Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 241
Filtrar
1.
Sci Rep ; 14(1): 18002, 2024 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-39097642

RESUMEN

Zika virus (ZIKV) infection was first reported in 2015 in Brazil as causing microcephaly and other developmental abnormalities in newborns, leading to the identification of Congenital Zika Syndrome (CZS). Viral infections have been considered an environmental risk factor for neurodevelopmental disorders outcome, such as Autism Spectrum Disorder (ASD). Moreover, not only the infection per se, but maternal immune system activation during pregnancy, has been linked to fetal neurodevelopmental disorders. To understand the impact of ZIKV vertical infection on brain development, we derived induced pluripotent stem cells (iPSC) from Brazilian children born with CZS, some of the patients also being diagnosed with ASD. Comparing iPSC-derived neurons from CZS with a control group, we found lower levels of pre- and postsynaptic proteins and reduced functional synapses by puncta co-localization. Furthermore, neurons and astrocytes derived from the CZS group showed decreased glutamate levels. Additionally, the CZS group exhibited elevated levels of cytokine production, one of which being IL-6, already associated with the ASD phenotype. These preliminary findings suggest that ZIKV vertical infection may cause long-lasting disruptions in brain development during fetal stages, even in the absence of the virus after birth. These disruptions could contribute to neurodevelopmental disorders manifestations such as ASD. Our study contributes with novel knowledge of the CZS outcomes and paves the way for clinical validation and the development of potential interventions to mitigate the impact of ZIKV vertical infection on neurodevelopment.


Asunto(s)
Encéfalo , Células Madre Pluripotentes Inducidas , Transmisión Vertical de Enfermedad Infecciosa , Sinapsis , Infección por el Virus Zika , Virus Zika , Humanos , Infección por el Virus Zika/virología , Infección por el Virus Zika/patología , Femenino , Virus Zika/patogenicidad , Sinapsis/patología , Sinapsis/metabolismo , Encéfalo/virología , Encéfalo/patología , Embarazo , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/virología , Neuronas/virología , Neuronas/metabolismo , Neuronas/patología , Masculino , Astrocitos/virología , Astrocitos/metabolismo , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/metabolismo , Complicaciones Infecciosas del Embarazo/virología , Complicaciones Infecciosas del Embarazo/patología , Brasil , Recién Nacido , Trastorno del Espectro Autista/virología , Niño
2.
Commun Biol ; 7(1): 811, 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38965360

RESUMEN

Experimental autoimmune encephalomyelitis (EAE) is a demyelinating disease affecting the central nervous system (CNS) in animals that parallels several clinical and molecular traits of multiple sclerosis in humans. Herpes simplex virus type 1 (HSV-1) infection mainly causes cold sores and eye diseases, yet eventually, it can also reach the CNS, leading to acute encephalitis. Notably, a significant proportion of healthy individuals are likely to have asymptomatic HSV-1 brain infection with chronic brain inflammation due to persistent latent infection in neurons. Because cellular senescence is suggested as a potential factor contributing to the development of various neurodegenerative disorders, including multiple sclerosis, and viral infections may induce a premature senescence state in the CNS, potentially increasing susceptibility to such disorders, here we examine the presence of senescence-related markers in the brains and spinal cords of mice with asymptomatic HSV-1 brain infection, EAE, and both conditions. Across all scenarios, we find a significant increases of senescence biomarkers in the CNS with some differences depending on the analyzed group. Notably, some senescence biomarkers are exclusively observed in mice with the combined conditions. These results indicate that asymptomatic HSV-1 brain infection and EAE associate with a significant expression of senescence biomarkers in the CNS.


Asunto(s)
Encéfalo , Senescencia Celular , Herpes Simple , Herpesvirus Humano 1 , Esclerosis Múltiple , Animales , Ratones , Encéfalo/virología , Encéfalo/patología , Encéfalo/metabolismo , Esclerosis Múltiple/virología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/patogenicidad , Herpes Simple/virología , Herpes Simple/patología , Femenino , Ratones Endogámicos C57BL , Encefalomielitis Autoinmune Experimental/virología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Fenotipo , Sistema Nervioso Central/virología , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Médula Espinal/virología , Médula Espinal/metabolismo , Médula Espinal/patología , Biomarcadores/metabolismo , Encefalitis por Herpes Simple/virología , Encefalitis por Herpes Simple/patología , Encefalitis por Herpes Simple/metabolismo
4.
Methods Mol Biol ; 2808: 177-195, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743371

RESUMEN

We describe the use of conventional histology and immunohistochemistry against canine distemper virus (CDV) to examine the brains of domestic dogs with a confirmed diagnosis of CDV infection. Histologically, to identify the main typical lesions, we used conventional H&E stain; to evaluate the progressive demyelination, we used Luxol Fast Blue stain; and to identify the presence of viral particles in these affected regions, we used immunohistochemistry against CDV. We confirm that the histopathological analysis of brains of distemper-infected dogs is a powerful tool to evaluate the typical brain lesions and could be used as an interesting natural model to continue studying the pathogenesis of canine distemper in different species and/or other morbillivirus infections, like measles.


Asunto(s)
Encéfalo , Virus del Moquillo Canino , Moquillo , Inmunohistoquímica , Animales , Virus del Moquillo Canino/patogenicidad , Moquillo/virología , Moquillo/patología , Perros , Encéfalo/virología , Encéfalo/patología , Inmunohistoquímica/métodos
5.
Proc Natl Acad Sci U S A ; 119(35): e2200960119, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-35951647

RESUMEN

Although increasing evidence confirms neuropsychiatric manifestations associated mainly with severe COVID-19 infection, long-term neuropsychiatric dysfunction (recently characterized as part of "long COVID-19" syndrome) has been frequently observed after mild infection. We show the spectrum of cerebral impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, ranging from long-term alterations in mildly infected individuals (orbitofrontal cortical atrophy, neurocognitive impairment, excessive fatigue and anxiety symptoms) to severe acute damage confirmed in brain tissue samples extracted from the orbitofrontal region (via endonasal transethmoidal access) from individuals who died of COVID-19. In an independent cohort of 26 individuals who died of COVID-19, we used histopathological signs of brain damage as a guide for possible SARS-CoV-2 brain infection and found that among the 5 individuals who exhibited those signs, all of them had genetic material of the virus in the brain. Brain tissue samples from these five patients also exhibited foci of SARS-CoV-2 infection and replication, particularly in astrocytes. Supporting the hypothesis of astrocyte infection, neural stem cell-derived human astrocytes in vitro are susceptible to SARS-CoV-2 infection through a noncanonical mechanism that involves spike-NRP1 interaction. SARS-CoV-2-infected astrocytes manifested changes in energy metabolism and in key proteins and metabolites used to fuel neurons, as well as in the biogenesis of neurotransmitters. Moreover, human astrocyte infection elicits a secretory phenotype that reduces neuronal viability. Our data support the model in which SARS-CoV-2 reaches the brain, infects astrocytes, and consequently, leads to neuronal death or dysfunction. These deregulated processes could contribute to the structural and functional alterations seen in the brains of COVID-19 patients.


Asunto(s)
Encéfalo , COVID-19 , Enfermedades Virales del Sistema Nervioso Central , SARS-CoV-2 , Astrocitos/patología , Astrocitos/virología , Encéfalo/patología , Encéfalo/virología , COVID-19/complicaciones , COVID-19/patología , Enfermedades Virales del Sistema Nervioso Central/etiología , Enfermedades Virales del Sistema Nervioso Central/patología , Humanos , Síndrome Post Agudo de COVID-19
7.
Viruses ; 13(10)2021 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-34696533

RESUMEN

The Zika virus (ZIKV) has shown a promising oncolytic effect against embryonal CNS tumors. However, studies on the effect of different administration routes and the ideal viral load in preclinical models are highly relevant aiming for treatment safety and efficiency. Here, we investigated the effect and effectiveness of different routes of administration, and the number of ZIKVBR injections on tumor tropism, destruction, and side effects. Furthermore, we designed an early-stage human brain organoid co-cultured with embryonal CNS tumors to analyze the ZIKVBR oncolytic effect. We showed that in the mice bearing subcutaneous tumors, the ZIKVBR systemically presented a tropism to the brain. When the tumor was located in the mice's brain, serial systemic injections presented efficient tumor destruction, with no neurological or other organ injury and increased mice survival. In the human cerebral organoid model co-cultured with embryonal CNS tumor cells, ZIKVBR impaired tumor progression. The gene expression of cytokines and chemokines in both models suggested an enhancement of immune cells recruitment and tumor inflammation after the treatment. These results open new perspectives for virotherapy using the ZIKVBR systemic administration route and multiple doses of low virus load for safe and effective treatment of embryonal CNS tumors, an orphan disease that urges new effective therapies.


Asunto(s)
Neoplasias Encefálicas/terapia , Viroterapia Oncolítica/métodos , Virus Zika/metabolismo , Animales , Encéfalo/virología , Neoplasias Encefálicas/patología , Línea Celular , Sistema Nervioso Central/efectos de los fármacos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/métodos , Inyecciones Intralesiones/métodos , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Virus Oncolíticos/metabolismo , Organoides , Virus Zika/inmunología , Infección por el Virus Zika/virología
8.
Front Immunol ; 12: 702048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335614

RESUMEN

Zika virus (ZIKV) is an arbovirus belonging to Flaviviridae family that emerged as a global health threat due to its association with microcephaly and other severe neurological complications, including Guillain-Barré Syndrome (GBS) and Congenital Zika Syndrome (CZS). ZIKV disease has been linked to neuroinflammation and neuronal cell death. Neurodegenerative processes may be exacerbated by metabolites produced by the kynurenine pathway, an important pathway for the degradation of tryptophan, which induces neuronal dysfunction due to enhanced excitotoxicity. Here, we exploited the hypothesis that ZIKV-induced neurodegeneration can be rescued by blocking a target enzyme of the kynurenine pathway, the Indoleamine 2,3-dioxygenase (IDO-1). RT-PCR analysis showed increased levels of IDO-1 RNA expression in undifferentiated primary neurons isolated from wild type (WT) mice infected by ZIKV ex vivo, as well as in the brain of ZIKV-infected A129 mice. Pharmacological inhibition of IDO-1 enzyme with 1-methyl-D-tryptophan (1-MT), in both in vitro and in vivo systems, led to significant reduction of ZIKV-induced neuronal death without interfering with the ability of ZIKV to replicate in those cells. Furthermore, in vivo analyses using both genetically modified mice (IDO-/- mice) and A129 mice treated with 1-MT resulted in reduced microgliosis, astrogliosis and Caspase-3 positive cells in the brain of ZIKV-infected A129 mice. Interestingly, increased levels of CCL5 and CXCL-1 chemokines were found in the brain of 1-MT treated-mice. Together, our data indicate that IDO-1 blockade provides a neuroprotective effect against ZIKV-induced neurodegeneration, and this is amenable to inhibition by pharmacological treatment.


Asunto(s)
Neuroprotección/fisiología , Triptófano/antagonistas & inhibidores , Triptófano/metabolismo , Infección por el Virus Zika/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/virología , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Microcefalia/metabolismo , Microcefalia/virología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/virología , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/virología , Neuronas/metabolismo , Neuronas/virología , Fármacos Neuroprotectores/metabolismo , Virus Zika/patogenicidad , Infección por el Virus Zika/virología
9.
PLoS Negl Trop Dis ; 15(8): e0009575, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34351896

RESUMEN

Since the 2015 to 2016 outbreak in America, Zika virus (ZIKV) infected almost 900,000 patients. This international public health emergency was mainly associated with a significant increase in the number of newborns with congenital microcephaly and abnormal neurologic development, known as congenital Zika syndrome (CZS). Furthermore, Guillain-Barré syndrome (GBS), a neuroimmune disorder of adults, has also been associated with ZIKV infection. Currently, the number of ZIKV-infected patients has decreased, and most of the cases recently reported present as a mild and self-limiting febrile illness. However, based on its natural history of a typical example of reemerging pathogen and the lack of specific therapeutic options against ZIKV infection, new outbreaks can occur worldwide, demanding the attention of researchers and government authorities. Here, we discuss the clinical spectrum and immunopathological mechanisms underlying ZIKV-induced neurological manifestations. Several studies have confirmed the tropism of ZIKV for neural progenitor stem cells by demonstrating the presence of ZIKV in the central nervous system (CNS) during fetal development, eliciting a deleterious inflammatory response that compromises neurogenesis and brain formation. Of note, while the neuropathology of CZS can be due to a direct viral neuropathic effect, adults may develop neuroimmune manifestations such as GBS due to poorly understood mechanisms. Antiganglioside autoantibodies have been detected in multiple patients with ZIKV infection-associated GBS, suggesting a molecular mimicry. However, further additional immunopathological mechanisms remain to be uncovered, paving the way for new therapeutic strategies.


Asunto(s)
Encéfalo/embriología , Síndrome de Guillain-Barré/virología , Microcefalia/virología , Infección por el Virus Zika/patología , Virus Zika/patogenicidad , Animales , Encéfalo/virología , Femenino , Síndrome de Guillain-Barré/etiología , Humanos , Ratones , Células-Madre Neurales/virología , Embarazo , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika/virología
10.
BMC Infect Dis ; 21(1): 311, 2021 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-33794785

RESUMEN

BACKGROUND: Dengue infection is caused by an arbovirus with a wide range of presentations, varying from asymptomatic disease to unspecific febrile illness and haemorrhagic syndrome with shock, which can evolve to death. In Brazil, the virus circulates since the 1980s with many introductions of new serotypes, genotypes, and lineages since then. Here we report a fatal case of dengue associated with a Dengue virus (DENV) lineage not detected in the country until now. CASE PRESENTATION: The patient, a 58-year-old man arrived at the hospital complaining of fever and severe abdominal pain due to intense gallbladder edema, mimicking acute abdomen. After 48 h of hospital admission, he evolved to refractory shock and death. DENV RNA was detected in all tissues collected (heart, lung, brain, kidney, spleen, pancreas, liver, and testis). Viral sequencing has shown that the virus belongs to serotype 2, American/Asian genotype, in a new clade, which has never been identified in Brazil before. The virus was phylogenetically related to isolates from central America [Puerto Rico (2005-2007), Martinique (2005), and Guadeloupe (2006)], most likely arriving in Brazil from Puerto Rico. CONCLUSION: In summary, this was the first fatal documented case with systemic dengue infection associated with the new introduction of Dengue type 2 virus in Brazil during the 2019 outbreak.


Asunto(s)
Virus del Dengue/aislamiento & purificación , Dengue/diagnóstico , Encéfalo/virología , Brasil , Dengue/virología , Virus del Dengue/clasificación , Virus del Dengue/genética , Vesícula Biliar/patología , Genotipo , Corazón/virología , Humanos , Pulmón/virología , Masculino , Persona de Mediana Edad , Filogenia , ARN Viral/metabolismo
11.
Sci Rep ; 11(1): 6099, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731800

RESUMEN

The absence of an adequate animal model for studies has limited the understanding of congenital Zika syndrome (CZS) in humans during the outbreak in America. In this study, we used squirrel monkeys (Saimiri collinsi), a neotropical primate (which mimics the stages of human pregnancy), as a model of Zika virus (ZIKV) infection. Seven pregnant female squirrel monkeys were experimentally infected at three different gestational stages, and we were able reproduce a broad range of clinical manifestations of ZIKV lesions observed in newborn humans. Histopathological and immunohistochemical analyses of early-infected newborns (2/4) revealed damage to various areas of the brain and ZIKV antigens in the cytoplasm of neurons and glial cells, indicative of CZS. The changes caused by ZIKV infection were intrauterine developmental delay, ventriculomegaly, simplified brain gyri, vascular impairment and neuroprogenitor cell dysfunction. Our data show that the ZIKV infection outcome in squirrel monkeys is similar to that in humans, indicating that this model can be used to help answer questions about the effect of ZIKV infection on neuroembryonic development and the morphological changes induced by CZS.


Asunto(s)
Encéfalo , Complicaciones Infecciosas del Embarazo , Infección por el Virus Zika , Virus Zika , Animales , Animales Recién Nacidos , Encéfalo/embriología , Encéfalo/patología , Encéfalo/virología , Femenino , Embarazo , Complicaciones Infecciosas del Embarazo/patología , Complicaciones Infecciosas del Embarazo/virología , Saimiri , Síndrome , Infección por el Virus Zika/embriología , Infección por el Virus Zika/patología
12.
J Gen Virol ; 102(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33528354

RESUMEN

Equine herpesvirus type 1 (EHV-1) is an emerging pathogen that causes encephalomyelitis in horses and non-equid species. Several aspects of the immune response in the central nervous system (CNS), mainly regarding the role of inflammatory mediators during EHV-1 encephalitis, remain unknown. Moreover, understanding the mechanisms underlying extensive neuropathology induced by viruses would be helpful to establish therapeutic strategies. Therefore, we aimed to evaluate some aspects of the innate immune response during highly neurovirulent EHV-1 infection. C57BL/6 mice infected intranasally with A4/72 and A9/92 EHV-1 strains developed a fulminant neurological disease at 3 days post-inoculation with high viral titres in the brain. These mice developed severe encephalitis with infiltration of monocytes and CD8+ T cells to the brain. The inflammatory infiltrate followed the detection of the chemokines CCL2, CCL3, CCL4, CCL5, CXCL2, CXCL9 and CXCL-10 in the brain. Notably, the levels of CCL3, CCL4, CCL5 and CXCL9 were higher in A4/72-infected mice, which presented higher numbers of inflammatory cells within the CNS. Pro-inflammatory cytokines, such as interleukins (ILs) IL-1α, IL-1ß, IL-6, IL-12ß, and tumour necrosis factor (TNF), were also detected in the CNS, and Toll-like receptor (TLR) TLR2, TLR3 and TLR9 genes were also upregulated within the brain of EHV-1-infected mice. However, no expression of interferon-γ (IFN-γ) and IL-12α, which are important for controlling the replication of other herpesviruses, was detected in EHV-1-infected mice. The results show that the activated innate immune mechanisms could not prevent EHV-1 replication within the CNS, but most likely contributed to the extensive neuropathology. The mouse model of viral encephalitis proposed here will also be useful to study the mechanisms underlying extensive neuropathology.


Asunto(s)
Encéfalo/inmunología , Encefalitis Viral/inmunología , Infecciones por Herpesviridae/inmunología , Herpesvirus Équido 1/inmunología , Herpesvirus Équido 1/patogenicidad , Animales , Encéfalo/metabolismo , Encéfalo/virología , Quimiocinas/genética , Quimiocinas/metabolismo , Citocinas/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis Viral/virología , Infecciones por Herpesviridae/virología , Inmunidad Innata , Leucocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores Toll-Like/genética , Regulación hacia Arriba , Carga Viral
13.
Virulence ; 12(1): 244-259, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33410731

RESUMEN

St. Louis encephalitis virus (SLEV) is a neglected mosquito-borne flavivirus that causes severe neurological disease in humans. SLEV replication in the central nervous system (CNS) induces the local production of interferons (IFNs), which are attributed to host protection. The antiviral response to SLEV infection in the CNS is not completely understood, which led us to characterize the roles of IFNs using mouse models of St. Louis encephalitis. We infected mice deficient in type I IFN receptor (ABR-/-) or deficient in Type II IFN (IFNγ-/-) and assessed the contribution of each pathway to disease development. We found that type I and II IFNs play different roles in SLEV infection. Deficiency in type I IFN signaling was associated to an early and increased mortality, uncontrolled SLEV replication and impaired ISG expression, leading to increased proinflammatory cytokine production and brain pathology. Conversely, IFNγ-/- mice were moderately resistant to SLEV infection. IFNγ deficiency caused no changes to viral load or SLEV-induced encephalitis and did not change the expression of ISGs in the brain. We found that type I IFN is essential for the control of SLEV replication whereas type II IFN was not associated with protection in this model.


Asunto(s)
Encéfalo/inmunología , Encéfalo/virología , Virus de la Encefalitis de San Luis/inmunología , Encefalitis de San Luis/inmunología , Interferón Tipo I/inmunología , Interferón gamma/inmunología , Animales , Encéfalo/patología , Modelos Animales de Enfermedad , Interferón Tipo I/genética , Interferón gamma/genética , Ratones , Ratones Endogámicos C57BL , Carga Viral , Replicación Viral/inmunología
14.
Virulence ; 12(1): 430-443, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33487119

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is an encephalitic alphavirus that can cause debilitating, acute febrile illness and potentially result in encephalitis. Currently, there are no FDA-licensed vaccines or specific therapeutics for VEEV. Previous studies have demonstrated that VEEV infection results in increased blood-brain barrier (BBB) permeability that is mediated by matrix metalloproteinases (MMPs). Furthermore, after subarachnoid hemorrhage in mice, MMP-9 is upregulated in the brain and mediates BBB permeability in a toll-like receptor 4 (TLR4)-dependent manner. Here, we demonstrate that disease in C3H mice during VEEV TC-83 infection is dependent on TLR4 because intranasal infection of C3H/HeN (TLR4 WT ) mice with VEEV TC-83 resulted in mortality as opposed to survival of TLR4-defective C3H/HeJ (TLR4 mut ) mice. In addition, BBB permeability was induced to a lesser extent in TLR4 mut mice compared with TLR4 WT mice during VEEV TC-83 infection as determined by sodium fluorescein and fluorescently-conjugated dextran extravasation. Moreover, MMP-9, MMP-2, ICAM-1, CCL2 and IFN-γ were all induced to significantly lower levels in the brains of infected TLR4 mut mice compared with infected TLR4 WT mice despite the absence of significantly different viral titers or immune cell populations in the brains of infected TLR4 WT and TLR4 mut mice. These data demonstrate the critical role of TLR4 in mediating BBB permeability and disease in C3H mice during VEEV TC-83 infection, which suggests that TLR4 is a potential target for the development of therapeutics for VEEV.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Virus de la Encefalitis Equina Venezolana/patogenicidad , Receptor Toll-Like 4/genética , Animales , Encéfalo/virología , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/inmunología , Encefalomielitis Equina Venezolana/virología , Femenino , Ratones , Ratones Endogámicos C3H , Permeabilidad , Receptor Toll-Like 4/metabolismo , Replicación Viral
15.
Mol Neurobiol ; 58(2): 520-535, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32978729

RESUMEN

The main discussion above of the novel pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has focused substantially on the immediate risks and impact on the respiratory system; however, the effects induced to the central nervous system are currently unknown. Some authors have suggested that SARS-CoV-2 infection can dramatically affect brain function and exacerbate neurodegenerative diseases in patients, but the mechanisms have not been entirely described. In this review, we gather information from past and actual studies on coronaviruses that informed neurological dysfunction and brain damage. Then, we analyzed and described the possible mechanisms causative of brain injury after SARS-CoV-2 infection. We proposed that potential routes of SARS-CoV-2 neuro-invasion are determinant factors in the process. We considered that the hematogenous route of infection can directly affect the brain microvascular endothelium cells that integrate the blood-brain barrier and be fundamental in initiation of brain damage. Additionally, activation of the inflammatory response against the infection represents a critical step on injury induction of the brain tissue. Consequently, the virus' ability to infect brain cells and induce the inflammatory response can promote or increase the risk to acquire central nervous system diseases. Here, we contribute to the understanding of the neurological conditions found in patients with SARS-CoV-2 infection and its association with the blood-brain barrier integrity.


Asunto(s)
Barrera Hematoencefálica/virología , Encéfalo/virología , COVID-19/complicaciones , Enfermedades del Sistema Nervioso Central/virología , Inflamación/virología , Barrera Hematoencefálica/patología , Encéfalo/patología , COVID-19/patología , Enfermedades del Sistema Nervioso Central/patología , Humanos , Inflamación/patología
16.
J Infect Dis ; 223(3): 435-440, 2021 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-32614431

RESUMEN

The recent increase in babies born with brain and eye malformations in Brazil is associated with Zika virus (ZIKV) infection in utero. ZIKV alters host DNA methylation in vitro. Using genome-wide DNA methylation profiling we compared 18 babies born with congenital ZIKV microcephaly with 20 controls. We found ZIKV-associated alteration of host methylation patterns, notably at RABGAP1L which is important in brain development, at viral host immunity genes MX1 and ISG15, and in an epigenetic module containing the causal microcephaly gene MCPH1. Our data support the hypothesis that clinical signs of congenital ZIKV are associated with changes in DNA methylation.


Asunto(s)
Metilación de ADN , Inmunidad/genética , Microcefalia/virología , Neurogénesis/genética , Infección por el Virus Zika , Encéfalo/crecimiento & desarrollo , Encéfalo/virología , Brasil , Proteínas de Ciclo Celular/genética , Preescolar , Proteínas del Citoesqueleto/genética , Femenino , Humanos , Lactante , Masculino , Embarazo , Complicaciones Infecciosas del Embarazo/virología , Virus Zika/inmunología
17.
Neurosci Lett ; 739: 135448, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33129847

RESUMEN

Zika virus (ZIKV) is a mosquito-borne Flavivirus structurally and antigenically related to Dengue virus (DENV). Zika virus has been associated with congenital anomalies and most ZIKV outbreaks have occurred in endemic areas of DENV. The present study investigated the effects of prior DENV serotype 1 (DENV1) immunity in immunocompetent female Swiss mice on gestational ZIKV infection in offspring. Physical/reflex development, locomotor activity, anxiety, visual acuity, and brain-derived neurotrophic factor (BDNF) levels were evaluated in offspring during infancy and adolescence. Anti-DENV1 and anti-ZIKV antibodies were detected in sera of the progenitors, whereas no ZIKV genomes were detected in the offspring brain. Pups from dams with only DENV1 immunity presented alterations of physical/reflex development. Pups from all infected dams exhibited time-related impairments in locomotor activity and anxiolytic-like behavior. Offspring from DENV/ZIKV-infected dams exhibited impairments in visual acuity during infancy but not during adolescence, which was consistent with morphometric analysis of the optic nerve. Pups from DENV1-, ZIKV-, and DENV/ZIKV-infected dams exhibited a decrease in BDNF levels during infancy and an increase during adolescence in distinct brain regions. In summary, we found no influence of prior DENV1 immunity on gestational ZIKV infection in offspring, with the exception of alterations of early visual parameters, and an increase in BDNF levels in the hippocampus during adolescence.


Asunto(s)
Conducta Animal , Dengue/inmunología , Complicaciones Infecciosas del Embarazo/inmunología , Complicaciones Infecciosas del Embarazo/psicología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/psicología , Animales , Encéfalo/inmunología , Encéfalo/virología , Factor Neurotrófico Derivado del Encéfalo/inmunología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Femenino , Masculino , Aprendizaje por Laberinto , Ratones , Embarazo
18.
ACS Chem Neurosci ; 11(24): 4012-4016, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33244974

RESUMEN

The current pandemic of coronavirus disease 2019 (COVID-19) has gained increased attention in the neuroscience community, especially taking into account the neuroinvasive potential of its causative agent, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the impact of its infection on the structure and function of the brain. Apart from the neurotropic properties of SARS-CoV-2, it is likewise important the observation that virus infection may perturb specific cellular processes that are believed to play an important role in the pathogenesis of diverse neurological disorders, particularly in Parkinson's disease (PD). In this scenario, viral infection-induced colon inflammation, gut microbial imbalance, and α-synuclein upregulation are of particular interest with regard to the interplay between the gastrointestinal tract and the central nervous system (microbiome-gut-brain axis). In this Perspective, we present a critical view on the different hypotheses that are recently being raised by neuroscientists about the relationship between SARS-CoV-2 infection and long-lasting neurodegenerative disorders, opening the question of whether COVID-19 might represent a risk factor for the development of PD.


Asunto(s)
Disbiosis/virología , Enfermedades Neurodegenerativas/virología , SARS-CoV-2/patogenicidad , Virosis/virología , Encéfalo/patología , Encéfalo/virología , Disbiosis/complicaciones , Microbioma Gastrointestinal/fisiología , Humanos , Enfermedades Neurodegenerativas/patología , Virosis/complicaciones , Virosis/patología
19.
Pediatr Infect Dis J ; 39(10): 894-898, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32936599

RESUMEN

BACKGROUND: Viral meningoencephalitis is highly heterogeneous, varying by geographic location. The aim of this study was to characterize the etiology and reporting the clinical findings and outcome of viral encephalitis in children in southern Brazil. METHODS: A cross-Sectional study was conducted at Hospital Pequeno Príncipe, Curitiba, Brazil, between January 2013 and December 2017. It included patients younger than 18 years, who fulfilled the criteria: altered mental status as a major criteria and 2 or more minor criteria (1) fever, (2) seizures, (3) focal neurologic findings, (4) central system fluid white cell count of ≥5 cells/mm, (5) abnormal brain imaging, and/or (6) electroencephalogram abnormalities. RESULTS: Viral meningoencephalitis was diagnosed in 270 children, with median age of 2 years (interquartile range: 0-4), The etiology of viral meningoencephalitis was confirmed in 47% of patients. Enterovirus (18%) was the major cause of encephalitis in Southern Brazilian children, and a high prevalence of Epstein-Barr virus (6%) was demonstrated. Most patients presented with fever (81%), followed by vomiting (50%), focal neurologic findings (46%), seizures (31%) and headache (30%). Few abnormalities were detected on electroencephalograms and brain magnetic resonance images. On discharge from hospital, symptoms resolved completely in 87% of children. Sequelae were mainly observed in patients with focal neurologic symptoms (P<0.001), presence of seizures (P<0.001) and electroencephalogram abnormalities (P=0.024). CONCLUSIONS: Enterovirus was the major cause of encephalitis. Etiologic agent of encephalitis seems to be influenced by the local virologic pattern. A poor outcome was identified in patients with seizures, focal neurologic findings and electroencephalogram abnormalities.


Asunto(s)
Encefalitis Viral/epidemiología , Virus/patogenicidad , Enfermedad Aguda/epidemiología , Adolescente , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Encéfalo/virología , Brasil/epidemiología , Niño , Preescolar , Estudios Transversales , Electroencefalografía , Encefalitis Viral/complicaciones , Encefalitis Viral/diagnóstico por imagen , Femenino , Humanos , Lactante , Recién Nacido , Espectroscopía de Resonancia Magnética , Masculino , Convulsiones/etiología , Virus/clasificación
20.
Comp Immunol Microbiol Infect Dis ; 73: 101495, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32889429

RESUMEN

Rabies and herpetic encephalitis are the main viral infections in bovines with neurological symptoms. Bovine rabies has a high prevalence in Central and South America, while bovine encephalitis associated with herpesvirus is especially important in South America. Viral isolation is the classical way to confirm herpesvirus infection, but molecular evidence of the presence of the virus in affected animals is gaining importance in the diagnosis of the disease in the laboratory. This study investigated the presence of herpesvirus type 1 and 5 (BoHV-1 and BoHV-5) in 182 encephalon of rabies-suspected cattle in Rio Grande do Sul state (RS), Brazil using multiplex real-time polymerase chain reaction (mRT-PCR). The rabies virus was investigated by direct fluorescent antibody assay and intracerebral suckling mouse inoculation. The genomes of BoHV-1 and BoHV-5 were detected in 17% of samples. BoHV-5 and BoHV-1 were detected in 100% and 19% of BoHV positive samples, respectively, indicating the circulation of the pathogens in cattle herds in RS. The high Ct values and the absence of isolation suggest viral latency. Coinfection of herpesvirus and the rabies virus was detected in 28% of samples, although no significant association between pathogens was observed. Rabies was detected in 57.7% of suspected samples, confirming the importance of the disease in the state. Concerning the method by which samples were conserved, no significant difference was observed between the number of positive results in frozen and refrigerated samples.


Asunto(s)
Enfermedades de los Bovinos/virología , Infecciones por Herpesviridae/veterinaria , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 5/genética , Rabia/veterinaria , Animales , Encéfalo/virología , Brasil/epidemiología , Bovinos , Enfermedades de los Bovinos/epidemiología , Criopreservación/veterinaria , ADN Viral/aislamiento & purificación , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Ratones , Reacción en Cadena de la Polimerasa Multiplex/veterinaria , Rabia/epidemiología , Refrigeración/veterinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA