Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Neurovirol ; 30(3): 303-315, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38943022

RESUMO

Although previous studies have suggested that subtype B HIV-1 proviruses in the brain are associated with physiological changes and immune activation accompanied with microgliosis and astrogliosis, and indicated that both HIV-1 subtype variation and geographical location might influence the neuropathogenicity of HIV-1 in the brain. The natural course of neuropathogenesis of the most widespread subtype C HIV-1 has not been adequately investigated, especially for people living with HIV (PLWH) in sub-Saharan Africa. To characterize the natural neuropathology of subtype C HIV-1, postmortem frontal lobe and basal ganglia tissues were collected from nine ART-naïve individuals who died of late-stage AIDS with subtype C HIV-1 infection, and eight uninfected deceased individuals as controls. Histological staining was performed on all brain tissues to assess brain pathologies. Immunohistochemistry (IHC) against CD4, p24, Iba-1, GFAP, and CD8 in all brain tissues was conducted to evaluate potential viral production and immune activation. Histological results showed mild perivascular cuffs of lymphocytes only in a minority of the infected individuals. Viral capsid p24 protein was only detected in circulating immune cells of one infected individual, suggesting a lack of productive HIV-1 infection of the brain even at the late-stage of AIDS. Notably, similar levels of Iba-1 or GFAP between HIV + and HIV- brain tissues indicated a lack of microgliosis and astrogliosis, respectively. Similar levels of CD8 + cytotoxic T lymphocyte (CTL) infiltration between HIV + and HIV- brain tissues indicated CTL were not likely to be involved within subtype C HIV-1 infected participants of this cohort. Results from this subtype C HIV-1 study suggest that there is a lack of productive infection and limited neuropathogenesis by subtype C HIV-1 even at late-stage disease, which is in contrast to what was reported for subtype B HIV-1 by other investigators.


Assuntos
Proteína Glial Fibrilar Ácida , HIV-1 , Humanos , HIV-1/imunologia , HIV-1/patogenicidade , Masculino , Feminino , Adulto , Pessoa de Meia-Idade , África Subsaariana , Proteína Glial Fibrilar Ácida/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , Infecções por HIV/patologia , Gânglios da Base/imunologia , Gânglios da Base/patologia , Gânglios da Base/virologia , Proteínas de Ligação ao Cálcio/imunologia , Proteínas de Ligação ao Cálcio/genética , Lobo Frontal/imunologia , Lobo Frontal/patologia , Lobo Frontal/virologia , Proteína do Núcleo p24 do HIV/imunologia , Complexo AIDS Demência/imunologia , Complexo AIDS Demência/patologia , Complexo AIDS Demência/virologia , Antígenos CD4/imunologia , Linfócitos T CD8-Positivos/imunologia , Gliose/imunologia , Gliose/patologia , Gliose/virologia , Astrócitos/imunologia , Astrócitos/patologia , Astrócitos/virologia , Encéfalo/patologia , Encéfalo/imunologia , Encéfalo/virologia , Proteínas dos Microfilamentos
2.
Development ; 144(22): 4114-4124, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28993398

RESUMO

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.


Assuntos
Encéfalo/patologia , Encéfalo/virologia , Vírus da Dengue/patogenicidade , Filogenia , Zika virus/patogenicidade , África , Animais , Animais Recém-Nascidos , Ásia , Encéfalo/embriologia , Morte Celular , Córtex Cerebral/patologia , Vírus da Dengue/crescimento & desenvolvimento , Gliose/patologia , Gliose/virologia , Camundongos Endogâmicos C57BL , Microcefalia/patologia , Microglia/patologia , Microglia/virologia , Células-Tronco Neurais/patologia , Neurônios/patologia , Virulência , Zika virus/crescimento & desenvolvimento
3.
J Neurovirol ; 26(6): 838-845, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32901392

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Disfunção Cognitiva/genética , Modelos Animais de Doenças , Gliose/genética , Infecções por HIV/genética , HIV-1/genética , Animais , Astrócitos/metabolismo , Astrócitos/virologia , Biomarcadores/metabolismo , Encéfalo/virologia , Disfunção Cognitiva/complicações , Disfunção Cognitiva/metabolismo , Disfunção Cognitiva/virologia , Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/complicações , Gliose/metabolismo , Gliose/virologia , Proteína do Núcleo p24 do HIV/genética , Proteína do Núcleo p24 do HIV/metabolismo , Infecções por HIV/complicações , Infecções por HIV/metabolismo , Infecções por HIV/virologia , HIV-1/metabolismo , HIV-1/patogenicidade , Humanos , Inflamação , Antígenos Comuns de Leucócito/genética , Antígenos Comuns de Leucócito/metabolismo , Macrófagos/metabolismo , Macrófagos/virologia , Masculino , Camundongos , Camundongos SCID , Microglia/metabolismo , Microglia/virologia , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/metabolismo , Neurônios/virologia , Fenótipo
4.
Int J Mol Sci ; 20(7)2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-30959793

RESUMO

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.


Assuntos
Astrócitos/patologia , Infecções por Cardiovirus/enzimologia , Infecções por Cardiovirus/virologia , Gliose/enzimologia , Gliose/virologia , Metaloproteinase 12 da Matriz/deficiência , Microglia/patologia , Medula Espinal/patologia , Animais , Movimento Celular , Proteína Glial Fibrilar Ácida/metabolismo , Imunofenotipagem , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/metabolismo , Metaloproteinase 12 da Matriz/metabolismo , Camundongos Knockout , Fenótipo
5.
J Infect Dis ; 216(11): 1386-1397, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-28482001

RESUMO

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.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo/patogenicidade , Febre Hemorrágica da Crimeia/patologia , Neuroglia/patologia , Neuroglia/virologia , Animais , Anticorpos Antivirais , Encéfalo/patologia , Linhagem Celular , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Chlorocebus aethiops , Feminino , Gliose/patologia , Gliose/virologia , Transplante de Células-Tronco Hematopoéticas , Febre Hemorrágica da Crimeia/virologia , Humanos , Fígado/patologia , Meningite/patologia , Meningite/virologia , Meningoencefalite/patologia , Meningoencefalite/virologia , Camundongos , Doenças Transmitidas por Carrapatos/patologia , Células Vero
6.
Vet Pathol ; 54(6): 933-944, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29065819

RESUMO

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.


Assuntos
Antígenos Virais/metabolismo , Coronavirus Felino/fisiologia , Infecções Oculares Virais/veterinária , Peritonite Infecciosa Felina/patologia , Inflamação/veterinária , Animais , Linfócitos B/patologia , Gatos , Olho/patologia , Olho/virologia , Infecções Oculares Virais/patologia , Infecções Oculares Virais/virologia , Peritonite Infecciosa Felina/virologia , Feminino , Gliose/patologia , Gliose/veterinária , Gliose/virologia , Imuno-Histoquímica/veterinária , Inflamação/patologia , Inflamação/virologia , Macrófagos/patologia , Masculino , Retinite/patologia , Retinite/veterinária , Retinite/virologia , Linfócitos T/patologia , Uveíte/patologia , Uveíte/veterinária , Uveíte/virologia
7.
Neurobiol Dis ; 92(Pt B): 137-43, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26851503

RESUMO

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.


Assuntos
Antivirais/farmacologia , Encefalite Viral/tratamento farmacológico , Infecções por HIV/tratamento farmacológico , Inibidores de Janus Quinases/farmacologia , Pirazóis/farmacologia , Replicação Viral/efeitos dos fármacos , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Astrócitos/virologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/virologia , Modelos Animais de Doenças , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Gliose/tratamento farmacológico , Gliose/metabolismo , Gliose/patologia , Gliose/virologia , Infecções por HIV/metabolismo , Infecções por HIV/patologia , HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/virologia , Masculino , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/virologia , Nitrilas , Pirimidinas , Tenofovir/farmacologia
8.
J Neuroinflammation ; 13(1): 272, 2016 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-27737697

RESUMO

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.


Assuntos
Antirretrovirais/farmacologia , Antirretrovirais/uso terapêutico , Cerebelo/patologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/patologia , Neurônios/efeitos dos fármacos , Análise de Variância , Animais , Antígenos CD/metabolismo , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Proteínas de Ligação a DNA/metabolismo , Didesoxinucleosídeos/farmacologia , Didesoxinucleosídeos/uso terapêutico , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/etiologia , Gliose/virologia , Proteína gp41 do Envelope de HIV/metabolismo , Humanos , Macaca mulatta , Masculino , Proteínas dos Microfilamentos , Microglia/efeitos dos fármacos , Microglia/patologia , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/virologia , Neurônios/metabolismo
9.
J Neurovirol ; 22(2): 140-8, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26419894

RESUMO

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.


Assuntos
Astrócitos/patologia , Febre de Chikungunya/patologia , Gliose/patologia , Imunidade Inata , Neurônios/patologia , Animais , Astrócitos/imunologia , Astrócitos/virologia , Febre de Chikungunya/genética , Febre de Chikungunya/imunologia , Febre de Chikungunya/virologia , Vírus Chikungunya/imunologia , Vírus Chikungunya/patogenicidade , Modelos Animais de Doenças , Expressão Gênica , Gliose/genética , Gliose/imunologia , Gliose/virologia , Substância Cinzenta/imunologia , Substância Cinzenta/patologia , Substância Cinzenta/virologia , Interações Hospedeiro-Patógeno , Humanos , Macaca fascicularis , Neurônios/imunologia , Neurônios/virologia , Transdução de Sinais , Telemetria , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Substância Branca/imunologia , Substância Branca/patologia , Substância Branca/virologia
10.
J Med Primatol ; 43(6): 468-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25082291

RESUMO

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.


Assuntos
Gliose/veterinária , Macaca mulatta , Doenças dos Macacos/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Vírus da Imunodeficiência Símia/fisiologia , Animais , Feminino , Gliose/imunologia , Gliose/patologia , Gliose/virologia , Imuno-Histoquímica/veterinária , Masculino , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Estudos Retrospectivos , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
11.
Sci Rep ; 11(1): 6357, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33737684

RESUMO

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.


Assuntos
Biomarcadores/sangue , COVID-19/sangue , Doenças do Sistema Nervoso Central/virologia , Proteoma , Idoso , COVID-19/complicações , Estudos de Casos e Controles , Estudos de Coortes , Feminino , Perfilação da Expressão Gênica , Gliose/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/sangue
12.
Dev Med Child Neurol ; 52(10): 901-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20561004

RESUMO

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.


Assuntos
Antivirais/uso terapêutico , Encéfalo/patologia , Encéfalo/virologia , Vírus do Sarampo/isolamento & purificação , Sarampo/complicações , Panencefalite Esclerosante Subaguda , Anticorpos Antivirais/isolamento & purificação , Apoptose , Encéfalo/fisiopatologia , Doenças Desmielinizantes/virologia , Progressão da Doença , Quimioterapia Combinada , Eletroencefalografia , Epilepsias Mioclônicas/virologia , Feminino , Gliose/virologia , Humanos , Inosina Pranobex/uso terapêutico , Interferon-alfa/uso terapêutico , Imageamento por Ressonância Magnética , Masculino , Vacina contra Sarampo/administração & dosagem , Vírus do Sarampo/imunologia , Ribavirina/uso terapêutico , Índice de Gravidade de Doença , Fatores Sexuais , Panencefalite Esclerosante Subaguda/líquido cefalorraquidiano , Panencefalite Esclerosante Subaguda/diagnóstico , Panencefalite Esclerosante Subaguda/tratamento farmacológico , Panencefalite Esclerosante Subaguda/epidemiologia , Panencefalite Esclerosante Subaguda/prevenção & controle , Panencefalite Esclerosante Subaguda/virologia , Fatores de Tempo , Vírion/efeitos dos fármacos
13.
Viruses ; 12(9)2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32906600

RESUMO

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.


Assuntos
Infecções por Astroviridae/virologia , Ecologia , Mamastrovirus/fisiologia , Doenças do Sistema Nervoso/virologia , Animais , Estudos de Casos e Controles , Fezes/virologia , Gliose/patologia , Gliose/virologia , Mamastrovirus/genética , Mamastrovirus/isolamento & purificação , Projetos Piloto , Suínos , Doenças dos Suínos/virologia , Viremia/veterinária , Eliminação de Partículas Virais
14.
Sci Rep ; 9(1): 17926, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31784616

RESUMO

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.


Assuntos
Encéfalo/virologia , Vírus da Dengue/patogenicidade , Dengue/patologia , Encefalite por Arbovirus/patologia , Gliose/patologia , Replicação Viral , Animais , Encéfalo/patologia , Células Cultivadas , Dengue/virologia , Vírus da Dengue/fisiologia , Encefalite por Arbovirus/virologia , Gliose/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microglia/patologia , Microglia/virologia , Células de Purkinje/patologia , Células de Purkinje/virologia
15.
J Neuroinflammation ; 5: 50, 2008 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19014432

RESUMO

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.


Assuntos
Astrócitos/imunologia , Doença de Borna/fisiopatologia , Encéfalo/fisiopatologia , Encefalite/fisiopatologia , Gliose/fisiopatologia , Microglia/imunologia , Animais , Astrócitos/virologia , Biomarcadores/análise , Biomarcadores/metabolismo , Doença de Borna/imunologia , Doença de Borna/patologia , Vírus da Doença de Borna/fisiologia , Encéfalo/imunologia , Encéfalo/virologia , Células Cultivadas , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Doença Crônica , Técnicas de Cocultura , Encefalite/imunologia , Encefalite/virologia , Gliose/imunologia , Gliose/virologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Interleucina-6/imunologia , Interleucina-6/metabolismo , Microglia/virologia , Degeneração Neural/imunologia , Degeneração Neural/fisiopatologia , Degeneração Neural/virologia , Neurônios/imunologia , Neurônios/patologia , Neurônios/virologia , Ratos , Ratos Endogâmicos F344
16.
Cell Biol Int ; 32(12): 1506-13, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18801452

RESUMO

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.


Assuntos
Astrócitos/imunologia , Encéfalo/imunologia , Encefalite Japonesa/imunologia , Gliose/imunologia , Estresse Oxidativo/imunologia , Animais , Animais Recém-Nascidos , Astrócitos/virologia , Astrocitoma/imunologia , Encéfalo/fisiopatologia , Encéfalo/virologia , Neoplasias Encefálicas/imunologia , Linhagem Celular Transformada , Linhagem Celular Tumoral , Ceruloplasmina/imunologia , Ceruloplasmina/metabolismo , Quimiocinas/imunologia , Quimiocinas/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/fisiopatologia , Gliose/fisiopatologia , Gliose/virologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Espécies Reativas de Oxigênio/imunologia , Espécies Reativas de Oxigênio/metabolismo , Tiorredoxinas/imunologia , Tiorredoxinas/metabolismo , Proteínas Vesiculares de Transporte de Glutamato/imunologia , Proteínas Vesiculares de Transporte de Glutamato/metabolismo
17.
Vet Microbiol ; 227: 41-44, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30473350

RESUMO

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.


Assuntos
Animais Recém-Nascidos/virologia , Infecções por Pestivirus/veterinária , Pestivirus/genética , Pestivirus/patogenicidade , Doenças dos Suínos/patologia , Animais , Encéfalo/citologia , Encéfalo/patologia , Encéfalo/virologia , Gliose/veterinária , Gliose/virologia , Pestivirus/isolamento & purificação , Infecções por Pestivirus/patologia , Infecções por Pestivirus/virologia , Filogenia , Suínos , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/virologia , Tremor
18.
Brain Res ; 1134(1): 214-9, 2007 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-17189621

RESUMO

Clinical and preclinical evidence suggests that cocaine exposure hastens progression of the HIV disease process. An established active, euphoric dose of cocaine (20 mg/kg) was administered to SCID mice according to a regimen consistent with exposure to the drug by cocaine-abusing HIV-infected patients to determine the effects of cocaine on four previously established pathological characteristics of HIV encephalitis: cognitive deficits, fatigue, astrogliosis, and microgliosis. Mice were intracranially inoculated with either HIV-infected, or uninfected macrophages and then injected with either cocaine or saline in a 2 (Infection)x2 (Cocaine) factorial design. Cognition was assessed by acquisition and retention of a spatially cued learning task. Fatigue was assessed by monitoring motor activity following a 2 min forced swim. Mice were then sacrificed to determine the extent of astrogliosis and microgliosis in the four groups. Results indicated that in comparison to uninfected controls, HIV positive mice had increased astrogliosis and microgliosis, cognitive deficits, and recovered more slowly from fatigue. However, despite evidence that the cocaine exposure regimen activated the central nervous system and had long-term CNS effects, the drug did not alter the behavioral or the neuropathological deficits noted in HIV-infected SCID mice.


Assuntos
Complexo AIDS Demência/induzido quimicamente , Complexo AIDS Demência/fisiopatologia , Encéfalo/efeitos dos fármacos , Transtornos Relacionados ao Uso de Cocaína/complicações , Transtornos Relacionados ao Uso de Cocaína/fisiopatologia , Cocaína/efeitos adversos , Complexo AIDS Demência/patologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Astrócitos/virologia , Encéfalo/patologia , Encéfalo/fisiopatologia , Doença Crônica , Transtornos Relacionados ao Uso de Cocaína/patologia , Transtornos Cognitivos/induzido quimicamente , Transtornos Cognitivos/fisiopatologia , Transtornos Cognitivos/virologia , Modelos Animais de Doenças , Inibidores da Captação de Dopamina/efeitos adversos , Fadiga/induzido quimicamente , Fadiga/fisiopatologia , Fadiga/virologia , Gliose/induzido quimicamente , Gliose/fisiopatologia , Gliose/virologia , Masculino , Camundongos , Camundongos SCID , Microglia/efeitos dos fármacos , Microglia/patologia , Microglia/virologia
19.
Neurosci Lett ; 414(1): 45-50, 2007 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-17287085

RESUMO

Chemokines and their receptors are important elements for the selective attraction and activation of various subsets of leukocytes. Interferon-gamma inducible protein (IP-10 or CXCL-10) is a potent chemoattractant and has been suggested to enhance the severity of virus infection and neuronal injury. In order to assess functional importance of this chemokine in viral encephalitis, we have exploited an experimental model of Japanese encephalitis. We report for the first time that in Japanese encephalitis, astrocytes are the predominant source of IP-10. A progressive increase in IP-10 induction following viral infection is concomitant with the increase in IFN-gamma a known inducer of IP-10. However, this increase in IFN-gamma level is not sufficient to confer protection as animals eventually succumb to the infection.


Assuntos
Astrócitos/imunologia , Encéfalo/imunologia , Quimiocinas CXC/biossíntese , Quimiotaxia/imunologia , Encefalite Japonesa/imunologia , Animais , Astrócitos/metabolismo , Encéfalo/fisiopatologia , Encéfalo/virologia , Linhagem Celular , Quimiocina CXCL10 , Quimiocinas CXC/genética , Encefalite Japonesa/patologia , Encefalite Japonesa/fisiopatologia , Feminino , Gliose/imunologia , Gliose/fisiopatologia , Gliose/virologia , Humanos , Imunidade Inata/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/biossíntese , Regulação para Cima/imunologia
20.
Neurosci Lett ; 420(2): 144-9, 2007 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-17531383

RESUMO

The anti-inflammatory cytokine interleukin (IL)-10 is synthesized in the central nervous system (CNS) and acts to limit clinical symptoms of stroke, multiple sclerosis, Alzheimer's disease, meningitis, and the behavioral changes that occur during bacterial infections. Expression of IL-10 is critical during the course of most major diseases in the CNS and promotes survival of neurons and all glial cells in the brain by blocking the effects of proinflammatory cytokines and by promoting expression of cell survival signals. In order to assess functional importance of this cytokine in viral encephalitis we have exploited an experimental model of Japanese encephalitis (JE). We report for the first time that in Japanese encephalitis, there is a progressive decline in level of IL-10. The extent of progressive decrease in IL-10 level following viral infection is inversely proportional to the increase in the level of proinflammatory cytokines as well as negative consequences that follows viral infection.


Assuntos
Encéfalo/imunologia , Encefalite Japonesa/imunologia , Gliose/imunologia , Interleucina-10/imunologia , Microglia/imunologia , Degeneração Neural/imunologia , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Encéfalo/metabolismo , Encéfalo/virologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Encefalite/imunologia , Encefalite/metabolismo , Encefalite/virologia , Vírus da Encefalite Japonesa (Espécie)/patogenicidade , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/metabolismo , Encefalite Japonesa/fisiopatologia , Feminino , Gliose/induzido quimicamente , Gliose/virologia , Interleucina-10/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-1beta/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Microglia/metabolismo , Microglia/virologia , Degeneração Neural/metabolismo , Degeneração Neural/virologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA