointing results, but preliminary data suggest that lithium may provide clinical benefit. In addition, the low cost of lithium would facilitate access in low- and middle-income countries which carry the greatest burden of HIV. METHODS: Our objective was to evaluate the 24-week efficacy and safety of lithium in patients with moderate to severe HAND. Our primary efficacy endpoint was the change in Global Deficit Score (GDS) from baseline to 24 weeks, whereas our secondary endpoint was the change in proton magnetic resonance spectroscopy (H-MRS) brain metabolite concentrations. We conducted a 24-week randomized placebo-controlled trial of lithium as adjunctive pharmacotherapy. We enrolled participants with moderate to severe HAND, on ART for at least 6 months, with suppressed viral loads and attending public sector primary care clinics in Cape Town, South Africa. We randomized 66 participants to lithium (n = 32) or placebo (n = 34). Lithium or placebo was dosed 12-hourly and titrated to achieve the maintenance target plasma concentration of 0.6 to 1.0 mmol/L. Sham lithium concentrations were generated for participants receiving placebo. RESULTS: Totally 61 participants completed the study (lithium arm = 30; placebo arm = 31). Participants at enrolment had a mean age of 40 years and a median CD4+ T-cell count of 500 cells/µL. The median change in GDS between baseline and week 24 for the lithium and placebo arms were -0.57 (95% confidence interval [CI] -0.77, -0.32) and -0.56 (-0.69, -0.34) respectively, with a mean difference of -0.054 (95% CI -0.26, 0.15); P = 0.716. The improvement remained similar when analyzed according to age, severity of impairment, CD4+ count, time on ART, and ART regimen. Standard H-MRS metabolite concentrations were similar between the treatment arms. The study drug was well tolerated in both study arms. Six serious adverse events occurred, but none were considered related to the study drug. CONCLUSION: Adjunctive lithium pharmacotherapy in patients on ART with HAND was well tolerated but had no additional benefit on neurocognitive impairment.


Subject(s)
Brain , HIV Infections , Lithium Compounds , Neurocognitive Disorders , Adult , Brain/diagnostic imaging , Brain/metabolism , CD4 Lymphocyte Count/methods , Drug Monitoring/methods , Female , HIV Infections/complications , HIV Infections/diagnosis , HIV Infections/psychology , Humans , Lithium Compounds/administration & dosage , Lithium Compounds/pharmacokinetics , Male , Middle Aged , Neurocognitive Disorders/diagnosis , Neurocognitive Disorders/drug therapy , Neurocognitive Disorders/etiology , Neurocognitive Disorders/metabolism , Neuropsychological Tests , Proton Magnetic Resonance Spectroscopy/methods , Psychotropic Drugs/administration & dosage , Psychotropic Drugs/pharmacokinetics , Severity of Illness Index , Treatment Outcome
6.
J Neural Transm (Vienna) ; 123(6): 643-51, 2016 06.
Article in English | MEDLINE | ID: mdl-27173383

ABSTRACT

While cortical thinning has been associated with HIV infection, it is unclear whether this reflects a direct effect of the virus, whether it is related to disruption of subcortical function or whether it is better explained by epiphenomena, such as drug abuse or comorbid medical conditions. The present study investigated the relationship between cortical thickness and subcortical function in HIV+ patients. Specifically, we examined the relationship between prefrontal cortical thickness and striatal function. Twenty-three largely treatment naïve, non-substance abusing HIV+ participants and 19 healthy controls matched for age, gender, and educational status were included. Cortical morphometry was performed using FreeSurfer software analysis. Striatal function was measured during an fMRI stop-signal anticipation task known to engage the striatum. Any cortical regions showing significant thinning were entered as dependent variables into a single linear regression model which included subcortical function, age, CD4 count, and a measure of global cognitive performance as independent predictors. The only cortical region that was significantly reduced after correction for multiple comparisons was the right superior frontal gyrus. Striatal activity was found to independently predict superior frontal gyral cortical thickness. While cortical thinning in HIV infection is likely multifactorial, viral induced subcortical dysfunction appears to play a role.


Subject(s)
Anticipation, Psychological/physiology , HIV Infections , Inhibition, Psychological , Neostriatum/physiopathology , Prefrontal Cortex/pathology , Adult , Female , HIV Infections/complications , HIV Infections/pathology , HIV Infections/physiopathology , Humans , Magnetic Resonance Imaging , Male , Neostriatum/diagnostic imaging , Prefrontal Cortex/diagnostic imaging , Prefrontal Cortex/physiopathology , Young Adult
7.
PLoS One ; 11(1): e0146678, 2016.
Article in English | MEDLINE | ID: mdl-26812052

ABSTRACT

BACKGROUND: HIV-disease progression correlates with immune activation. Here we investigated whether corticosteroid treatment can attenuate HIV disease progression in antiretroviral-untreated patients. METHODS: Double-blind, placebo-controlled randomized clinical trial including 326 HIV-patients in a resource-limited setting in Tanzania (clinicaltrials.gov NCT01299948). Inclusion criteria were a CD4 count above 300 cells/µl, the absence of AIDS-defining symptoms and an ART-naïve therapy status. Study participants received 5 mg prednisolone per day or placebo for 2 years. Primary endpoint was time to progression to an AIDS-defining condition or to a CD4-count below 200 cells/µl. RESULTS: No significant change in progression towards the primary endpoint was observed in the intent-to-treat (ITT) analysis (19 cases with prednisolone versus 28 cases with placebo, p = 0.1407). In a per-protocol (PP)-analysis, 13 versus 24 study participants progressed to the primary study endpoint (p = 0.0741). Secondary endpoints: Prednisolone-treatment decreased immune activation (sCD14, suPAR, CD38/HLA-DR/CD8+) and increased CD4-counts (+77.42 ± 5.70 cells/µl compared to -37.42 ± 10.77 cells/µl under placebo, p < 0.0001). Treatment with prednisolone was associated with a 3.2-fold increase in HIV viral load (p < 0.0001). In a post-hoc analysis stratifying for sex, females treated with prednisolone progressed significantly slower to the primary study endpoint than females treated with placebo (ITT-analysis: 11 versus 21 cases, p = 0.0567; PP-analysis: 5 versus 18 cases, p = 0.0051): No changes in disease progression were observed in men. CONCLUSIONS: This study could not detect any significant effects of prednisolone on disease progression in antiretroviral-untreated HIV infection within the intent-to-treat population. However, significant effects were observed on CD4 counts, immune activation and HIV viral load. This study contributes to a better understanding of the role of immune activation in the pathogenesis of HIV infection. TRIAL REGISTRATION: ClinicalTrials.gov NCT01299948.


Subject(s)
Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Immunologic Factors/pharmacology , Prednisolone/pharmacology , Adult , Anti-HIV Agents/pharmacology , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , Disease Progression , Double-Blind Method , Female , HIV Infections/epidemiology , Humans , Immunologic Factors/therapeutic use , Kaplan-Meier Estimate , Male , Medication Adherence , Prednisolone/therapeutic use , Treatment Outcome , Viral Load
8.
J Int Neuropsychol Soc ; 21(9): 722-31, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26435417

ABSTRACT

The aim of the present study was to investigate the effect of HIV infection on cortical and subcortical regions of the frontal-striatal system involved in the inhibition of voluntary movement. Functional MRI (fMRI) studies suggest that human immunodeficiency virus (HIV) infection is associated with frontostriatal dysfunction. While frontostriatal systems play a key role in behavioral inhibition, there are to our knowledge no fMRI studies investigating the potential impact of HIV on systems involved during the inhibition of voluntary movement. A total of 17 combined antiretroviral therapy (cART) naïve HIV+ participants as well as 18 age, gender, ethnic, education matched healthy controls performed a modified version of the stop-signal paradigm. This paradigm assessed behavior as well as functional brain activity associated with motor execution, reactive inhibition (outright stopping) and proactive inhibition (anticipatory response slowing before stopping). HIV+ participants showed significantly slower responses during motor execution compared to healthy controls, whereas they had normal proactive response slowing. Putamen hypoactivation was evident in the HIV+ participants based on successful stopping, indicating subcortical dysfunction during reactive inhibition. HIV+ participants showed normal cortical functioning during proactive inhibition. Our data provide evidence that HIV infection is associated with subcortical dysfunction during reactive inhibition, accompanied by relatively normal higher cortical functioning during proactive inhibition. This suggests that HIV infection may primarily involve basic striatal-mediated control processes in cART naïve participants.


