Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Brain Inj ; 35(5): 520-529, 2021 04 16.
Article in English | MEDLINE | ID: mdl-33587672

ABSTRACT

Purpose: SARS-CoV-2 infection can cause the coronavirus disease (COVID), ranging from flu-like symptoms to interstitial pneumonia. Mortality is high in COVID pneumonia and it is the highest among the frailest. COVID could be particularly serious in patients with severe acquired brain injury (SABI), such as those with a disorder of consciousness. We here describe a cohort of patients with a disorder of consciousness exposed to SARS-CoV-2 early after their SABI.Materials and methods: The full cohort of 11 patients with SABI hospitalized in March 2020 in the IRCCS Fondazione Don Gnocchi rehabilitation (Milan, Italy) was recruited. Participants received SARS-CoV-2 testing and different clinical and laboratory data were collected.Results: Six patients contracted SARS-CoV-2 and four of them developed the COVID. Of these, one patient had ground-glass opacities on the chest CT scan, while the remaining three developed consolidations. No patient died and the overall respiratory involvement was mild, requiring in the worst cases low-flow oxygen.Conclusions: Here we report the clinical course of a cohort of patients with SABI exposed to SARS-CoV-2. The infection spread among patients and caused COVID in some of them. Unexpectedly, COVID was moderate, caused at most mild respiratory distress and did not result in fatalities.


Subject(s)
Brain Injuries/complications , COVID-19/complications , Consciousness Disorders/complications , Brain Injuries/virology , COVID-19 Testing , Consciousness Disorders/virology , Humans , Italy
2.
Clin Microbiol Infect ; 27(8): 1131-1136, 2021 Aug.
Article in English | MEDLINE | ID: mdl-32979577

ABSTRACT

OBJECTIVES: The aim was to investigate the correlation between biomarkers of brain injury and long-term neurocognitive outcome, and the interplay with intrathecal inflammation and neuronal autoimmunity, in patients with herpes simplex encephalitis (HSE). METHODS: A total of 53 adult/adolescent HSE patients were included from a prospective cohort in a randomized placebo-controlled trial investigating the effect of a 3-month follow-up treatment with valaciclovir. Study subjects underwent repeated serum/cerebrospinal fluid (CSF) sampling and brain magnetic resonance imaging in the first 3 months along with cognitive assessment using the Mattis Dementia Rating Scale (MDRS) at 24 months. CSF samples were analysed for biomarkers of brain injury, inflammation and synaptic autoimmunity. The predefined primary analysis was the correlation between peak CSF neurofilament protein (NFL), a biomarker of neuronal damage, and MDRS at 24 months. RESULTS: Impaired cognitive performance significantly correlated with NFL levels (rho = -0.36, p = 0.020). Development of IgG anti-N-methyl-D-aspartate receptor (NDMAR) antibodies was associated with a broad and prolonged proinflammatory CSF response. In a linear regression model, lower MDRS at 24 months was associated with previous development of IgG anti-N-methyl-D-aspartate receptor (NMDAR) (beta = -0.6249, p = 0.024) and age (z-score beta = -0.2784, p = 0.024), but not CSF NFL, which however significantly correlated with subsequent NMDAR autoimmunization (p = 0.006). DISCUSSION: Our findings show that NFL levels are predictive of long-term neurocognitive outcome in HSE, and suggest a causative chain of events where brain tissue damage increases the risk of NMDAR autoimmunisation and subsequent prolongation of CSF inflammation. The data provides guidance for a future intervention study of immunosuppressive therapy administered in the recovery phase of HSE.


Subject(s)
Autoimmunity , Brain Injuries/cerebrospinal fluid , Encephalitis, Herpes Simplex , Inflammation/cerebrospinal fluid , Neurocognitive Disorders/virology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers/cerebrospinal fluid , Brain Injuries/virology , Encephalitis, Herpes Simplex/cerebrospinal fluid , Encephalitis, Herpes Simplex/diagnosis , Encephalitis, Herpes Simplex/drug therapy , Female , Humans , Immunoglobulin G , Male , Middle Aged , Prospective Studies , Receptors, N-Methyl-D-Aspartate/immunology , Young Adult
3.
Biofactors ; 47(2): 190-197, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33098588

ABSTRACT

Neuroinflammation leads to neurodegeneration, cognitive defects, and neurodegenerative disorders. Neurotrauma/traumatic brain injury (TBI) can cause activation of glial cells, neurons, and neuroimmune cells in the brain to release neuroinflammatory mediators. Neurotrauma leads to immediate primary brain damage (direct damage), neuroinflammatory responses, neuroinflammation, and late secondary brain damage (indirect) through neuroinflammatory mechanism. Secondary brain damage leads to chronic inflammation and the onset and progression of neurodegenerative diseases. Currently, there are no effective and specific therapeutic options to treat these brain damages or neurodegenerative diseases. Flavone luteolin is an important natural polyphenol present in several plants that show anti-inflammatory, antioxidant, anticancer, cytoprotective, and macrophage polarization effects. In this short review article, we have reviewed the neuroprotective effects of luteolin in neurotrauma and neurodegenerative disorders and pathways involved in this mechanism. We have collected data for this study from publications in the PubMed using the keywords luteolin and mast cells, neuroinflammation, neurodegenerative diseases, and TBI. Recent reports suggest that luteolin suppresses systemic and neuroinflammatory responses in Coronavirus disease 2019 (COVID-19). Studies have shown that luteolin exhibits neuroprotective effects through various mechanisms, including suppressing immune cell activation, such as mast cells, and inflammatory mediators released from these cells. In addition, luteolin can suppress neuroinflammatory response, activation of microglia and astrocytes, oxidative stress, neuroinflammation, and the severity of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and TBI pathogenesis. In conclusion, luteolin can improve cognitive decline and enhance neuroprotection in neurodegenerative diseases, TBI, and stroke.


Subject(s)
Brain Injuries, Traumatic/drug therapy , COVID-19 Drug Treatment , Inflammation/drug therapy , Luteolin/therapeutic use , Neuroprotective Agents/therapeutic use , Brain/drug effects , Brain/virology , Brain Injuries/complications , Brain Injuries/drug therapy , Brain Injuries/virology , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/virology , COVID-19/complications , COVID-19/virology , Flavones/therapeutic use , Humans , Inflammation/complications , Inflammation/virology , Neurons/drug effects , Neurons/virology , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity
5.
J Virol ; 94(19)2020 09 15.
Article in English | MEDLINE | ID: mdl-32669339

ABSTRACT

Brain injury occurs within days in simian immunodeficiency virus (SIV) or human immunodeficiency virus (HIV) infection, and some recovery may occur within weeks. Inflammation and oxidative stress associate with such injury, but what drives recovery is unknown. Chronic HIV infection associates with reduced brain frontal cortex expression of the antioxidant/anti-inflammatory enzyme heme oxygenase-1 (HO-1) and increased neuroinflammation in individuals with cognitive impairment. We hypothesized that acute regional brain injury and recovery associate with differences in regional brain HO-1 expression. Using SIV-infected rhesus macaques, we analyzed multiple brain regions through acute and chronic infection (90 days postinfection [dpi]) and quantified viral (SIV gag RNA), synaptic (PSD-95; synaptophysin), axonal (neurofilament/neurofilament light chain [NFL]), inflammatory, and antioxidant (enzymes, including heme oxygenase isoforms [HO-1, HO-2]) markers. PSD-95 was reduced in the brainstem, basal ganglia, neocortex, and cerebellum within 13 dpi, indicating acute synaptic injury throughout the brain. All areas except the brainstem recovered. Unchanged NFL was consistent with no acute axonal injury. SIV RNA expression was highest in the brainstem throughout infection, and it associated with neuroinflammation. Surprisingly, during the synaptic injury and recovery phases, HO-2, and not HO-1, progressively decreased in the brainstem. Thus, acute SIV synaptic injury occurs throughout the brain, with spontaneous recovery in regions other than the brainstem. Within the brainstem, the high SIV load and inflammation, along with reduction of HO-2, may impair recovery. In other brain regions, stable HO-2 expression, with or without increasing HO-1, may promote recovery. Our data support roles for heme oxygenase isoforms in modulating recovery from synaptic injury in SIV infection and suggest their therapeutic targeting for promoting neuronal recovery.IMPORTANCE Brain injury induced by acute simian (or human) immunodeficiency virus infection may persist or spontaneously resolve in different brain regions. Identifying the host factor(s) that promotes spontaneous recovery from such injury may reveal targets for therapeutic drug strategies for promoting recovery from acute neuronal injury. The gradual recovery from such injury observed in many, but not all, brain regions in the rhesus macaque model is consistent with the possible existence of a therapeutic window of opportunity for intervening to promote recovery, even in those regions not showing spontaneous recovery. In persons living with human immunodeficiency virus infection, such neuroprotective treatments could ultimately be considered as adjuncts to the initiation of antiretroviral drug therapy.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Heme Oxygenase (Decyclizing)/metabolism , Simian Acquired Immunodeficiency Syndrome/metabolism , Simian Immunodeficiency Virus/immunology , Animals , Anti-Inflammatory Agents , Brain/pathology , Brain/virology , Brain Injuries/pathology , Brain Injuries/virology , Disease Models, Animal , Female , HIV Infections , Heme Oxygenase-1/metabolism , Inflammation , Macaca mulatta , Male , Oxidative Stress , Protein Isoforms , Simian Acquired Immunodeficiency Syndrome/pathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/pathogenicity
6.
Sci Rep ; 9(1): 8004, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142789

ABSTRACT

Despite treatment, immune activation is thought to contribute to cerebral injury in children perinatally infected with human immunodeficiency virus (HIV). We aimed to characterize immune activation in relation to neuroimaging and cognitive outcomes. We therefore measured immunological, coagulation, and neuronal biomarkers in plasma and cerebrospinal fluid (CSF) samples of 34 perinatally HIV-infected children aged 8-18 years, and in plasma samples of 37 controls of comparable age, sex, ethnicity, and socio-economic status. We then compared plasma biomarker levels between groups, and explored associations between plasma/CSF biomarkers and neuroimaging and cognitive outcomes using network analysis. HIV-infected children showed higher plasma levels of C-reactive protein, interferon-gamma, interferon-gamma-inducible protein-10, and monocyte chemoattractant protein-1 than controls. In HIV-infected participants, plasma soluble CD14 was positively associated with microstructural white matter (WM) damage, and plasma D-dimer was negatively associated with WM blood flow. In CSF, IL-6 was negatively associated with WM volume, and neurofilament heavy-chain (NFH) was negatively associated with intelligence quotient and working memory. These markers of ongoing inflammation, immune activation, coagulation, and neuronal damage could be used to further evaluate the pathophysiology and clinical course of cerebral and cognitive deficits in perinatally acquired HIV.


Subject(s)
Cognitive Dysfunction/immunology , HIV Infections/immunology , Immunity, Cellular/genetics , Inflammation/immunology , Adolescent , Biomarkers/blood , Brain Injuries/complications , Brain Injuries/immunology , Brain Injuries/pathology , Brain Injuries/virology , Chemokine CCL2/genetics , Chemokine CXCL10/genetics , Child , Cognitive Dysfunction/complications , Cognitive Dysfunction/pathology , Cognitive Dysfunction/virology , Female , HIV/pathogenicity , HIV Infections/complications , HIV Infections/pathology , HIV Infections/virology , Humans , Inflammation/complications , Inflammation/pathology , Inflammation/virology , Male , Neuroimaging , Pediatrics , White Matter/immunology , White Matter/pathology , White Matter/virology
7.
Article in English | MEDLINE | ID: mdl-31921708

ABSTRACT

Hepatitis E virus (HEV) infection has been associated with extrahepatic manifestations, particularly neurological disorders. Although it has been reported that HEV infection induced hepatocyte apoptosis associated with mitochondria injury, activation of mitochondrial apoptotic pathway in the central nervous system during HEV infection was not clearly understood. In this study, the induction of mitochondrial apoptosis-associated proteins and pro-inflammatory cytokines were detected in HEV infected Mongolian gerbil model and primary human brain microvascular endothelial cells (HBMVECs). Mitochondrial exhibited fragments with loss of cristae and matrix in HEV infected brain tissue by transmission electron microscope (TEM). In vitro studies showed that expression of NADPH oxidase 4 (NOX4) was significantly increased in HEV infected HBMVECs (p < 0.05), while ATP5A1 was significantly decreased (p < 0.01). Expressions of pro-apoptotic proteins were further evaluated. Bax was significantly increased in both HEV infected brain tissues and HBMVECs (p < 0.01). In vivo studies showed that caspase-9 and caspase-3 were activated after HEV inoculation (p < 0.01), associated with PCNA overexpression as response to apoptosis. Cytokines were measured to evaluate tissue inflammatory levels. Results showed that the release of TNFα and IL-1ß were significantly increased after HEV infection (p < 0.01), which might be attributed to microglia activation characterized by high level of IBA1 expression (p < 0.01). Taken together, these data support that HEV infection induces high levels of pro-inflammatory cytokines, associated with mitochondria-mediated apoptosis. The results provide new insight into mechanisms of extra-hepatic injury of HEV infection, especially in the central nervous system.


Subject(s)
Apoptosis/physiology , Brain Injuries/virology , Cytokines/metabolism , Hepatitis E/pathology , Mitochondria/pathology , Animals , Brain/blood supply , Brain/pathology , Brain/virology , Caspase 3/metabolism , Caspase 9/metabolism , Cells, Cultured , Endothelial Cells/virology , Gerbillinae/virology , Hepatitis E virus/pathogenicity , Humans , Mitochondrial Proton-Translocating ATPases/metabolism , NADPH Oxidase 4/metabolism , Proliferating Cell Nuclear Antigen/metabolism , bcl-2-Associated X Protein/metabolism
8.
Fetal Diagn Ther ; 46(1): 1-11, 2019.
Article in English | MEDLINE | ID: mdl-30032153

ABSTRACT

BACKGROUND: Infection with parvovirus B19 (B19V) during pregnancy may cause severe fetal anemia, hydrops, and fe tal death. Furthermore, neurodevelopmental impairment among survivors may occur despite appropriate prenatal management, including intrauterine transfusion (IUT). OBJECTIVES: Our primary objective was to describe cerebral lesions on MRI in fetuses with severe anemia requiring IUT for B19V infection. Our secondary objective was to search for clinical and biological characteristics associated with the occurrence of such lesions. STUDY DESIGN: We performed a retrospective review of data on fetuses infected with B19V and requiring at least one IUT between 2005 and 2016. Fetuses with abnormal cerebral MRI results in the 3rd trimester were compared to those with normal MRI results. RESULTS: Of 34 transfused fetuses, 26 children were born at full term. Five intrauterine fetal deaths, 1 neonatal death, and 2 terminations of pregnancy occurred. Cerebral anomalies were observed in 7/27 fetuses on MRI, including cerebellar hemorrhage or a small cerebellum. Only viral load in fetal blood appeared to be associated with brain lesions (11.5 log10 copies/mL [10.5-12.5] in case of abnormal MRI results vs. 9.5 log10 copies/mL [7.8-10.0]; p = 0.05). CONCLUSIONS: Among the fetuses transfused for B19V infection, 26% presented with prenatal abnormal cerebral imaging results. In our study, viral load in fetal blood appeared to be the only factor associated with fetal brain lesions.


Subject(s)
Brain Injuries/virology , Erythema Infectiosum/diagnostic imaging , Prenatal Diagnosis , Blood Transfusion, Intrauterine , Erythema Infectiosum/complications , Erythema Infectiosum/therapy , Hemodynamics , Humans , Intracranial Hemorrhages/diagnostic imaging , Intracranial Hemorrhages/etiology , Retrospective Studies
9.
Virol J ; 14(1): 243, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29282065

ABSTRACT

BACKGROUND: Enterovirus (EV) infection has been a serious health issue in Asia-Pacific region. It has been indicated that the occurrence of fatal hand foot and mouth disease (HFMD) cases following EV71 infection is mainly attributed to pulmonary edema. However, the development of pulmonary disorders after EV71 infection remains largely unknown. To establish an EV71-infected animal model and further explore the underlying association of central nervous system (CNS) invasion with pulmonary edema, we isolated a clinical source EV71 strain (ZZ1350) from a severe case in Henan Province. METHODS: We evaluated the cytotoxicity of ZZ1350 strain and the susceptibility in 3-day-old BALB/c mice with intraperitoneal, intracerebral and intramuscular inoculation. Various histopathological and immunohistochemical techniques were applied to determine the target organs or tissue damage after infection. Correlation analysis was used to identify the relationship between CNS injury and pulmonary disorders. RESULTS: Our experimental results suggested that ZZ1350 (C4 subtype) had high cytotoxicity against African green monkey kidney (Vero) cells and human rhabdomyosarcoma (RD) cells and neonatal BALB/c mice were highly susceptible to the infection with ZZ1350 through three different inoculation routes (2 × 106 pfu/mouse) exhibiting severe neurological and respiratory symptoms that were similar to clinical observation. Viral replication was found in brain, spinal cord, skeletal muscle, lung, spleen, liver, heart of infected mice and these sections also showed histopathological changes. We found that brain histology score was positive correlated with lung histology score in total experimental mice and mice under the three inoculation routes (P < 0.05). At the same time, there were positive correlations between spinal cord score and lung score in total experimental mice and mice with intracerebral inoculation (P < 0.05). CONCLUSIONS: ZZ1350 strain is effective to establish animal model of EV71 infection with severe neurological and respiratory symptoms. The development of pulmonary disorders after EV71 infection is associated with severity of CNS damage.


Subject(s)
Brain Injuries/virology , Enterovirus A, Human/pathogenicity , Enterovirus Infections/complications , Lung/virology , Pulmonary Edema/virology , Spinal Cord Injuries/virology , Animals , Brain Injuries/pathology , Cell Line, Tumor , Cell Survival , China , Chlorocebus aethiops , Disease Models, Animal , Disease Susceptibility , Enterovirus A, Human/isolation & purification , Enterovirus Infections/pathology , Humans , Lung/pathology , Mice , Spinal Cord Injuries/pathology , Vero Cells
10.
Article in English | MEDLINE | ID: mdl-28469998

ABSTRACT

Enterovirus 71 (EV71) infects the central nervous system (CNS) and causes brainstem encephalitis in children. MiRNAs have been found to play various functions in EV71 infection in human cell lines. To identify potential miRNAs involved in the inflammatory injury in CNS, our study, for the first time, performed a miRNA microarray assay in vivo using EV71 infected mice brains. Twenty differentially expressed miRNAs were identified (four up- and 16 down-regulated) and confirmed by qRT-PCR. The target genes of these miRNAs were analyzed using KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis, revealing that the miRNAs were mainly involved in the regulation of inflammation and neural system function. MiR-150-5p, -3082-5p, -3473a, -468-3p, -669n, -721, -709, and -5107-5p that regulate MAPK and chemokine signaling were all down-regulated, which might result in increased cytokine production. In addition, miR-3473a could also regulate focal adhesion and leukocyte trans-endothelial migration, suggesting a role in virus-induced blood-brain barrier disruption. The miRNAs and pathways identified in this study could help to understand the intricate interactions between EV71 and the brain injury, offering new insight for the future research of the molecular mechanism of EV71 induced brainstem encephalitis.


Subject(s)
Brain Injuries/pathology , Brain/virology , Enterovirus A, Human/pathogenicity , MicroRNAs/metabolism , MicroRNAs/pharmacology , Animals , Blood-Brain Barrier , Brain/pathology , Brain Injuries/virology , Cell Line , Central Nervous System/pathology , Central Nervous System/virology , Chemokines/metabolism , Cytokines/metabolism , Down-Regulation , Enterovirus A, Human/genetics , Enterovirus Infections , Gene Expression Profiling , Humans , Inflammation , Mice , MicroRNAs/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Real-Time Polymerase Chain Reaction , Survival Rate
11.
Sci Rep ; 7: 46514, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28425451

ABSTRACT

Infection with human immunodeficiency virus-1 (HIV-1) causes brain injury. Type I interferons (IFNα/ß) are critical mediators of any anti-viral immune response and IFNß has been implicated in the temporary control of lentiviral infection in the brain. Here we show that transgenic mice expressing HIV-1 envelope glycoprotein 120 in their central nervous system (HIVgp120tg) mount a transient IFNß response and provide evidence that IFNß confers neuronal protection against HIVgp120 toxicity. In cerebrocortical cell cultures, neuroprotection by IFNß against gp120 toxicity is dependent on IFNα receptor 1 (IFNAR1) and the ß-chemokine CCL4, as IFNAR1 deficiency and neutralizing antibodies against CCL4, respectively, abolish the neuroprotective effects. We find in vivo that IFNß mRNA is significantly increased in HIVgp120tg brains at 1.5, but not 3 or 6 months of age. However, a four-week intranasal IFNß treatment of HIVgp120tg mice starting at 3.5 months of age increases expression of CCL4 and concomitantly protects neuronal dendrites and pre-synaptic terminals in cortex and hippocampus from gp120-induced damage. Moreover, in vivo and in vitro data suggests astrocytes are a major source of IFNß-induced CCL4. Altogether, our results suggest exogenous IFNß as a neuroprotective factor that has potential to ameliorate in vivo HIVgp120-induced brain injury.


Subject(s)
Brain Injuries/prevention & control , Disease Models, Animal , Interferon-beta/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain Injuries/virology , Cells, Cultured , Chemokine CCL4/genetics , Chemokine CCL4/metabolism , Gene Expression/drug effects , HIV Envelope Protein gp120/antagonists & inhibitors , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV-1/physiology , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Mice, Transgenic , Neurons/pathology , Neurons/virology , Rats , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism
12.
Medicine (Baltimore) ; 96(51): e9178, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29390455

ABSTRACT

RATIONALE: A major epidemic of Zika virus (ZIKV) infection occurred in French Guiana and West Indies. French national epidemiological surveillance estimated that 1650 pregnant women contracted the ZIKV during epidemic period from January 2016 to October 2016 in French Guiana. PATIENT CONCERNS: ZIKV infection during pregnancy is a cause of microcephaly and birth defects. DIAGNOSES: In this report, we describe 2 children with proven in utero ZIKV exposure. Their mothers were both symptomatic and ZIKV infection occurred early in pregnancy. Ultrasonography monitoring in utero did not show any abnormality for both patient. They were born at full-term, healthy, without any birth defects and no sign of congenital ZIKV infection. INTERVENTIONS: ZIKV was neither found on placenta fragments nor children blood and urine at birth. Their neurodevelopment outcomes in early-life fitted the expectations. As recommended in national guidelines, we performed cerebral MRIs at 2 months old, showing severe brain abnormalities, especially of white matter areas. After a large screening, we did not find any differential diagnosis for their brain lesions. OUTCOMES: We concluded it was due to their in utero ZIKV exposure. LESSONS: In this report, pathogenicity of ZIKV may involve mother's immunological response or metabolic disorder during the infection.


Subject(s)
Brain Injuries/virology , Magnetic Resonance Imaging/methods , Pregnancy Complications, Infectious/diagnostic imaging , Prenatal Exposure Delayed Effects/diagnostic imaging , Zika Virus Infection/diagnosis , Adult , Brain Injuries/diagnostic imaging , Brain Injuries/etiology , Cesarean Section , Child Development/physiology , Female , Follow-Up Studies , French Guiana , Humans , Infant , Infant, Newborn , Male , Pregnancy , Pregnancy Complications, Infectious/virology , Prenatal Exposure Delayed Effects/pathology , Risk Assessment , Ultrasonography, Prenatal/methods , Zika Virus Infection/complications
13.
J Neurovirol ; 22(2): 149-58, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26424107

ABSTRACT

A wide spectrum of neurocognitive deficits characterises HIV infection in adults. HIV infection is additionally associated with morphological brain abnormalities affecting neural substrates that subserve neurocognitive function. Early life stress (ELS) also has a direct influence on brain morphology. However, the combined impact of ELS and HIV on brain structure and neurocognitive function has not been examined in an all-female sample with advanced HIV disease. The present study examined the effects of HIV and childhood trauma on brain morphometry and neurocognitive function. Structural data were acquired using a 3T Magnetom MRI scanner, and a battery of neurocognitive tests was administered to 124 women: HIV-positive with ELS (n = 32), HIV-positive without ELS (n = 30), HIV-negative with ELS (n = 31) and HIV-negative without ELS (n = 31). Results revealed significant group volumetric differences for right anterior cingulate cortex (ACC), bilateral hippocampi, corpus callosum, left and right caudate and left and right putamen. Mean regional volumes were lowest in HIV-positive women with ELS compared to all other groups. Although causality cannot be inferred, findings also suggest that alterations in the left frontal lobe, right ACC, left hippocampus, corpus callosum, left and right amygdala and left caudate may be associated with poorer neurocognitive performance in the domains of processing speed, attention/working memory, abstraction/executive functions, motor skills, learning and language/fluency with these effects more pronounced in women living with both HIV and childhood trauma. This study highlights the potential contributory role of childhood trauma to brain alterations and neurocognitive decline in HIV-infected individuals.


Subject(s)
Brain Injuries/physiopathology , Cognitive Dysfunction/physiopathology , HIV Infections/physiopathology , Stress, Psychological/physiopathology , Adolescent , Adult , Aged , Attention , Brain Injuries/complications , Brain Injuries/pathology , Brain Injuries/virology , Case-Control Studies , Caudate Nucleus/pathology , Caudate Nucleus/physiopathology , Caudate Nucleus/virology , Child , Cognitive Dysfunction/complications , Cognitive Dysfunction/pathology , Cognitive Dysfunction/virology , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Corpus Callosum/virology , Executive Function , Female , Gyrus Cinguli/pathology , Gyrus Cinguli/physiopathology , Gyrus Cinguli/virology , HIV Infections/complications , HIV Infections/pathology , HIV Infections/virology , Hippocampus/pathology , Hippocampus/physiopathology , Hippocampus/virology , Humans , Magnetic Resonance Imaging , Memory, Short-Term , Motor Skills , Neuropsychological Tests , Putamen/pathology , Putamen/physiopathology , Putamen/virology , Stress, Psychological/complications , Stress, Psychological/pathology , Stress, Psychological/virology , Time Factors
14.
Neurology ; 86(1): 19-27, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26561287

ABSTRACT

OBJECTIVE: The current study aims to evaluate the neurologic state of perinatally HIV-infected children on combination antiretroviral therapy and to attain a better insight into the pathogenesis of their persistent neurologic and cognitive deficits. METHODS: We included perinatally HIV-infected children between 8 and 18 years and healthy controls matched for age, sex, ethnicity, and socioeconomic status. All participants underwent a 3.0 T MRI with 3D-T1-weighted, 3D-fluid-attenuated inversion recovery, and diffusion-weighted series for the evaluation of cerebral volumes, white matter hyperintensities (WMH), and white matter (WM) diffusion characteristics. Associations with disease-related parameters and cognitive performance were explored using linear regression models. RESULTS: We included 35 cases (median age 13.8 years) and 37 controls (median age 12.1 years). A lower gray matter and WM volume, more WMH, and a higher WM diffusivity were observed in the cases. Within the HIV-infected children, a poorer clinical, immunologic, and virologic state were negatively associated with volumetric, WMH, and diffusivity markers. CONCLUSIONS: In children with HIV, even when long-term clinically and virologically controlled, we found lower brain volumes, a higher WMH load, and poorer WM integrity compared to matched controls. These differences occur in the context of a poor cognitive performance in the HIV-infected group, and larger, longitudinal studies are needed to increase our understanding of the pathogenesis of cerebral injury in perinatally HIV-infected children.


Subject(s)
Brain Injuries/diagnosis , Brain Injuries/etiology , HIV Infections/complications , HIV Infections/diagnosis , Adolescent , Brain/pathology , Brain Injuries/virology , Child , Female , Humans , Male , Organ Size , Viral Load
15.
J Neuroinflammation ; 12: 171, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26377670

ABSTRACT

BACKGROUND: HIV-associated neuroinflammation is believed to be a major contributing factor in the development of HIV-associated neurocognitive disorders (HAND). In this study, we used micropositron emission tomography (PET) imaging to quantify neuroinflammation in HIV-1 transgenic rat (Tg), a small animal model of HIV, known to develop neurological and behavioral problems. METHODS: Dynamic [(18)F]DPA-714 PET imaging was performed in Tg and age-matched wild-type (WT) rats in three age groups: 3-, 9-, and 16-month-old animals. As a positive control for neuroinflammation, we performed unilateral intrastriatal injection of quinolinic acid (QA) in a separate group of WT rats. To confirm our findings, we performed multiplex immunofluorescent staining for Iba1 and we measured cytokine/chemokine levels in brain lysates of Tg and WT rats at different ages. RESULTS: [(18)F]DPA-714 uptake in HIV-1 Tg rat brains was generally higher than in age-matched WT rats but this was not statistically significant in any age group. [(18)F]DPA-714 uptake in the QA-lesioned rats was significantly higher ipsilateral to the lesion compared to contralateral side indicating neuroinflammatory changes. Iba1 immunofluorescence showed no significant differences in microglial activation between the Tg and WT rats, while the QA-lesioned rats showed significant activation. Finally, cytokine/chemokine levels in brain lysates of the Tg rats and WT rats were not significantly different. CONCLUSION: Microglial activation might not be the primary mechanism for neuropathology in the HIV-1 Tg rats. Although [(18)F]DPA-714 is a good biomarker of neuroinflammation, it cannot be reliably used as an in vivo biomarker of neurodegeneration in the HIV-1 Tg rat.


Subject(s)
Brain Injuries/virology , Encephalitis/diagnostic imaging , Fluorodeoxyglucose F18/pharmacokinetics , HIV-1/metabolism , Positron-Emission Tomography , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/genetics , Brain Injuries/chemically induced , Brain Injuries/complications , Brain Injuries/diagnostic imaging , Brain Mapping , Cytokines/metabolism , Encephalitis/etiology , Fluorodeoxyglucose F18/blood , Functional Laterality , HIV-1/genetics , Male , Pyrazoles/blood , Pyrimidines/blood , Quinolinic Acid/toxicity , Rats , Rats, Inbred F344 , Rats, Transgenic , Time Factors
16.
J Neurovirol ; 20(3): 209-18, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24549970

ABSTRACT

Despite modern antiretroviral therapy, HIV-associated sensory neuropathy affects over 50 % of HIV patients. The clinical expression of HIV neuropathy is highly variable: many individuals report few symptoms, but about half report distal neuropathic pain (DNP), making it one of the most prevalent, disabling, and treatment-resistant complications of HIV disease. The presence and intensity of pain is not fully explained by the degree of peripheral nerve damage, making it unclear why some patients do, and others do not, report pain. To better understand central nervous system contributions to HIV DNP, we performed a cross-sectional analysis of structural magnetic resonance imaging volumes in 241 HIV-infected participants from an observational multi-site cohort study at five US sites (CNS HIV Anti-Retroviral Treatment Effects Research Study, CHARTER). The association between DNP and the structural imaging outcomes was investigated using both linear and nonlinear (Gaussian Kernel support vector) multivariable regression, controlling for key demographic and clinical variables. Severity of DNP symptoms was correlated with smaller total cerebral cortical gray matter volume (r = -0.24; p = 0.004). Understanding the mechanisms for this association between smaller total cortical volumes and DNP may provide insight into HIV DNP chronicity and treatment-resistance.


Subject(s)
AIDS Dementia Complex/epidemiology , AIDS Dementia Complex/pathology , Magnetic Resonance Imaging , Neuralgia , AIDS Dementia Complex/drug therapy , Adult , Anti-Retroviral Agents/therapeutic use , Brain Injuries/epidemiology , Brain Injuries/pathology , Brain Injuries/virology , Cerebral Cortex/pathology , Cerebral Cortex/virology , Cognition Disorders/epidemiology , Cognition Disorders/pathology , Cognition Disorders/virology , Confounding Factors, Epidemiologic , Cross-Sectional Studies , Female , Gray Matter/pathology , Gray Matter/virology , Humans , Male , Mental Disorders/epidemiology , Mental Disorders/pathology , Mental Disorders/virology , Middle Aged , Neuralgia/epidemiology , Neuralgia/pathology , Neuralgia/virology , Prevalence , Risk Factors , Substance-Related Disorders/epidemiology , Substance-Related Disorders/pathology , Substance-Related Disorders/virology
17.
J Neuropathol Exp Neurol ; 71(1): 15-27, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22157616

ABSTRACT

The simian immunodeficiency virus (SIV) macaque model resembles human immunodeficiency virus-acquired immunodeficiency syndrome (AIDS) and associated brain dysfunction. Altered expression of synaptic markers and transmitters in neuro-AIDS has been reported, but limited data exist for the cholinergic system and lipid mediators such as prostaglandins. Here, we analyzed cholinergic basal forebrain neurons with their telencephalic projections and the rate-limiting enzymes for prostaglandin synthesis, cyclooxygenase isotypes 1 and 2 (COX1 and COX2) in the brains of SIV-infected macaques with or without encephalitis and antiretroviral therapy and uninfected controls.Cyclooxygenase isotype 1, but not COX2, was coexpressed with markers of cholinergic phenotype, that is, choline acetyltransferase and vesicular acetylcholine transporter (VAChT), in basal forebrain neurons of monkey, as well as human, brain. Cyclooxygenase isotype 1 was decreased in basal forebrain neurons in macaques with AIDS versus uninfected and asymptomatic SIV-infected macaques. The VAChT-positive fiber density was reduced in frontal, parietal, and hippocampal-entorhinal cortex. Although brain SIV burden and associated COX1- and COX2-positive mononuclear and endothelial inflammatory reactions were mostly reversed in AIDS-diseased macaques that received 6-chloro-2',3'-dideoxyguanosine treatment, decreased VAChT-positive terminal density and reduced cholinergic COX1 expression were not. Thus, COX1 expression is a feature of primate cholinergic basal forebrain neurons; it may be functionally important and a critical biomarker of cholinergic dysregulation accompanying lentiviral encephalopathy. These results further imply that insufficiently prompt initiation of antiretroviral therapy in lentiviral infection may lead to neurostructurally unremarkable but neurochemically prominent irreversible brain damage.


Subject(s)
Cerebral Cortex/virology , Cholinergic Neurons/metabolism , Hippocampus/virology , Prosencephalon/virology , Prostaglandins/biosynthesis , Simian Acquired Immunodeficiency Syndrome/metabolism , Animals , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/virology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Cholinergic Neurons/pathology , Cholinergic Neurons/virology , Hippocampus/metabolism , Hippocampus/pathology , Lentivirus Infections/metabolism , Lentivirus Infections/pathology , Macaca mulatta , Neural Pathways/metabolism , Neural Pathways/pathology , Neural Pathways/virology , Prosencephalon/metabolism , Prosencephalon/pathology , Retroviruses, Simian , Simian Acquired Immunodeficiency Syndrome/pathology , Time Factors
18.
J Virol ; 83(1): 420-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18945784

ABSTRACT

Cytomegalovirus (CMV) is one of the most common viral pathogens leading to neurological dysfunction in individuals with depressed immune systems. How CMV enters the brain remains an open question. The hypothesis that brain injury may enhance the entrance of CMV into the brain was tested. Insertion of a sterile needle into the brain caused a dramatic increase in mouse CMV in the brains of immunodeficient SCID mice inoculated peripherally within an hour of injury and examined 1 week later; peripheral inoculation 48 h after injury and a 1-week survival resulted in only a modest infection at the site of injury. In contrast, uninjured SCID mice, as well as injured immunocompetent control mice, showed little sign of viral infection at the same time intervals. Direct inoculation of the brain resulted in widespread dispersal and enhanced replication of mCMV in SCID brains tested 1 week later but not in parallel control brains. Differential viremia was unlikely to account for the greater viral load in the SCID brain, since increased mCMV in the blood of SCID compared to controls was not detected until a longer interval. These data suggest that brain injury enhances CMV invasion of the brain, but only when the adaptive immune system is compromised, and that the brain's ability to resist viral infection recovers rapidly after injury.


Subject(s)
Brain Injuries/complications , Brain Injuries/virology , Cytomegalovirus Infections , Animals , Immunocompromised Host , Mice , Mice, SCID , Survival Analysis , Viremia
19.
Neuropediatrics ; 36(2): 117-9, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15822026

ABSTRACT

Bilateral striatal lesion is characterised by a specific clinical syndrome (encephalopathy with rigidity, irritability, variable pyramidal, and extrapyramidal symptoms, speech abnormalities) and symmetrical lesion of the basal ganglia including the caudate nucleus, the putamen, and occasionally other nuclei. We report three cases in whom bilateral striatal lesion developed in association with varicella. Each patient recovered completely and showed no signs of cognitive deficiency, chorea or hyperkinetic syndrome, all of which have been reported as sequelae of BSL associated with other conditions. These cases suggest that bilateral striatal lesion may be an immune-mediated complication of varicella.


Subject(s)
Brain Injuries/etiology , Chickenpox/complications , Corpus Striatum/physiopathology , Brain Injuries/pathology , Brain Injuries/virology , Chickenpox/pathology , Chickenpox/virology , Child, Preschool , Cognition/physiology , Corpus Striatum/pathology , Corpus Striatum/virology , Female , Humans , Magnetic Resonance Imaging/methods , Male
20.
Ann Neurol ; 56(2): 259-72, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15293278

ABSTRACT

Human immunodeficiency virus (HIV)-positive patients commonly have attention and concentration problems. However, it remains unclear how HIV infection affects the attention network. Therefore, blood oxygenation level dependent functional magnetic resonance imaging (BOLD-fMRI) was performed in 36 subjects (18 HIV and 18 seronegative [SN] controls) during a set of visual attention tasks with increasing levels of attentional load. Compared with SN controls, HIV subjects showed similar task performance (accuracies and reaction times) but decreased activation in the normal visual attention network (dorsal parietal, bilateral prefrontal, and cerebellar regions) and increased activation in adjacent or contralateral brain regions. Cognitive performance (assessed with NPZ-8), CD4, and viral load all correlated with activated BOLD signals in brain regions that activated more in HIV subjects. Furthermore, HIV subjects activated more than SN controls in brain regions that showed load-dependent increase in activation (right prefrontal and right parietal regions) but less in regions that showed a saturation effect with increasing load. These findings suggest that HIV-associated brain injury leads to reduced efficiency in the normal attention network, thus requiring reorganization and increased usage of neural reserves to maintain performance during attention-requiring tasks. Exceeding the brain reserve capacity may lead to attention deficits and cognitive impairment in HIV patients.


Subject(s)
Attention/physiology , Brain Injuries/physiopathology , HIV Infections/complications , HIV , Nerve Net/physiopathology , Adult , Analysis of Variance , Brain Injuries/virology , Brain Mapping , CD4 Lymphocyte Count/methods , Cerebrovascular Circulation/physiology , Female , Functional Laterality , Humans , Image Processing, Computer-Assisted/methods , Magnetic Resonance Imaging/methods , Male , Nerve Net/virology , Neuropsychological Tests , Oxygen/blood , Viral Load/methods , Visual Perception/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...