Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Sci Rep ; 11(1): 6357, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737684

ABSTRACT

The recent SARS-CoV-2 pandemic manifests itself as a mild respiratory tract infection in most individuals, leading to COVID-19 disease. However, in some infected individuals, this can progress to severe pneumonia and acute respiratory distress syndrome (ARDS), leading to multi-organ failure and death. This study explores the proteomic differences between mild, severe, and critical COVID-19 positive patients to further understand the disease progression, identify proteins associated with disease severity, and identify potential therapeutic targets. Blood protein profiling was performed on 59 COVID-19 mild (n = 26), severe (n = 9) or critical (n = 24) cases and 28 controls using the OLINK inflammation, autoimmune, cardiovascular and neurology panels. Differential expression analysis was performed within and between disease groups to generate nine different analyses. From the 368 proteins measured per individual, more than 75% were observed to be significantly perturbed in COVID-19 cases. Six proteins (IL6, CKAP4, Gal-9, IL-1ra, LILRB4 and PD-L1) were identified to be associated with disease severity. The results have been made readily available through an interactive web-based application for instant data exploration and visualization, and can be accessed at https://phidatalab-shiny.rosalind.kcl.ac.uk/COVID19/ . Our results demonstrate that dynamic changes in blood proteins associated with disease severity can potentially be used as early biomarkers to monitor disease severity in COVID-19 and serve as potential therapeutic targets.


Subject(s)
Biomarkers/blood , COVID-19/blood , Central Nervous System Diseases/virology , Proteome , Aged , COVID-19/complications , Case-Control Studies , Cohort Studies , Female , Gene Expression Profiling , Gliosis/virology , Humans , Male , Middle Aged , Nerve Tissue Proteins/blood
2.
Viruses ; 12(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32906600

ABSTRACT

Astroviruses (AstVs) cause disease in a wide variety of species. Porcine AstVs are highly genetically diverse and conventionally assigned to five genetic lineages (PoAstV1-5). Due to the increasing evidence that porcine astrovirus type 3 (PoAstV3) is a cause of encephalomyelitis in swine and to elucidate important ecologic characteristics, the infection dynamics and environmental distribution of PoAstV3 were investigated in a herd with PoAstV3-associated neurologic disease. Over a 22 week period, the frequency of PoAstV3 fecal shedding varied by pig and age. The peak detection by RT-qPCR of PoAstV3 on fecal swabs (95%; 61 of 64) occurred at 3 weeks of age. The lowest frequency of detection was at 21 weeks of age (4%; 2 of 47); however, the frequency increased to 41% (19 of 46) at the final sampling time point (25 weeks of age). Viremia was rare (0.9%: 4 of 433). Detection in oral fluid was consistent with 75% to 100% of samples positive at each time point. Pens and feeders also had a high rate of detection with a majority of samples positive at a majority of sampling time points. Based on the data presented, PoAstV3 can be consistently detected in the environment with a majority of pigs being infected and a subset intermittently shedding the virus in feces out to 25 weeks of age. These findings suggest the importance of as-yet unidentified risk factors associated with the development of PoAstV3-associated polioencephalomyelitis.


Subject(s)
Astroviridae Infections/virology , Ecology , Mamastrovirus/physiology , Nervous System Diseases/virology , Animals , Case-Control Studies , Feces/virology , Gliosis/pathology , Gliosis/virology , Mamastrovirus/genetics , Mamastrovirus/isolation & purification , Pilot Projects , Swine , Swine Diseases/virology , Viremia/veterinary , Virus Shedding
3.
J Neurovirol ; 26(6): 838-845, 2020 12.
Article in English | MEDLINE | ID: mdl-32901392

ABSTRACT

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


Subject(s)
Brain/metabolism , Cognitive Dysfunction/genetics , Disease Models, Animal , Gliosis/genetics , HIV Infections/genetics , HIV-1/genetics , Animals , Astrocytes/metabolism , Astrocytes/virology , Biomarkers/metabolism , Brain/virology , Cognitive Dysfunction/complications , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/virology , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/complications , Gliosis/metabolism , Gliosis/virology , HIV Core Protein p24/genetics , HIV Core Protein p24/metabolism , HIV Infections/complications , HIV Infections/metabolism , HIV Infections/virology , HIV-1/metabolism , HIV-1/pathogenicity , Humans , Inflammation , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Macrophages/metabolism , Macrophages/virology , Male , Mice , Mice, SCID , Microglia/metabolism , Microglia/virology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Neurons/metabolism , Neurons/virology , Phenotype
4.
Sci Rep ; 9(1): 17926, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31784616