Subject(s)
Brain/physiopathology , Corpus Striatum/physiopathology , HIV Infections/physiopathology , Adult , Case-Control Studies , Female , Functional Neuroimaging , Humans , Inhibition, Psychological , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Psychomotor Performance/physiology , Reaction Time
9.
AIDS ; 29(11): 1335-43, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26091294

ABSTRACT

OBJECTIVE: Functional MRI has thus far demonstrated that HIV has an impact on frontal-striatal systems involved in executive functioning. The potential impact of HIV on frontal-striatal systems involved in reward processing has yet to be examined by functional MRI. This study therefore aims to investigate the effects of HIV infection on reward processing by examining the function of the ventral-striatal reward system during a monetary incentive delay task. DESIGN: This is a cross-sectional case-control study. METHODS: Eighteen combined antiretroviral therapy-naive HIV-positive (HIV+) participants, as well as 16 matched healthy controls, performed a monetary incentive delay task. This paradigm assesses behaviour as well as functional brain activity-associated reward anticipation and reward outcome. RESULTS: HIV+ participants showed a general decrease in activation associated with both neutral as well as potentially rewarding cues in their ventral striatum. We found normal activity related to reward outcome in the orbito-frontal cortex. Despite HIV+ participants' reaction times being significantly slower when independently measured from the reward paradigm, this performance deficit normalized during the performance of the reward task. CONCLUSION: HIV caused a decrease in activity during cue processing in the ventral striatum, with normal cortical functioning during reward outcome processing. Our results therefore suggest that HIV not only has an impact on fronto-striatal systems involved in executive functioning, but also has a direct impact on the function of the ventral-striatal reward system.


Subject(s)
Frontal Lobe/physiopathology , HIV Seropositivity/physiopathology , HIV Seropositivity/psychology , Ventral Striatum/physiopathology , Adult , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Magnetic Resonance Imaging , Male , Neuropsychological Tests , Reaction Time , Regression Analysis , Young Adult
10.
AIDS Res Ther ; 11: 17, 2014.
Article in English | MEDLINE | ID: mdl-25013451

ABSTRACT

BACKGROUND: Opioids may have effects on susceptibility to HIV-infection, viral replication and disease progression. Injecting drug users (IDU), as well as anyone receiving opioids for anesthesia and analgesia may suffer the clinical consequences of such interactions. There is conflicting data between in vitro experiments showing an enhancing effect of opioids on HIV replication and clinical data, mostly showing no such effect. For clarification we studied the effects of the opioids heroin and morphine on HIV replication in cultured CD4-positive T cells at several concentrations and we related the observed effects with the relevant reached plasma concentrations found in IDUs. METHODS: Latently-infected ACH-2 T lymphoblasts were incubated with different concentrations of morphine and heroine. Reactivation of HIV was assessed by intracellular staining of viral Gag p24 protein and subsequent flow cytometric quantification of p24-positive cells. The influence of the opioid antagonist naloxone and the antioxidants N-acetyl-cysteine (NAC) and glutathione (GSH) on HIV reactivation was determined. Cell viability was investigated by 7-AAD staining and flow cytometric quantification. RESULTS: Morphine and heroine triggered reactivation of HIV replication in ACH-2 cells in a dose-dependent manner at concentrations above 1 mM (EC50 morphine 2.82 mM; EC50 morphine 1.96 mM). Naloxone did not interfere with heroine-mediated HIV reactivation, even at high concentrations (1 mM). Opioids also triggered necrotic cell death at similar concentrations at which HIV reactivation was observed. Both opioid-mediated reactivation of HIV and opioid-triggered cell death could be inhibited by the antioxidants GSH and NAC. CONCLUSIONS: Opioids reactivate HIV in vitro but at concentrations that are far above the plasma levels of analgesic regimes or drug concentrations found in IDUs. HIV reactivation was mediated by effects unrelated to opioid-receptor activation and was tightly linked to the cytotoxic activity of the substances at millimolar concentrations, suggesting that opioid-mediated reactivation of HIV was due to accompanying effects of cellular necrosis such as activation of reactive oxygen species and NF-κB.

11.
Atten Defic Hyperact Disord ; 6(3): 203-9, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24696011

ABSTRACT

The dopamine transporter (DAT) is a functional element of the dopaminergic synapse in the brain. Its primary role is the regulation of dopamine (DA) availability by forward and reverse transport of DA from and to the synaptic cleft by which extracellular DA concentrations are being regulated. The DAT gene and especially the DAT 10/10 genotype have been intensively discussed as a candidate for several neuropsychiatric disorders including attention-deficit-hyperactivity disorder (ADHD). We found recently that the DAT 10/10 genotype is associated with increased levels of CSF DA and is present more frequently in HIV-infected individuals than in uninfected subjects, suggesting that personality traits related to this polymorphism may increase the risk of acquisition of HIV. In this article, we review studies on the DAT 10/10 genotype and the association with ADHD and its endophenotypes, express concerns on the reported DA neurochemistry in ADHD and discuss consequences of the DAT 10/10 genotype on the epidemiology of HIV infection.


Subject(s)
Attention Deficit Disorder with Hyperactivity/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , HIV Infections/genetics , Alleles , Genetic Predisposition to Disease/genetics , Genotype , Humans , Risk Factors
12.
AIDS ; 28(6): 803-11, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24300546

ABSTRACT

Functional MRI studies investigating the impact of HIV on the brain have implicated the involvement of fronto-striatal circuitry. However, to date there is no review and meta-analysis of this work. We systematically reviewed the literature and performed a meta-analysis of functional magnetic resonance imaging (fMRI) studies in HIV-infected individuals using a well validated tool recently developed for use in fMRI, 'GingerALE'. Twenty-one studies (468 HIV+, 270 HIV- controls) were qualitatively reviewed, of which six (105 HIV+, 102 controls) utilized fMRI paradigms engaging the fronto-striatal-parietal network, making a quantitative analysis possible. Our meta-analysis revealed consistent functional differences in the left inferior frontal gyrus and caudate nucleus between infected participants and controls across these studies. This fronto-striatal dysfunction was qualitatively related to cognitive impairment, disease progression and treatment effects. Although further work needs to be done to further delineate the potentially confounding influence of substance abuse and HIV-related comorbidities, as well as HIV's effect on functional haemodynamic vascular coupling, these findings indicate that further investigation of the fronto-striatal sub-networks in HIV-infected patients is warranted.


Subject(s)
AIDS Dementia Complex/pathology , AIDS Dementia Complex/physiopathology , Frontal Lobe/pathology , Frontal Lobe/physiopathology , HIV Infections/complications , Visual Cortex/pathology , Visual Cortex/physiopathology , Humans , Magnetic Resonance Imaging
13.
J Neural Transm (Vienna) ; 120(10): 1411-9, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24057505

ABSTRACT

Dysfunction of dopaminergic neurotransmission has been implicated in HIV infection. We showed previously increased dopamine (DA) levels in CSF of therapy-naïve HIV patients and an inverse correlation between CSF DA and CD4 counts in the periphery, suggesting adverse effects of high levels of DA on HIV infection. In the current study including a total of 167 HIV-positive and negative donors from Germany and South Africa (SA), we investigated the mechanistic background for the increase of CSF DA in HIV individuals. Interestingly, we found that the DAT 10/10-repeat allele is present more frequently within HIV individuals than in uninfected subjects. Logistic regression analysis adjusted for gender and ethnicity showed an odds ratio for HIV infection in DAT 10/10 allele carriers of 3.93 (95% CI 1.72-8.96; p = 0.001, Fishers exact test). 42.6% HIV-infected patients harbored the DAT 10/10 allele compared to only 10.5% uninfected DAT 10/10 carriers in SA (odds ratio 6.31), whereas 68.1 versus 40.9%, respectively, in Germany (odds ratio 3.08). Subjects homozygous for the 10-repeat allele had higher amounts of CSF DA and reduced DAT mRNA expression but similar disease severity compared with those carrying other DAT genotypes. These intriguing and novel findings show the mutual interaction between DA and HIV, suggesting caution in the interpretation of CNS DA alterations in HIV infection solely as a secondary phenomenon to the virus and open the door for larger studies investigating consequences of the DAT functional polymorphism on HIV epidemiology and progression of disease.


Subject(s)
Dopamine Plasma Membrane Transport Proteins/genetics , Dopamine/cerebrospinal fluid , HIV Infections/cerebrospinal fluid , HIV Infections/genetics , AIDS Dementia Complex/cerebrospinal fluid , AIDS Dementia Complex/genetics , Adult , Aged , Alleles , Female , Genotype , Humans , Male , Middle Aged , Odds Ratio , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Young Adult
14.
J Neurovirol ; 18(3): 157-61, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22528475

ABSTRACT

HIV-associated dementia and its precursors are frequently observed complications of HIV infection, even in the presence of combination antiretroviral treatment (cART). The development, surveillance and treatment of this condition are still not completely understood. Cytokines, as immunological transmitters, may be one key to gaining a deeper understanding of the disease. A total of 33 HIV-positive male patients were evaluated by neuropsychological testing, lumbar and venous puncture, neuroimaging and neurological examination. The cytokine content in the CSF (cerebrospinal fluid) was examined by a solid-phase protein array. The Digit-Symbol Test, contraction time analysis, Rey-Osterrieth Figure and Grooved-Pegboard Test showed inferior results in the presence of an inflammatory CSF environment, whereas neuroprotective or anti-inflammatory conditions were correlated to better results in contraction time analysis. Higher CSF levels of cytokines were independently correlated with the duration of HIV infection. The study showed a correlation of cytokine levels in the CSF of HIV patients with test results of their neuropsychological functioning. The effect was pronounced with regard to the more complex executive tasks. Determining CSF cytokine levels may be a useful supplement to the assessment of HIV patients and contribute helpful information to predict neurocognitive performance. Therapeutic strategies to ameliorate a negative impact of an altered cytokine milieu may aid in slowing the evolution of neurocognitive dysfunction.


Subject(s)
Cytokines/cerebrospinal fluid , HIV Infections/cerebrospinal fluid , HIV Infections/psychology , HIV-1/physiology , Adult , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/therapeutic use , Cognition , HIV Infections/blood , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuropsychological Tests , Prospective Studies , Protein Array Analysis , Task Performance and Analysis , Time Factors , Viral Load
15.
BMC Infect Dis ; 12: 14, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22264238

ABSTRACT

BACKGROUND: HIV-associated general immune activation is a strong predictor for HIV disease progression, suggesting that chronic immune activation may drive HIV pathogenesis. Consequently, immunomodulating agents may decelerate HIV disease progression. METHODS: In an observational study, we determined immune activation in HIV patients receiving low-dose (5 mg/day) prednisolone with or without highly-active antiretroviral therapy (HAART) compared to patients without prednisolone treatment. Lymphocyte activation was determined by flow cytometry detecting expression of CD38 on CD8(+) T cells. The monocyte activation markers sCD14 and LPS binding protein (LBP) as well as inflammation markers soluble urokinase plasminogen activated receptor (suPAR) and sCD40L were determined from plasma by ELISA. RESULTS: CD38-expression on CD8+ T lymphocytes was significantly lower in prednisolone-treated patients compared to untreated patients (median 55.40% [percentile range 48.76-67.70] versus 73.34% [65.21-78.92], p = 0.0011, Mann-Whitney test). Similarly, we detected lower levels of sCD14 (3.6 µg/ml [2.78-5.12] vs. 6.11 µg/ml [4.58-7.70]; p = 0.0048), LBP (2.18 ng/ml [1.59-2.87] vs. 3.45 ng/ml [1.84-5.03]; p = 0.0386), suPAR antigen (2.17 µg/ml [1.65-2.81] vs. 2.56 µg/ml [2.24-4.26]; p = 0.0351) and a trend towards lower levels of sCD40L (2.70 pg/ml [1.90-4.00] vs. 3.60 pg/ml [2.95-5.30]; p = 0.0782). Viral load in both groups was similar (0.8 × 105 ng/ml [0.2-42.4 × 105] vs. 1.1 × 105 [0.5-12.2 × 105]; p = 0.3806). No effects attributable to prednisolone were observed when patients receiving HAART in combination with prednisolone were compared to patients who received HAART alone. CONCLUSIONS: Patients treated with low-dose prednisolone display significantly lower general immune activation than untreated patients. Further longitudinal studies are required to assess whether treatment with low-dose prednisolone translates into differences in HIV disease progression.


Subject(s)
HIV Infections/drug therapy , HIV Infections/immunology , Immunologic Factors/administration & dosage , Lymphocyte Activation/drug effects , Prednisolone/administration & dosage , ADP-ribosyl Cyclase 1/analysis , Acute-Phase Proteins/analysis , Adult , Anti-HIV Agents/administration & dosage , CD40 Ligand/blood , CD8-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Carrier Proteins/analysis , Female , Flow Cytometry , Humans , Immunophenotyping , Lipopolysaccharide Receptors/analysis , Male , Membrane Glycoproteins/analysis , Middle Aged , Monocytes/chemistry , Monocytes/immunology , Receptors, Urokinase Plasminogen Activator/blood
16.
BMC Neurosci ; 12: 116, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-22085464

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is characterized at the cellular level by a destruction of neuromelanin (NM)-containing dopaminergic cells and a profound reduction in striatal dopamine. It has been shown recently that anti-melanin antibodies are increased in sera of Parkinson patients, suggesting that NM may act as an autoantigen. In this study we tested whether NM is being recognized by dendritic cells (DCs), the major cell type for inducing T- and B-cell responses in vivo. This recognition of NM by DCs is a prerequisite to trigger an adaptive autoimmune response directed against NM-associated structures. RESULTS: Murine DCs were treated with NM of substantia nigra (SN) from human subjects or with synthetic dopamine melanin (DAM). DCs effectively phagocytized NM and subsequently developed a mature phenotype (CD86(high)/MHCII(high)). NM-activated DCs secreted the proinflammatory cytokines IL-6 and TNF-α. In addition, they potently triggered T cell proliferation in a mixed lymphocyte reaction, showing that DC activation was functional to induce a primary T cell response. In contrast, DAM, which lacks the protein and lipid components of NM but mimics the dopamine-melanin backbone of NM, had only very little effect on DC phenotype and function. CONCLUSIONS: NM is recognized by DCs in vitro and triggers their maturation. If operative in vivo, this would allow the DC-mediated transport and presentation of SN antigens to the adaptive immune system, leading to autoimmmunity in susceptible individuals. Our data provide a rationale for an autoimmune-based pathomechanism of PD with NM as the initial trigger.


Subject(s)
Autoimmune Diseases of the Nervous System/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Melanins/pharmacology , Parkinson Disease/immunology , Parkinson Disease/pathology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Autoimmune Diseases of the Nervous System/pathology , Cells, Cultured , Dendritic Cells/drug effects , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
17.
PLoS One ; 6(8): e23091, 2011.
Article in English | MEDLINE | ID: mdl-21886779

ABSTRACT

BACKGROUND: The World Health Organization (WHO) has recommended guidelines for a HIV drug resistance (HIVDR) survey for resource-limited countries. Eligibility criteria for patients include age below 25 years in order to focus on the prevalence of transmitted HIVDR (tHIVDR) in newly-infected individuals. Most of the participating sites across Africa have so far reported tHIVDR prevalences of below 5%. In this study we investigated whether the rate of HIVDR in patients <25 years is representative for HIVDR in the rest of the therapy-naïve population. METHODS AND FINDINGS: HIVDR was determined in 88 sequentially enrolled ART-naïve patients from Mwanza, Tanzania (mean age 35.4 years). Twenty patients were aged <25 years and 68 patients were aged 25-63 years. The frequency of HIVDR in the study population was 14.8% (95%; CI 0.072-0.223) and independent of NVP-resistance induced by prevention of mother-to-child transmission programs. Patients >25 years had a significantly higher HIVDR frequency than younger patients (19.1%; 95% CI 0.095-0.28) versus 0%, P = 0.0344). In 2 out of the 16 patients with HIVDR we found traces of antiretrovirals (ARVs) in plasma. CONCLUSIONS: ART-naïve patients aged over 25 years exhibited significantly higher HIVDR than younger patients. Detection of traces of ARVs in individuals with HIVDR suggests that besides transmission, undisclosed misuse of ARVs may constitute a significant factor in the generation of the observed high HIVDR rate. The current WHO tHIVDR survey that is solely focused on the transmission of HIVDR and that excludes patients over 25 years of age may therefore result in substantial underestimation of the prevalence of HIVDR in the therapy-naïve population. Similar studies should be performed also in other areas to test whether the so far reported optimistic picture of low HIVDR prevalence in young individuals is really representative for the rest of the ART-naïve HIV-infected population.


Subject(s)
Anti-HIV Agents/therapeutic use , Drug Resistance, Viral , Eligibility Determination/statistics & numerical data , HIV Infections/drug therapy , Health Care Surveys/statistics & numerical data , World Health Organization , Adult , Aging , Cohort Studies , Demography , Drug Monitoring , Female , HIV Infections/epidemiology , HIV Infections/transmission , HIV Infections/virology , HIV-1/classification , HIV-1/genetics , Humans , Infectious Disease Transmission, Vertical/prevention & control , Infectious Disease Transmission, Vertical/statistics & numerical data , Male , Middle Aged , Molecular Sequence Data , Mutation/genetics , Phylogeny , Tanzania/epidemiology , Time Factors , Treatment Outcome , Young Adult
18.
Cytometry A ; 79(6): 436-45, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21425237

ABSTRACT

Haematopoietic immune cell populations play an important role in the pathogenesis of numerous neurological disorders. To better understand the function of resident mononuclear phagocytes and migrating leukocytes in the central nervous system (CNS), the definition of these populations in healthy individuals is crucial. Therefore, the composition of CNS-associated leukocytes, isolated from macaque brain tissue, was assessed using multicolor flow cytometry. We established a combination of antibodies directed against nine different antigens that enabled a precise classification of all major immune cell populations in a single tube. Macrophages, dendritic cells (DCs), B and T lymphocytes, and natural killer (NK) cells were differentiated in CNS and peripheral blood. Additionally, microglia cells were detected in the brain. Using this antibody combination also allowed the discrimination of functionally different subsets among the distinct immunocyte populations, for example, CD8 positive cytotoxic T lymphocytes. About 95% of the leukocytes in the brain are microglia cells. Two additional myeloid cell populations, CD14 positive macrophages and CD11c-positive DCs, were also identified. In contrast to blood, where macrophages are more abundant, DCs outnumbered macrophages in the brain. Among lymphocytes, proportions of CD20 positive B lymphocytes were decreased, and T lymphocytes as well as NK cells were increased in brain compared to blood. Significant changes were also detected for macrophage and T-cell subpopulations. The nonexclusive expression of certain surface makers on different cell populations demanded a simultaneous classification of all intrathecal immune cells. Knowing their exact composition offers new insights on interaction and regulation in inflammatory processes and will be instrumental to monitor alterations in the course of neurological diseases.


Subject(s)
B-Lymphocyte Subsets/cytology , Brain/cytology , Dendritic Cells/cytology , Flow Cytometry/methods , Ganglia/cytology , Killer Cells, Natural/cytology , Macrophages/cytology , T-Lymphocyte Subsets/cytology , Animals , Antigens, CD , B-Lymphocyte Subsets/immunology , Brain/immunology , Cell Differentiation , Dendritic Cells/immunology , Female , Ganglia/immunology , Killer Cells, Natural/immunology , Lymphocyte Count , Macaca , Macrophages/immunology , Male , T-Lymphocyte Subsets/immunology
19.
J Neurovirol ; 15(5-6): 390-400, 2009 Sep.
Article in English | MEDLINE | ID: mdl-20001608

ABSTRACT

The objective of this study was to evaluate immune cytokine expression in cerebrospinal fluid (CSF) of patients with human immunodeficiency virus-1 (HIV-1)-associated dementia (HAD) using a novel cytokine array assay. HIV-1 induces a condition resembling classical subcortical dementia, known as HAD. The immune mechanisms contributing to HAD have not been elucidated. Cytokine expression in CSF was determined by solid-phase protein array in 33 neurologically asymptomatic HIV-positive male patients and were compared to levels in non-HIV controls and patients with HAD. Neurological examinations and lumbar and venous punctures were conducted in all patients and controls. Interleukin (IL)-1, IL-4, and IL-10, were up-regulated in all treated acquired immunodeficiency syndrome (AIDS) patients independent of neurological status compared to controls. In contrast, interferon gamma (IFN-gamma), IL-1alpha, IL-15, and tumor necrosis factor alpha (TNF-alpha) were highly expressed in patients with HAD compared to undemented HIV-positive patients. These results show that solid-phase protein array can detect immunological changes in patients infected with HIV. Cytokine expression levels differ in different disease stages and in patients on different treatment paradigms. Pending further validation on a larger number of patients, this method may be a useful tool in CSF diagnostics and the longitudinal evaluation of patient with HAD.


Subject(s)
AIDS Dementia Complex/cerebrospinal fluid , AIDS Dementia Complex/immunology , Cytokines/cerebrospinal fluid , HIV-1 , Inflammation Mediators/cerebrospinal fluid , Protein Array Analysis/methods , AIDS Dementia Complex/diagnosis , AIDS Dementia Complex/epidemiology , Adult , Biomarkers/cerebrospinal fluid , Humans , Interferon-gamma/cerebrospinal fluid , Interleukin-10/cerebrospinal fluid , Interleukin-15/cerebrospinal fluid , Interleukin-1alpha/cerebrospinal fluid , Interleukin-4/cerebrospinal fluid , Male , Middle Aged , Neuropsychological Tests , Protein Array Analysis/standards , Reproducibility of Results , Risk Factors , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/virology , Tumor Necrosis Factor-alpha/cerebrospinal fluid
20.
Neuropsychopharmacology ; 33(9): 2228-36, 2008 Aug.
Article in English | MEDLINE | ID: mdl-17971830

ABSTRACT

N-methyl-D-aspartate (NMDA) receptor activation is involved in the pathogenetic cascades of neurodegenerative disorders including human immunodeficiency virus (HIV) dementia. Memantine, an uncompetitive NMDA receptor antagonist, which has been recently approved for the treatment of Alzheimer's disease, is being discussed as a potential adjunctive therapeutic substance for HIV dementia. We used simian immunodeficiency virus-infected rhesus macaques to assess the effects of memantine on brain dysfunction and brain pathology within 3-5 months after initial infection during early asymptomatic stage of disease. We had shown previously that within this time frame, marked changes were evident in the dopaminergic systems. Memantine was administered two weeks post infection, at peak viremia, in order to prevent early NMDA receptor activation due to immune mediators. We found that memantine prevented onset of dopamine deficits in the brains of SIV-infected macaques, without affecting early brain pathology or peripheral course of infection. Memantine specifically upregulated mRNA and protein expression of the neurotrophic factor brain-derived neurotrophic factor (BDNF), suggesting that the protective effect of memantine on dopamine function may be mechanistically remote from NMDA receptor antagonism. This novel pharmacological action of memantine may also be relevant for other neurodegenerative disorders and supports the involvement of neurotrophic factors in adult brain neuroprotection.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Dopamine/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Memantine/pharmacology , Simian Acquired Immunodeficiency Syndrome/metabolism , Up-Regulation/drug effects , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain/virology , Brain-Derived Neurotrophic Factor/genetics , Disease Models, Animal , Excitatory Amino Acid Antagonists/therapeutic use , Homovanillic Acid/metabolism , Humans , Macaca mulatta , Memantine/therapeutic use , Oligonucleotide Array Sequence Analysis/methods , RNA, Messenger/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Viral Load/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...