ABSTRACT

Dengue is an important arboviral infection, causing a broad range symptom that varies from life-threatening mild illness to severe clinical manifestations. Recent studies reported the impairment of the central nervous system (CNS) after dengue infection, a characteristic previously considered as atypical and underreported. However, little is known about the neuropathology associated to dengue. Since animal models are important tools for helping to understand the dengue pathogenesis, including neurological damages, the aim of this work was to investigate the effects of intracerebral inoculation of a neuroadapted dengue serotype 2 virus (DENV2) in immunocompetent BALB/c mice, mimicking some aspects of the viral encephalitis. Mice presented neurological morbidity after the 7th day post infection. At the same time, histopathological analysis revealed that DENV2 led to damages in the CNS, such as hemorrhage, reactive gliosis, hyperplastic and hypertrophied microglia, astrocyte proliferation, Purkinje neurons retraction and cellular infiltration around vessels in the pia mater and in neuropil. Viral tropism and replication were detected in resident cells of the brain and cerebellum, such as neurons, astrocyte, microglia and oligodendrocytes. Results suggest that this classical mice model might be useful for analyzing the neurotropic effect of DENV with similarities to what occurs in human.


Subject(s)
Brain/virology , Dengue Virus/pathogenicity , Dengue/pathology , Encephalitis, Arbovirus/pathology , Gliosis/pathology , Virus Replication , Animals , Brain/pathology , Cells, Cultured , Dengue/virology , Dengue Virus/physiology , Encephalitis, Arbovirus/virology , Gliosis/virology , Male , Mice , Mice, Inbred BALB C , Microglia/pathology , Microglia/virology , Purkinje Cells/pathology , Purkinje Cells/virology
5.
Int J Mol Sci ; 20(7)2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30959793

ABSTRACT

Theiler's murine encephalomyelitis (TME) represents a versatile animal model for studying the pathogenesis of demyelinating diseases such as multiple sclerosis. Hallmarks of TME are demyelination, astrogliosis, as well as inflammation. Previous studies showed that matrix metalloproteinase 12 knockout (Mmp12-/-) mice display an ameliorated clinical course associated with reduced demyelination. The present study aims to elucidate the impact of MMP12 deficiency in TME with special emphasis on astrogliosis, macrophages infiltrating the central nervous system (CNS), and the phenotype of microglia/macrophages (M1 or M2). SJL wild-type and Mmp12-/- mice were infected with TME virus (TMEV) or vehicle (mock) and euthanized at 28 and 98 days post infection (dpi). Immunohistochemistry or immunofluorescence of cervical and thoracic spinal cord for detecting glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba1), chemokine receptor 2 (CCR2), CD107b, CD16/32, and arginase I was performed and quantitatively evaluated. Statistical analyses included the Kruskal⁻Wallis test followed by Mann⁻Whitney U post hoc tests. TMEV-infected Mmp12-/- mice showed transiently reduced astrogliosis in association with a strong trend (p = 0.051) for a reduced density of activated/reactive microglia/macrophages compared with wild-type mice at 28 dpi. As astrocytes are an important source of cytokine production, including proinflammatory cytokines triggering or activating phagocytes, the origin of intralesional microglia/macrophages as well as their phenotype were determined. Only few phagocytes in wild-type and Mmp12-/- mice expressed CCR2, indicating that the majority of phagocytes are represented by microglia. In parallel to the reduced density of activated/reactive microglia at 98 dpi, TMEV-infected Mmp12-/- showed a trend (p = 0.073) for a reduced density of M1 (CD16/32- and CD107b-positive) microglia, while no difference regarding the density of M2 (arginase I- and CD107b-positive) cells was observed. However, a dominance of M1 cells was detected in the spinal cord of TMEV-infected mice at all time points. Reduced astrogliosis in Mmp12-/- mice was associated with a reduced density of activated/reactive microglia and a trend for a reduced density of M1 cells. This indicates that MMP12 plays an important role in microglia activation, polarization, and migration as well as astrogliosis and microglia/astrocyte interaction.


Subject(s)
Astrocytes/pathology , Cardiovirus Infections/enzymology , Cardiovirus Infections/virology , Gliosis/enzymology , Gliosis/virology , Matrix Metalloproteinase 12/deficiency , Microglia/pathology , Spinal Cord/pathology , Animals , Cell Movement , Glial Fibrillary Acidic Protein/metabolism , Immunophenotyping , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Matrix Metalloproteinase 12/metabolism , Mice, Knockout , Phenotype
6.
Vet Microbiol ; 227: 41-44, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30473350

ABSTRACT

Atypical porcine pestivirus (APPV) has been associated with congenital tremor (CT) type A-II in newborn piglets. Although the number of APPV-based studies is increasing, the associated pathologic findings in infected piglets are underreported. This study describes the histopathologic features of spontaneous APPV infection in CT-affected piglets and complements a previous report by our group. Four two-day-old piglets with CT were evaluated by histopathology, immunohistochemistry (IHC), and molecular assay. The main histopathologic findings at the brain and spinal cord included neuronal necrosis, gliosis, neuronophagia, satellitosis, demyelination, Wallerian degeneration, and Purkinje cell necrosis. An IHC assay designed to detect the proliferation of glial fibrillary acidic protein (GFAP) in affected areas of the brain and spinal cord revealed that the proliferation of GFAP + cells and fibers was predominant in APPV-infected piglets relative to asymptomatic piglets of the same age group. The RT-nested-PCR assays identified APPV RNA in the cerebrum, cerebellum, and brainstem of all piglets; other viruses known to produce similar manifestations were not detected. These results suggest that the APPV-induced histopathologic findings are predominantly degenerative and necrotic and correlate with our previous findings. Consequently, it is proposed that neuronal necrosis, gliosis, neuronophagia, and satellitosis should be considered as important histologic features of APPV-induced infection in symptomatic CT piglets.


Subject(s)
Animals, Newborn/virology , Pestivirus Infections/veterinary , Pestivirus/genetics , Pestivirus/pathogenicity , Swine Diseases/pathology , Animals , Brain/cytology , Brain/pathology , Brain/virology , Gliosis/veterinary , Gliosis/virology , Pestivirus/isolation & purification , Pestivirus Infections/pathology , Pestivirus Infections/virology , Phylogeny , Swine , Swine Diseases/epidemiology , Swine Diseases/virology , Tremor
7.
Vet Pathol ; 54(6): 933-944, 2017 11.
Article in English | MEDLINE | ID: mdl-29065819

ABSTRACT

Feline infectious peritonitis (FIP) is a serious, widely distributed systemic disease caused by feline coronavirus (FCoV), in which ocular disease is common. However, questions remain about the patterns of ocular inflammation and the distribution of viral antigen in the eyes of cats with FIP. This study characterized the ocular lesions of FIP including the expression of glial fibrillary acidic protein and proliferating cell nuclear antigen by Müller cells in the retina in cases of FIP and to what extent macrophages are involved in ocular inflammation in FIP. Immunohistochemistry for FCoV, CD3, CD79a, glial fibrillary acidic protein, calprotectin, and proliferating cell nuclear antigen was performed on paraffin sections from 15 naturally occurring cases of FIP and from controls. Glial fibrillary acidic protein expression was increased in the retina in cases of FIP. Müller cell proliferation was present within lesions of retinal detachment. Macrophages were present in FIP-associated ocular lesions, but they were the most numerous inflammatory cells only within granulomas (2/15 cats, 13%). In cases of severe inflammation of the ciliary body with damage to blood vessel walls and ciliary epithelium (3/15, 20%), some macrophages expressed FCoV antigens, and immunolabeling for calprotectin on consecutive sections suggested that these FCoV-positive macrophages were likely to be recently derived from blood. In cases of severe and massive inflammation of most ocular structures (4/15, 26%), B cells and plasma cells predominated over T cells and macrophages. These results indicate that gliosis can be present in FIP-affected retinas and suggest that breakdown of the blood-ocular barrier can allow FCoV-bearing macrophages to access the eye.


Subject(s)
Antigens, Viral/metabolism , Coronavirus, Feline/physiology , Eye Infections, Viral/veterinary , Feline Infectious Peritonitis/pathology , Inflammation/veterinary , Animals , B-Lymphocytes/pathology , Cats , Eye/pathology , Eye/virology , Eye Infections, Viral/pathology , Eye Infections, Viral/virology , Feline Infectious Peritonitis/virology , Female , Gliosis/pathology , Gliosis/veterinary , Gliosis/virology , Immunohistochemistry/veterinary , Inflammation/pathology , Inflammation/virology , Macrophages/pathology , Male , Retinitis/pathology , Retinitis/veterinary , Retinitis/virology , T-Lymphocytes/pathology , Uveitis/pathology , Uveitis/veterinary , Uveitis/virology
8.
Development ; 144(22): 4114-4124, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28993398

ABSTRACT

The Zika virus (ZIKV) has two lineages, Asian and African, and their impact on developing brains has not been compared. Dengue virus (DENV) is a close family member of ZIKV and co-circulates with ZIKV. Here, we performed intracerebral inoculation of embryonic mouse brains with dengue virus 2 (DENV2), and found that DENV2 is sufficient to cause smaller brain size due to increased cell death in neural progenitor cells (NPCs) and neurons. Compared with the currently circulating Asian lineage of ZIKV (MEX1-44), DENV2 grows slower, causes less neuronal death and fails to cause postnatal animal death. Surprisingly, our side-by-side comparison uncovered that the African ZIKV isolate (MR-766) is more potent at causing brain damage and postnatal lethality than MEX1-44. In comparison with MEX1-44, MR-766 grows faster in NPCs and in the developing brain, and causes more pronounced cell death in NPCs and neurons, resulting in more severe neuronal loss. Together, these results reveal that DENV2 is sufficient to cause smaller brain sizes, and suggest that the ZIKV African lineage is more toxic and causes more potent brain damage than the Asian lineage.


Subject(s)
Brain/pathology , Brain/virology , Dengue Virus/pathogenicity , Phylogeny , Zika Virus/pathogenicity , Africa , Animals , Animals, Newborn , Asia , Brain/embryology , Cell Death , Cerebral Cortex/pathology , Dengue Virus/growth & development , Gliosis/pathology , Gliosis/virology , Mice, Inbred C57BL , Microcephaly/pathology , Microglia/pathology , Microglia/virology , Neural Stem Cells/pathology , Neurons/pathology , Virulence , Zika Virus/growth & development
9.
J Infect Dis ; 216(11): 1386-1397, 2017 12 12.
Article in English | MEDLINE | ID: mdl-28482001

ABSTRACT

Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne viral hemorrhagic disease seen exclusively in humans. Central nervous system (CNS) infection and neurological involvement have also been reported in CCHF. In the current study, we inoculated NSG-SGM3 mice engrafted with human hematopoietic CD34+ stem cells with low-passage CCHF virus strains isolated from human patients. In humanized mice, lethal disease develops, characterized by histopathological change in the liver and brain. To date, targets of neurological infection and disease have not been investigated in CCHF. CNS disease in humanized mice was characterized by gliosis, meningitis, and meningoencephalitis, and glial cells were identified as principal targets of infection. Humanized mice represent a novel lethal model for studies of CCHF countermeasures, and CCHF-associated CNS disease. Our data suggest a role for astrocyte dysfunction in neurological disease and identify key regions of infection in the CNS for future investigations of CCHF.


Subject(s)
Hemorrhagic Fever Virus, Crimean-Congo/pathogenicity , Hemorrhagic Fever, Crimean/pathology , Neuroglia/pathology , Neuroglia/virology , Animals , Antibodies, Viral , Brain/pathology , Cell Line , Central Nervous System/pathology , Central Nervous System/virology , Chlorocebus aethiops , Female , Gliosis/pathology , Gliosis/virology , Hematopoietic Stem Cell Transplantation , Hemorrhagic Fever, Crimean/virology , Humans , Liver/pathology , Meningitis/pathology , Meningitis/virology , Meningoencephalitis/pathology , Meningoencephalitis/virology , Mice , Tick-Borne Diseases/pathology , Vero Cells
10.
J Neuroinflammation ; 13(1): 272, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27737697

ABSTRACT

BACKGROUND: The majority of investigations on HIV-associated neurocognitive disorders (HAND) neglect the cerebellum in spite of emerging evidence for its role in higher cognitive functions and dysfunctions in common neurodegenerative diseases. METHODS: We systematically investigated the molecular and cellular responses of the cerebellum as contributors to lentiviral infection-induced neurodegeneration, in the simian immunodeficiency virus (SIV)-infected rhesus macaque model for HIV infection and HAND. Four cohorts of animals were studied: non-infected controls, SIV-infected asymptomatic animals, and SIV-infected AIDS-diseased animals with and without brain-permeant antiretroviral treatment. The antiretroviral utilized was 6-chloro-2',3'-dideoxyguanosine (6-Cl-ddG), a CNS-permeable nucleoside reverse transcriptase inhibitor. Quantitation of granule cells and Purkinje cells, of an established biomarker of SIV infection (gp41), of microglial/monocyte/macrophage markers (IBA-1, CD68, CD163), and of the astroglial marker (GFAP) were used to reveal cell-specific cerebellar responses to lentiviral infection and antiretroviral therapy (ART). The macromolecular integrity of the blood brain barrier was tested by albumin immunohistochemistry. RESULTS: Productive CNS infection was observed in the symptomatic stage of disease, and correlated with extensive microglial/macrophage and astrocyte activation, and widespread macromolecular blood brain barrier defects. Signs of productive infection, and inflammation, were reversed upon treatment with 6-Cl-ddG, except for a residual low-grade activation of microglial cells and astrocytes. There was an extensive loss of granule cells in the SIV-infected asymptomatic cohort, which was further increased in the symptomatic stage of the disease and persisted after 6-Cl-ddG (administered after the onset of symptoms of AIDS). In the symptomatic stage, Purkinje cell density was reduced. Purkinje cell loss was likewise unaffected by 6-Cl-ddG treatment at this time. CONCLUSIONS: Our findings suggest that neurodegenerative mechanisms are triggered by SIV infection early in the disease process, i. e., preceding large-scale cerebellar productive infection and marked neuroinflammation. These affect primarily granule cells early in disease, with later involvement of Purkinje cells, indicating differential vulnerability of the two neuronal populations. The results presented here indicate a role for the cerebellum in neuro-AIDS. They also support the conclusion that, in order to attenuate the development of motor and cognitive dysfunctions in HIV-positive individuals, CNS-permeant antiretroviral therapy combined with anti-inflammatory and neuroprotective treatment is indicated even before overt signs of CNS inflammation occur.


Subject(s)
Anti-Retroviral Agents/pharmacology , Anti-Retroviral Agents/therapeutic use , Cerebellum/pathology , HIV Infections/drug therapy , HIV Infections/pathology , Neurons/drug effects , Analysis of Variance , Animals , Antigens, CD/metabolism , Calcium-Binding Proteins , Case-Control Studies , DNA-Binding Proteins/metabolism , Dideoxynucleosides/pharmacology , Dideoxynucleosides/therapeutic use , Glial Fibrillary Acidic Protein/metabolism , Gliosis/etiology , Gliosis/virology , HIV Envelope Protein gp41/metabolism , Humans , Macaca mulatta , Male , Microfilament Proteins , Microglia/drug effects , Microglia/pathology , Nerve Degeneration/drug therapy , Nerve Degeneration/etiology , Nerve Degeneration/virology , Neurons/metabolism
11.
Neurobiol Dis ; 92(Pt B): 137-43, 2016 08.
Article in English | MEDLINE | ID: mdl-26851503

ABSTRACT

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


Subject(s)
Antiviral Agents/pharmacology , Encephalitis, Viral/drug therapy , HIV Infections/drug therapy , Janus Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Virus Replication/drug effects , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/virology , Disease Models, Animal , Encephalitis, Viral/metabolism , Encephalitis, Viral/pathology , Gliosis/drug therapy , Gliosis/metabolism , Gliosis/pathology , Gliosis/virology , HIV Infections/metabolism , HIV Infections/pathology , HIV-1/drug effects , HIV-1/physiology , Humans , Macrophages/drug effects , Macrophages/virology , Male , Mice , Monocytes/drug effects , Monocytes/virology , Nitriles , Pyrimidines , Tenofovir/pharmacology
12.
J Neurovirol ; 22(2): 140-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26419894

ABSTRACT

Chikungunya, "that which bends up" in the Makonde dialect, is an emerging global health threat, with increasing incidence of neurological complications. Until 2013, Chikungunya infection had been largely restricted to East Africa and the Indian Ocean, with cases within the USA reported to be from foreign travel. However, in 2014, over 1 million suspected cases were reported in the Americas, and a recently infected human could serve as an unwitting reservoir for the virus resulting in an epidemic in the continental USA. Chikungunya infection is increasingly being associated with neurological sequelae. In this study, we sought to understand the role of astrocytes in the neuropathogenesis of Chikungunya infection. Even after virus has been cleared form the circulation, astrocytes were activated with regard to TLR2 expression. In addition, white matter astrocytes were hypertrophic, with increased arbor volume in gray matter astrocytes. Combined, these would alter the number and distribution of synapses that each astrocyte would be capable of forming. These results provide the first evidence that Chikungunya infection induces morphometric and innate immune activation of astrocytes in vivo. Perturbed glia-neuron signaling could be a major driving factor in the development of Chikungunya-associated neuropathology.


Subject(s)
Astrocytes/pathology , Chikungunya Fever/pathology , Gliosis/pathology , Immunity, Innate , Neurons/pathology , Animals , Astrocytes/immunology , Astrocytes/virology , Chikungunya Fever/genetics , Chikungunya Fever/immunology , Chikungunya Fever/virology , Chikungunya virus/immunology , Chikungunya virus/pathogenicity , Disease Models, Animal , Gene Expression , Gliosis/genetics , Gliosis/immunology , Gliosis/virology , Gray Matter/immunology , Gray Matter/pathology , Gray Matter/virology , Host-Pathogen Interactions , Humans , Macaca fascicularis , Neurons/immunology , Neurons/virology , Signal Transduction , Telemetry , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/immunology , White Matter/immunology , White Matter/pathology , White Matter/virology
13.
Brain Struct Funct ; 220(3): 1395-420, 2015.
Article in English | MEDLINE | ID: mdl-24585022

ABSTRACT

The use of viruses as transneuronal tracers has become an increasingly powerful technique for defining the synaptic organization of neural networks. Although a number of recombinant alpha herpesviruses are known to spread selectively in the retrograde direction through neural circuits only one strain, the H129 strain of herpes simplex virus type 1, is reported to selectively spread in the anterograde direction. However, it is unclear from the literature whether there is an absolute block or an attenuation of retrograde spread of H129. Here, we demonstrate efficient anterograde spread, and temporally delayed retrograde spread, of H129 and three novel recombinants. In vitro studies revealed no differences in anterograde and retrograde spread of parental H129 and its recombinants through superior cervical ganglion neurons. In vivo injections of rat striatum revealed a clear bias of anterograde spread, although evidence of deficient retrograde transport was also present. Evidence of temporally delayed retrograde transneuronal spread of H129 in the retina was observed following injection of the lateral geniculate nucleus. The data also demonstrated that three novel recombinants efficiently express unique fluorescent reporters and have the capacity to infect the same neurons in dual infection paradigms. From these experiments we conclude that H129 and its recombinants not only efficiently infect neurons through anterograde transneuronal passage, but also are capable of temporally delayed retrograde transneuronal spread. In addition, the capacity to produce dual infection of projection targets following anterograde transneuronal passage provides an important addition to viral transneuronal tracing technology.


Subject(s)
Brain/cytology , Brain/virology , Herpesvirus 1, Human/physiology , Neuroanatomical Tract-Tracing Techniques/methods , Neurons/cytology , Neurons/virology , Animals , Antigens, Viral , Axonal Transport , Basal Ganglia/cytology , Basal Ganglia/virology , Cells, Cultured , Chlorocebus aethiops , Genes, Reporter , Gliosis/pathology , Gliosis/virology , Humans , Male , Rats , Rats, Sprague-Dawley , Visual Pathways/cytology , Visual Pathways/virology
14.
J Med Primatol ; 43(6): 468-76, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25082291

ABSTRACT

BACKGROUND: Simian immunodeficiency virus (SIV), a model for HIV pathogenesis, is associated with neuropathology. METHODS: Five SIV-infected animals were selected following a database search of 1206 SIV-infected animals for nodular or astrocytic lesions. Two of five had neurologic dysfunction, and 3 of 5 were incidental findings. RESULTS: Histologic examination revealed multifocal nodular foci in the gray and white matter formed by interlacing astrocytes with abundant cytoplasm and large, reactive nuclei. Nodules were often enmeshed with small capillaries. Immunohistochemistry revealed variable immunoreactivity for a panel of markers: GFAP (4/5), vimentin (5/5), Glut-1 (1/5), CNPase (0/5), S100 (5/5), Iba1 (0/5), Ki67 (0/5), and p53 (4/4). In situ hybridization failed to detect any SIV RNA (0/5). Immunohistochemistry for simian virus 40, rhesus cytomegalovirus, and rhesus lymphocryptovirus failed to detect any antigen within the lesions. CONCLUSION: The immunoreactivity of p53 in the lesions compared with adjacent tissue suggests a local derangement in astrocyte proliferation and function.


Subject(s)
Gliosis/veterinary , Macaca mulatta , Monkey Diseases/pathology , Simian Acquired Immunodeficiency Syndrome/complications , Simian Immunodeficiency Virus/physiology , Animals , Female , Gliosis/immunology , Gliosis/pathology , Gliosis/virology , Immunohistochemistry/veterinary , Male , Monkey Diseases/immunology , Monkey Diseases/virology , Retrospective Studies , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
15.
Dev Med Child Neurol ; 52(10): 901-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20561004

ABSTRACT

Subacute sclerosing panencephalitis (SSPE) is a chronic encephalitis occurring after infection with measles virus. The prevalence of the disease varies depending on uptake of measles vaccination, with the virus disproportionally affecting regions with low vaccination rates. The physiopathology of the disease is not fully understood; however, there is evidence that it involves factors that favour humoral over cellular immune response against the virus. As a result, the virus is able to infect the neurons and to survive in a latent form for years. The clinical manifestations occur, on average, 6 years after measles virus infection. The onset of SSPE is insidious, and psychiatric manifestations are prominent. Subsequently, myoclonic seizures usually lead to a final stage of akinetic mutism. The diagnosis is clinical, supported by periodic complexes on electroencephalography, brain imaging suggestive of demyelination, and immunological evidence of measles infection. Management of the disease includes seizure control and avoidance of secondary complications associated with the progressive disability. Trials of treatment with interferon, ribavirin, and isoprinosine using different methodologies have reported beneficial results. However, the disease shows relentless progression; only 5% of individuals with SSPE undergo spontaneous remission, with the remaining 95% dying within 5 years of diagnosis.


Subject(s)
Antiviral Agents/therapeutic use , Brain/pathology , Brain/virology , Measles virus/isolation & purification , Measles/complications , Subacute Sclerosing Panencephalitis , Antibodies, Viral/isolation & purification , Apoptosis , Brain/physiopathology , Demyelinating Diseases/virology , Disease Progression , Drug Therapy, Combination , Electroencephalography , Epilepsies, Myoclonic/virology , Female , Gliosis/virology , Humans , Inosine Pranobex/therapeutic use , Interferon-alpha/therapeutic use , Magnetic Resonance Imaging , Male , Measles Vaccine/administration & dosage , Measles virus/immunology , Ribavirin/therapeutic use , Severity of Illness Index , Sex Factors , Subacute Sclerosing Panencephalitis/cerebrospinal fluid , Subacute Sclerosing Panencephalitis/diagnosis , Subacute Sclerosing Panencephalitis/drug therapy , Subacute Sclerosing Panencephalitis/epidemiology , Subacute Sclerosing Panencephalitis/prevention & control , Subacute Sclerosing Panencephalitis/virology , Time Factors , Virion/drug effects
16.
J Clin Exp Neuropsychol ; 32(5): 522-7, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19844819

ABSTRACT

Commensurate with the hypothesized neural dissociation between verb and noun generation, research in HIV infection shows that, relative to noun fluency, action (verb) fluency is disproportionately impaired, more strongly related to executive dysfunction, and more sensitive to declines in everyday functioning. However, whether the neurobiological correlates of HIV-associated deficits in verb and noun generation are separable have not heretofore been investigated. The present study examined the biomarker correlates of action and noun fluency in 74 participants with HIV infection. Biomarkers of viral burden, neuroaxonal damage, macrophage activation, neuroprotection, inflammation, and astrocytosis were measured in plasma and cerebrospinal fluid (CSF). Deficits in action, but not noun generation, were significantly associated with higher CSF levels of S100beta, a marker of astrocyte activation, even after controlling for antiretroviral therapy, current immune compromise, and general cognitive impairment. Concurrent validity for the frontal systems hypothesis of verb generation was provided by post hoc analyses demonstrating that S100beta was also associated with measures of executive functions, but not semantic memory or psychomotor speed. Overall, these findings suggest that HIV-associated impairment in action fluency, and executive dysfunction more generally, may reflect astrocytosis (i.e., elevated S100beta). Complementing the literature in HIV and other clinical populations with frontal systems involvement, these data also support the possible neurobiological dissociation of noun and verb generation.


Subject(s)
Gliosis/etiology , HIV Infections/complications , Language Disorders/etiology , Verbal Behavior/physiology , Adult , CD4 Antigens/metabolism , Chemokine CCL2/metabolism , Chemokine CXCL10/metabolism , Executive Function/physiology , Female , Gliosis/virology , Humans , Language Tests , Male , Middle Aged , Nerve Growth Factors/metabolism , Neuropsychological Tests , S100 Calcium Binding Protein beta Subunit , S100 Proteins/metabolism , Statistics, Nonparametric , tau Proteins/metabolism
17.
Brain Res ; 1286: 174-84, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19523933

ABSTRACT

The ts1 mutant of the Moloney murine leukemia virus (MoMuLV) causes neurodegeneration in infected mice that resembles HIV-associated dementia. We have shown previously that ts1 infects glial cells in the brain, but not neurons. The most likely mechanism for ts1-mediated neurodegeneration is loss of glial redox support and glial cell toxicity to neurons. Minocycline has been shown to have neuroprotective effects in various models of neurodegeneration. This study was designed to determine whether and how minocycline prevents paralysis and death in ts1-infected mice. We show here that minocycline delays neurodegeneration in ts1-infected mice, and that it prevents death of cultured astrocytes infected by ts1 through attenuating oxidative stress, inflammation and apoptosis. Although minocycline reduces virus titers in the CNS of infected mice, it does not affect virus titers in infected mice thymi, spleens or infected C1 astrocytes. In addition, minocycline prevents death of primary neurons when they are cocultured with ts1-infected astrocytes, through mechanisms involving both inhibition of oxidative stress and upregulation of the transcription factor NF-E2-related factor 2 (Nrf2), which controls cellular antioxidant defenses. We conclude that minocycline delays retrovirus ts1-induced neurodegeneration involving antioxidant, anti-inflammation and anti-apoptotic mechanisms.


Subject(s)
Apoptosis/drug effects , Inflammation/drug therapy , Minocycline/pharmacology , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/virology , Blotting, Western , Brain Stem/drug effects , Brain Stem/pathology , Brain Stem/virology , Cells, Cultured , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Gliosis/drug therapy , Gliosis/pathology , Gliosis/virology , Immunohistochemistry , Inflammation/virology , Mice , Moloney murine leukemia virus , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Nerve Degeneration/virology , Neurons/drug effects , Neurons/pathology , Neurons/virology , Reactive Oxygen Species/metabolism , Retroviridae Infections/complications , Retroviridae Infections/pathology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/virology , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Up-Regulation
18.
J Neuroinflammation ; 5: 50, 2008 Nov 11.
Article in English | MEDLINE | ID: mdl-19014432

ABSTRACT

Neonatal Borna disease virus (BDV) infection of the rat brain is associated with microglial activation and damage to certain neuronal populations. Since persistent BDV infection of neurons is nonlytic in vitro, activated microglia have been suggested to be responsible for neuronal cell death in vivo. However, the mechanisms of activation of microglia in neonatally BDV-infected rat brains remain unclear. Our previous studies have shown that activation of microglia by BDV in culture requires the presence of astrocytes as neither the virus nor BDV-infected neurons alone activate microglia. Here, we evaluated the mechanisms whereby astrocytes can contribute to activation of microglia in neuron-glia-microglia mixed cultures. We found that persistent infection of neuronal cells leads to activation of uninfected astrocytes as measured by elevated expression of RANTES. Activation of astrocytes then produces activation of microglia as evidenced by increased formation of round-shaped, MHCI-, MHCII- and IL-6-positive microglia cells. Our analysis of possible molecular mechanisms of activation of astrocytes and/or microglia in culture indicates that the mediators of activation may be soluble heat-resistant, low molecular weight factors. The findings indicate that astrocytes may mediate activation of microglia by BDV-infected neurons. The data are consistent with the hypothesis that microglia activation in the absence of neuronal damage may represent initial steps in the gradual neurodegeneration observed in brains of neonatally BDV-infected rats.


Subject(s)
Astrocytes/immunology , Borna Disease/physiopathology , Brain/physiopathology , Encephalitis/physiopathology , Gliosis/physiopathology , Microglia/immunology , Animals , Astrocytes/virology , Biomarkers/analysis , Biomarkers/metabolism , Borna Disease/immunology , Borna Disease/pathology , Borna disease virus/physiology , Brain/immunology , Brain/virology , Cells, Cultured , Chemokine CCL5/immunology , Chemokine CCL5/metabolism , Chronic Disease , Coculture Techniques , Encephalitis/immunology , Encephalitis/virology , Gliosis/immunology , Gliosis/virology , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Microglia/virology , Nerve Degeneration/immunology , Nerve Degeneration/physiopathology , Nerve Degeneration/virology , Neurons/immunology , Neurons/pathology , Neurons/virology , Rats , Rats, Inbred F344
19.
Cell Biol Int ; 32(12): 1506-13, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801452

ABSTRACT

Astrocytes become activated in response to many CNS pathologies. The process of astrocyte activation remains rather enigmatic and results in so-called reactive gliosis, a reaction with specific structural and functional characteristics. Astrocytes play a vital role in regulating aspects of inflammation and in the homeostatic maintenance of the CNS. However, the responses of different human astroglial cell-lines in viral encephalitis mediated inflammation are not well documented. We have shown that Japanese encephalitis virus (JEV) infection causes morphological and functional changes in astrocytic cell-lines. We have demonstrated that besides reactive oxygen species (ROS) JEV infection differentially regulated the induction pattern of IL-6, IL-1 beta and IL-8. IP-10, MCP-1, MIG and RANTES secretions in different astroglial cell-lines. The expression of different proteins such as astrocyte-specific glial fibrillary acidic protein (GFAP), the glutamate aspartate transporter/essential amino acid transporter-1 (GLAST/EAAT-1), glutamate transporter-1/essential amino acid transporter-2 (GLT-1/EAAT-2), Ceruloplasmin and Thioredoxin (TRX) expression level also differ in different human astrocyte cell-lines following infection.


Subject(s)
Astrocytes/immunology , Brain/immunology , Encephalitis, Japanese/immunology , Gliosis/immunology , Oxidative Stress/immunology , Animals , Animals, Newborn , Astrocytes/virology , Astrocytoma/immunology , Brain/physiopathology , Brain/virology , Brain Neoplasms/immunology , Cell Line, Transformed , Cell Line, Tumor , Ceruloplasmin/immunology , Ceruloplasmin/metabolism , Chemokines/immunology , Chemokines/metabolism , Cytokines/immunology , Cytokines/metabolism , Encephalitis Virus, Japanese/immunology , Encephalitis, Japanese/physiopathology , Gliosis/physiopathology , Gliosis/virology , Humans , Mice , Mice, Inbred BALB C , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Thioredoxins/immunology , Thioredoxins/metabolism , Vesicular Glutamate Transport Proteins/immunology , Vesicular Glutamate Transport Proteins/metabolism
20.
J Neurochem ; 103(3): 1184-95, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17680994

ABSTRACT

The canine distemper virus (CDV) belongs to the Morbillivirus genus which includes important human pathogens like the closely related measles virus. CDV infection can reach the nervous system where it causes serious malfunctions. Although this pathology is well described, the molecular events in brain infection are still poorly understood. Here we studied infection in vitro by CDV using a model of dissociated cell cultures from newborn rat hippocampus. We used a recombinant CDV closely related to the neurovirulent A75/17 which also expresses the enhanced green fluorescent protein. We found that infected neurons and astrocytes could be clearly detected, and that infection spreads only slowly to neighboring cells. Interestingly, this infection causes a massive cell death of neurons, which includes also non-infected neurons. Antagonists of NMDA-type or alpha-amino-3-hydroxy-5-methylisoxazole-4-propinate (AMPA)-type glutamate receptors could slow down this neuron loss, indicating an involvement of the glutamatergic system in the induction of cell death in infected and non-infected cells. Finally, we show that, following CDV infection, there is a steady increase in extracellular glutamate in infected cultures. These results indicate that CDV infection induces excitotoxic insults on neurons via glutamatergic signaling.


Subject(s)
Distemper/metabolism , Glutamic Acid/metabolism , Hippocampus/metabolism , Hippocampus/virology , Nerve Degeneration/metabolism , Nerve Degeneration/virology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/virology , Cell Death/physiology , Cells, Cultured , Chlorocebus aethiops , Distemper/physiopathology , Distemper Virus, Canine/pathogenicity , Distemper Virus, Canine/physiology , Excitatory Amino Acid Antagonists/pharmacology , Extracellular Fluid/metabolism , Gliosis/metabolism , Gliosis/physiopathology , Gliosis/virology , Green Fluorescent Proteins , Hippocampus/physiopathology , Nerve Degeneration/physiopathology , Neurons/metabolism , Neurons/virology , Rats , Receptors, Glutamate/drug effects , Receptors, Glutamate/metabolism , Recombinant Proteins , Up-Regulation/physiology , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...