Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.856
Filtrar
1.
Vet Res ; 55(1): 71, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822398

RESUMEN

In the wake of the COVID-19 pandemic caused by SARS-CoV-2, questions emerged about the potential effects of Bacillus Calmette-Guérin (BCG) vaccine on the immune response to SARS-CoV-2 infection, including the neurodegenerative diseases it may contribute to. To explore this, an experimental study was carried out in BCG-stimulated and non-stimulated k18-hACE2 mice challenged with SARS-CoV-2. Viral loads in tissues determined by RT-qPCR, histopathology in brain and lungs, immunohistochemical study in brain (IHC) as well as mortality rates, clinical signs and plasma inflammatory and coagulation biomarkers were assessed. Our results showed BCG-SARS-CoV-2 challenged mice presented higher viral loads in the brain and an increased frequency of neuroinvasion, with the greatest differences observed between groups at 3-4 days post-infection (dpi). Histopathological examination showed a higher severity of brain lesions in BCG-SARS-CoV-2 challenged mice, mainly consisting of neuroinflammation, increased glial cell population and neuronal degeneration, from 5 dpi onwards. This group also presented higher interstitial pneumonia and vascular thrombosis in lungs (3-4 dpi), BCG-SARS-CoV-2 mice showed higher values for TNF-α and D-dimer values, while iNOS values were higher in SARS-CoV-2 mice at 3-4 dpi. Results presented in this study indicate that BCG stimulation could have intensified the inflammatory and neurodegenerative lesions promoting virus neuroinvasion and dissemination in this experimental model. Although k18-hACE2 mice show higher hACE2 expression and neurodissemination, this study suggests that, although the benefits of BCG on enhancing heterologous protection against pathogens and tumour cells have been broadly demonstrated, potential adverse outcomes due to the non-specific effects of BCG should be considered.


Asunto(s)
Vacuna BCG , Encéfalo , COVID-19 , SARS-CoV-2 , Animales , Ratones , Vacuna BCG/administración & dosificación , COVID-19/inmunología , COVID-19/virología , SARS-CoV-2/fisiología , Encéfalo/patología , Encéfalo/virología , Carga Viral , Pulmón/patología , Pulmón/virología , Pulmón/inmunología , Enzima Convertidora de Angiotensina 2/metabolismo , Ratones Transgénicos , Femenino
2.
Arch Virol ; 169(6): 133, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829449

RESUMEN

Akabane virus (AKAV), Aino virus, Peaton virus, Sathuperi virus, and Shamonda virus are arthropod-borne viruses belonging to the order Elliovirales, family Peribunyaviridae, genus Orthobunyavirus. These viruses cause or may cause congenital malformations in ruminants, including hydranencephaly, poliomyelitis, and arthrogryposis, although their pathogenicity may vary among field cases. AKAV may cause relatively severe congenital lesions such as hydranencephaly in calves. Furthermore, strains of AKAV genogroups I and II exhibit different disease courses. Genogroup I strains predominantly cause postnatal viral encephalomyelitis, while genogroup II strains are primarily detected in cases of congenital malformation. However, the biological properties of AKAV and other orthobunyaviruses are insufficiently investigated in hosts in the field and in vitro. Here, we used an immortalized bovine brain cell line (FBBC-1) to investigate viral replication efficiency, cytopathogenicity, and host innate immune responses. AKAV genogroup II and Shamonda virus replicated to higher titers in FBBC-1 cells compared with the other viruses, and only AKAV caused cytopathic effects. These results may be associated with the severe congenital lesions in the brain caused by AKAV genogroup II. AKAV genogroup II strains replicated to higher titers in FBBC-1 cells than AKAV genogroup I strains, suggesting that genogroup II strains replicated more efficiently in fetal brain cells, accounting for the detection of the latter strains mainly in fetal infection cases. Therefore, FBBC-1 cells may serve as a valuable tool for investigating the virulence and tropism of the orthobunyaviruses for bovine neonatal brain tissues in vitro.


Asunto(s)
Encéfalo , Infecciones por Bunyaviridae , Orthobunyavirus , Replicación Viral , Animales , Bovinos , Orthobunyavirus/patogenicidad , Orthobunyavirus/genética , Orthobunyavirus/fisiología , Orthobunyavirus/clasificación , Encéfalo/virología , Encéfalo/patología , Línea Celular , Infecciones por Bunyaviridae/virología , Infecciones por Bunyaviridae/veterinaria , Infecciones por Bunyaviridae/patología , Enfermedades de los Bovinos/virología , Feto/virología , Efecto Citopatogénico Viral , Inmunidad Innata
3.
Front Immunol ; 15: 1374301, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38835765

RESUMEN

Background: Human immunodeficiency virus (HIV) affects nearly 40 million people globally, with roughly 80% of all people living with HIV receiving antiretroviral therapy. Antiretroviral treatment suppresses viral load in peripheral tissues but does not effectively penetrate the blood-brain barrier. Thus, viral reservoirs persist in the central nervous system and continue to produce low levels of inflammatory factors and early viral proteins, including the transactivator of transcription (Tat). HIV Tat is known to contribute to chronic neuroinflammation and synaptodendritic damage, which is associated with the development of cognitive, motor, and/or mood problems, collectively known as HIV-associated neurocognitive disorders (HAND). Cannabinoid anti-inflammatory effects are well documented, but therapeutic utility of cannabis remains limited due to its psychotropic effects, including alterations within brain regions encoding reward processing and motivation, such as the nucleus accumbens. Alternatively, inhibiting monoacylglycerol lipase (MAGL) has demonstrated therapeutic potential through interactions with the endocannabinoid system. Methods: The present study utilized a reward-related operant behavioral task to quantify motivated behavior in female Tat transgenic mice treated with vehicle or MAGL inhibitor MJN110 (1 mg/kg). Brain tissue was collected to assess dendritic injury and neuroinflammatory profiles, including dendritic microtubule-associated protein (MAP2ab) intensity, microglia density, microglia morphology, astrocyte density, astrocytic interleukin-1ß (IL-1ß) colocalization, and various lipid mediators. Results: No significant behavioral differences were observed; however, MJN110 protected against Tat-induced dendritic injury by significantly upregulating MAP2ab intensity in the nucleus accumbens and in the infralimbic cortex of Tat(+) mice. No or only minor effects were noted for Iba-1+ microglia density and/or microglia morphology. Further, Tat increased GFAP+ astrocyte density in the infralimbic cortex and GFAP+ astrocytic IL-1ß colocalization in the nucleus accumbens, with MJN110 significantly reducing these measures in Tat(+) subjects. Lastly, selected HETE-related inflammatory lipid mediators in the striatum were downregulated by chronic MJN110 treatment. Conclusions: These findings demonstrate anti-inflammatory and neuroprotective properties of MJN110 without cannabimimetic behavioral effects and suggest a promising alternative to cannabis for managing neuroinflammation.


Asunto(s)
VIH-1 , Ratones Transgénicos , Monoacilglicerol Lipasas , Enfermedades Neuroinflamatorias , Animales , Ratones , VIH-1/fisiología , Enfermedades Neuroinflamatorias/tratamiento farmacológico , Enfermedades Neuroinflamatorias/etiología , Femenino , Monoacilglicerol Lipasas/antagonistas & inhibidores , Monoacilglicerol Lipasas/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/metabolismo , Infecciones por VIH/tratamiento farmacológico , Humanos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/virología , Encéfalo/patología , Modelos Animales de Enfermedad , Microglía/efectos de los fármacos , Microglía/metabolismo , Complejo SIDA Demencia/tratamiento farmacológico
4.
Sci Rep ; 14(1): 10709, 2024 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-38729980

RESUMEN

Three years after SARS-CoV-2 emerged as a global infectious threat, the virus has become endemic. The neurological complications such as depression, anxiety, and other CNS complications after COVID-19 disease are increasing. The brain, and CSF have been shown as viral reservoirs for SARS-CoV-2, yielding a potential hypothesis for CNS effects. Thus, we investigated the CNS pharmacology of orally dosed nirmatrelvir/ritonavir (NMR/RTV). Using both an in vitro and an in vivo rodent model, we investigated CNS penetration and potential pharmacodynamic activity of NMR. Through pharmacokinetic modeling, we estimated the median CSF penetration of NMR to be low at 18.11% of plasma with very low accumulation in rodent brain tissue. Based on the multiples of the 90% maximal effective concentration (EC90) for SARS-CoV-2, NMR concentrations in the CSF and brain do not achieve an exposure level similar to that of plasma. A median of only 16% of all the predicted CSF concentrations in rats were > 3xEC90 (unadjusted for protein binding). This may have implications for viral persistence and neurologic post-acute sequelae of COVID-19 if increased NMR penetration in the CNS leads to decreased CNS viral loads and decreased CNS inflammation.


Asunto(s)
Leucocitos Mononucleares , Ritonavir , SARS-CoV-2 , Animales , Ratas , Ritonavir/farmacocinética , SARS-CoV-2/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/virología , Humanos , Masculino , Encéfalo/metabolismo , Encéfalo/virología , Tratamiento Farmacológico de COVID-19 , COVID-19/virología , COVID-19/líquido cefalorraquídeo , Antivirales/farmacocinética , Antivirales/farmacología , Ratas Sprague-Dawley , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/virología
5.
Acta Neuropathol ; 147(1): 92, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38801558

RESUMEN

The SARS-CoV-2 virus that led to COVID-19 is associated with significant and long-lasting neurologic symptoms in many patients, with an increased mortality risk for people with Alzheimer's disease (AD) and/or Down syndrome (DS). However, few studies have evaluated the neuropathological and inflammatory sequelae in postmortem brain tissue obtained from AD and people with DS with severe SARS-CoV-2 infections. We examined tau, beta-amyloid (Aß), inflammatory markers and SARS-CoV-2 nucleoprotein in DS, AD, and healthy non-demented controls with COVID-19 and compared with non-infected brain tissue from each disease group (total n = 24). A nested ANOVA was used to determine regional effects of the COVID-19 infection on arborization of astrocytes (Sholl analysis) and percent-stained area of Iba-1 and TMEM 119. SARS-CoV-2 antibodies labeled neurons and glial cells in the frontal cortex of all subjects with COVID-19, and in the hippocampus of two of the three DS COVID-19 cases. SARS-CoV-2-related alterations were observed in peri-vascular astrocytes and microglial cells in the gray matter of the frontal cortex, hippocampus, and para-hippocampal gyrus. Bright field microscopy revealed scattered intracellular and diffuse extracellular Aß deposits in the hippocampus of controls with confirmed SARS-CoV-2 infections. Overall, the present preliminary findings suggest that SARS-CoV-2 infections induce abnormal inflammatory responses in Down syndrome.


Asunto(s)
Enfermedad de Alzheimer , Encéfalo , COVID-19 , Síndrome de Down , Humanos , Síndrome de Down/patología , Síndrome de Down/metabolismo , Síndrome de Down/complicaciones , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/virología , Enfermedad de Alzheimer/metabolismo , COVID-19/patología , COVID-19/complicaciones , Masculino , Femenino , Anciano , Persona de Mediana Edad , Encéfalo/patología , Encéfalo/virología , Anciano de 80 o más Años , Astrocitos/patología , Astrocitos/virología , Astrocitos/metabolismo , Péptidos beta-Amiloides/metabolismo , SARS-CoV-2/patogenicidad , Microglía/patología , Microglía/metabolismo , Adulto , Proteínas tau/metabolismo
6.
Viruses ; 16(5)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38793666

RESUMEN

SARS-CoV-2 primarily infects the lungs via the ACE2 receptor but also other organs including the kidneys, the gastrointestinal tract, the heart, and the skin. SARS-CoV-2 also infects the brain, but the hematogenous route of viral entry to the brain is still not fully characterized. Understanding how SARS-CoV-2 traverses the blood-brain barrier (BBB) as well as how it affects the molecular functions of the BBB are unclear. In this study, we investigated the roles of the receptors ACE2 and DPP4 in the SARS-CoV-2 infection of the discrete cellular components of a transwell BBB model comprising HUVECs, astrocytes, and pericytes. Our results demonstrate that direct infection on the BBB model does not modulate paracellular permeability. Also, our results show that SARS-CoV-2 utilizes clathrin and caveolin-mediated endocytosis to traverse the BBB, resulting in the direct infection of the brain side of the BBB model with a minimal endothelial infection. In conclusion, the BBB is susceptible to SARS-CoV-2 infection in multiple ways, including the direct infection of endothelium, astrocytes, and pericytes involving ACE2 and/or DPP4 and the blood-to-brain transcytosis, which is an event that does not require the presence of host receptors.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Astrocitos , Barrera Hematoencefálica , COVID-19 , Dipeptidil Peptidasa 4 , Pericitos , SARS-CoV-2 , Transcitosis , Internalización del Virus , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Humanos , SARS-CoV-2/fisiología , Enzima Convertidora de Angiotensina 2/metabolismo , Pericitos/virología , Pericitos/metabolismo , COVID-19/virología , COVID-19/metabolismo , Astrocitos/virología , Astrocitos/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Encéfalo/virología , Encéfalo/metabolismo , Endocitosis , Células Endoteliales de la Vena Umbilical Humana/virología , Permeabilidad
7.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(4): 303-310, 2024 Apr.
Artículo en Chino | MEDLINE | ID: mdl-38710514

RESUMEN

Objective To clarify the relationship between astrocyte activation patterns and disease progression in epidemic encephalitis B (Japanese encephalitis). Methods First, a mouse model of epidemic encephalitis B was constructed by foot-pad injection of Japanese encephalitis virus (JEV), and the expression of viral protein NS3 in different brain regions was detected by immunofluorescence assay (IFA). Next, IFA, RNA sequencing (RNA-seq) and real-time quantitative PCR (qRT-PCR) were used to clarify the changes in the astrocyte activation patterns at different stages of epidemic encephalitis B. Finally, intracerebroventricular administration of irisin was conducted to regulate the proportion of activation in complement C3-positive A1 astrocytes and S100A10-positive A2 astrocytes, investigating whether it could improve the body mass, behavioral scores, and brain tissue damage in a mouse model. Results NS3 protein was detected by IFA predominantly in the M1/M2 region of the motor cortex and the hippocampus. The number and volume of GFAP-positive astrocytes significantly increased in JEV-infected brain regions, in which the expression of multiple genes associated with A1/A2 astrocyte activation was significantly enhanced. Although intracerebroventricular or intraperitoneal injection of irisin did not improve the prognosis of epidemic encephalitis B, it inhibited the activation of A1 astrocytes and ameliorate neuroinflammation. Conclusion Neurons in the M1/M2 motor cortex and hippocampus are susceptible to JEV infection, in which the abnormal astrocyte activation contributes to the neuroinflammatory injury. Irisin administration may restrain A1 astrocyte activation and alleviate neuroinflammation following JEV infection.


Asunto(s)
Astrocitos , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , Animales , Astrocitos/metabolismo , Astrocitos/virología , Ratones , Encefalitis Japonesa/inmunología , Virus de la Encefalitis Japonesa (Especie)/fisiología , Encéfalo/metabolismo , Encéfalo/virología , Encéfalo/patología , Masculino , Fibronectinas/metabolismo , Fibronectinas/genética
8.
Methods Mol Biol ; 2807: 261-270, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743234

RESUMEN

The development of 3D-organoid models has revolutionized the way diseases are studied. Recently, our brain organoid model has been shown to recapitulate in in vitro the human brain cytoarchitecture originally encountered in HIV-1 neuropathogenesis, allowing downstream applications. Infected monocytes, macrophages, and microglia are critically important immune cells for infection and dissemination of HIV-1 throughout brain during acute and chronic phase of the disease. Once in the brain parenchyma, long-lived infected monocytes/macrophages along with resident microglia contribute to the establishment of CNS latency in people with HIV (PWH). Hence, it is important to better understand how HIV-1 enters and establishes infection and latency in CNS to further develop cure strategies. Here we detailed an accessible protocol to incorporate monocytes (infected and/or labeled) as a model of transmigration of peripheral monocytes into brain organoids that can be applied to characterize HIV-1 neuroinvasion and virus dissemination.


Asunto(s)
Encéfalo , Infecciones por VIH , VIH-1 , Monocitos , Organoides , Organoides/virología , Organoides/patología , Humanos , VIH-1/fisiología , VIH-1/patogenicidad , Monocitos/virología , Monocitos/inmunología , Infecciones por VIH/virología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Encéfalo/virología , Encéfalo/patología , Encéfalo/inmunología , Microglía/virología , Microglía/inmunología , Microglía/patología , Macrófagos/virología , Macrófagos/inmunología , Latencia del Virus
9.
Methods Mol Biol ; 2807: 271-283, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743235

RESUMEN

The blood-brain barrier (BBB) is one of several barriers between the brain and the peripheral blood system to maintain homeostasis. Understanding the interactions between infectious agents such as human immunodeficiency virus type 1 (HIV-1), which are capable of traversing the BBB and causing neuroinflammation requires modeling an authentic BBB in vitro. Such an in vitro BBB model also helps develop means of targeting viruses that reside in the brain via natural immune effectors such as antibodies. The BBB consists of human brain microvascular endothelial cells (HBMECs), astrocytes, and pericytes. Here we report in vitro methods to establish a dual-cell BBB model consisting of primary HBMECs and primary astrocytes to measure the integrity of the BBB and antibody penetration of the BBB, as well as a method to establish a single cell BBB model to study the impact of HIV-1 infected medium on the integrity of such a BBB.


Asunto(s)
Astrocitos , Barrera Hematoencefálica , Células Endoteliales , Infecciones por VIH , VIH-1 , Barrera Hematoencefálica/virología , Barrera Hematoencefálica/metabolismo , Humanos , Astrocitos/virología , Astrocitos/metabolismo , Astrocitos/inmunología , Células Endoteliales/virología , Células Endoteliales/metabolismo , Células Endoteliales/inmunología , VIH-1/inmunología , VIH-1/fisiología , Infecciones por VIH/virología , Infecciones por VIH/inmunología , Pericitos/virología , Pericitos/metabolismo , Pericitos/inmunología , Enfermedades Neuroinflamatorias/virología , Enfermedades Neuroinflamatorias/inmunología , Técnicas de Cocultivo/métodos , Células Cultivadas , Encéfalo/virología , Encéfalo/inmunología , Encéfalo/metabolismo
10.
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
11.
PLoS One ; 19(5): e0303059, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743751

RESUMEN

Non-human primate (NHP)-based model systems are highly relevant for biomedical research. However, only few NHP cell lines are available and the generation of additional cell lines is an urgent need to help in the refinement and replacement of these models. Using lentiviral transduction of c-Fos, we established cell lines from the brain of rhesus macaques (Macaca mulatta). Transcriptome analysis revealed that these cell lines are closely related to astrocytes, which was confirmed by immunoblot and immunofluorescence microscopy detecting expression of the astrocyte marker glial fibrillary acidic protein (GFAP). Quantitative real-time PCR (qRT-PCR) demonstrated that major pathways of the interferon (IFN) system are intact. Using retroviral pseudotypes we found that the cell lines are susceptible to entry driven by the glycoproteins of vesicular stomatitis virus (VSV), lymphocytic choriomeningitis virus (LCMV) and to a lesser extent influenza A virus (IAV). Finally, these cells supported growth of Zika virus (ZIKV) and Papiine alphaherpesvirus 2 (PaHV2). In summary, we developed IFN-responsive cell lines from the rhesus macaque brain that allowed entry driven by several viral glycoproteins and were permissive to infection with ZIKV and a primate simplexvirus. These cell lines will be useful for efforts to analyze neurotropic viral infections in rhesus macaque models.


Asunto(s)
Astrocitos , Macaca mulatta , Animales , Astrocitos/virología , Astrocitos/metabolismo , Línea Celular , Encéfalo/virología , Encéfalo/metabolismo , Humanos
12.
BMC Med Imaging ; 24(1): 119, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38783187

RESUMEN

BACKGROUND: Magnetic Resonance Imaging (MRI)-based imaging techniques are useful for assessing white matter (WM) structural and microstructural integrity in the context of infection and inflammation. The purpose of this scoping review was to assess the range of work on the use of WM neuroimaging approaches to understand the impact of congenital and perinatal viral infections or exposures on the developing brain. METHODS: This scoping review was conducted according to the Arksey and O' Malley framework. A literature search was performed in Web of Science, Scopus and PubMed for primary research articles published from database conception up to January 2022. Studies evaluating the use of MRI-based WM imaging techniques in congenital and perinatal viral infections or exposures were included. Results were grouped by age and infection. RESULTS: A total of 826 articles were identified for screening and 28 final articles were included. Congenital and perinatal infections represented in the included studies were cytomegalovirus (CMV) infection (n = 12), human immunodeficiency virus (HIV) infection (n = 11) or exposure (n = 2) or combined (n = 2), and herpes simplex virus (HSV) infection (n = 1). The represented MRI-based WM imaging methods included structural MRI and diffusion-weighted and diffusion tensor MRI (DWI/ DTI). Regions with the most frequently reported diffusion metric group differences included the cerebellar region, corticospinal tract and association fibre WM tracts in both children with HIV infection and children who are HIV-exposed uninfected. In qualitative imaging studies, WM hyperintensities were the most frequently reported brain abnormality in children with CMV infection and children with HSV infection. CONCLUSION: There was evidence that WM imaging techniques can play a role as diagnostic and evaluation tools assessing the impact of congenital infections and perinatal viral exposures on the developing brain. The high sensitivity for identifying WM hyperintensities suggests structural brain MRI is a useful neurodiagnostic modality in assessing children with congenital CMV infection, while the DTI changes associated with HIV suggest metrics such as fractional anisotropy have the potential to be specific markers of subtle impairment or WM damage in neuroHIV.


Asunto(s)
Imagen por Resonancia Magnética , Sustancia Blanca , Humanos , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/virología , Imagen por Resonancia Magnética/métodos , Femenino , Embarazo , Recién Nacido , Encéfalo/diagnóstico por imagen , Encéfalo/virología , Encéfalo/patología , Infecciones por Citomegalovirus/diagnóstico por imagen , Infecciones por Citomegalovirus/congénito , Infecciones por VIH/diagnóstico por imagen , Neuroimagen/métodos , Imagen de Difusión Tensora/métodos , Complicaciones Infecciosas del Embarazo/diagnóstico por imagen , Complicaciones Infecciosas del Embarazo/virología , Lactante , Virosis/diagnóstico por imagen
13.
Front Immunol ; 15: 1384516, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38765009

RESUMEN

Viral variant is one known risk factor associated with post-acute sequelae of COVID-19 (PASC), yet the pathogenesis is largely unknown. Here, we studied SARS-CoV-2 Delta variant-induced PASC in K18-hACE2 mice. The virus replicated productively, induced robust inflammatory responses in lung and brain tissues, and caused weight loss and mortality during the acute infection. Longitudinal behavior studies in surviving mice up to 4 months post-acute infection revealed persistent abnormalities in neuropsychiatric state and motor behaviors, while reflex and sensory functions recovered over time. In the brain, no detectable viral RNA and minimal residential immune cell activation was observed in the surviving mice post-acute infection. Transcriptome analysis revealed persistent activation of immune pathways, including humoral responses, complement, and phagocytosis, and gene expression levels associated with ataxia telangiectasia, impaired cognitive function and memory recall, and neuronal dysfunction and degeneration. Furthermore, surviving mice maintained potent systemic T helper 1 prone cellular immune responses and strong sera neutralizing antibodies against Delta and Omicron variants months post-acute infection. Overall, our findings suggest that infection in K18-hACE2 mice recapitulates the persistent clinical symptoms reported in long-COVID patients and provides new insights into the role of systemic and brain residential immune factors in PASC pathogenesis.


Asunto(s)
COVID-19 , Modelos Animales de Enfermedad , Síndrome Post Agudo de COVID-19 , SARS-CoV-2 , Animales , COVID-19/inmunología , SARS-CoV-2/inmunología , Ratones , Humanos , Encéfalo/virología , Encéfalo/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Femenino
14.
Rev Med Virol ; 34(3): e2539, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38719789

RESUMEN

The viral infection of the central nervous system is a significant public health concern. So far, most clinical cases of viral neuroinvasion are dealt with supportive and/or symptomatic treatments due to the unavailability of specific treatments. Thus, developing specific therapies is required to alleviate neurological symptoms and disorders. In this review, we shed light on molecular aspects of viruses' entry into the brain which upon targeting with specific drugs have shown promising efficacy in vitro and in preclinical in vivo model systems. Further assessing the therapeutic potential of these drugs in clinical trials may offer opportunities to halt viral neuroinvasion in humans.


Asunto(s)
Antivirales , Humanos , Animales , Antivirales/uso terapéutico , Antivirales/farmacología , Internalización del Virus/efectos de los fármacos , Encéfalo/virología , Encéfalo/patología , Encéfalo/efectos de los fármacos , Enfermedades Virales del Sistema Nervioso Central/tratamiento farmacológico , Enfermedades Virales del Sistema Nervioso Central/virología
15.
Cell Rep Med ; 5(5): 101570, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38749422

RESUMEN

While an association between Parkinson's disease (PD) and viral infections has been recognized, the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) on PD progression remains unclear. Here, we demonstrate that SARS-CoV-2 infection heightens the risk of PD using human embryonic stem cell (hESC)-derived dopaminergic (DA) neurons and a human angiotensin-converting enzyme 2 (hACE2) transgenic (Tg) mouse model. Our findings reveal that SARS-CoV-2 infection exacerbates PD susceptibility and cellular toxicity in DA neurons pre-treated with human preformed fibrils (hPFFs). Additionally, nasally delivered SARS-CoV-2 infects DA neurons in hACE2 Tg mice, aggravating the damage initiated by hPFFs. Mice infected with SARS-CoV-2 display persisting neuroinflammation even after the virus is no longer detectable in the brain. A comprehensive analysis suggests that the inflammatory response mediated by astrocytes and microglia could contribute to increased PD susceptibility associated with SARS-CoV-2. These findings advance our understanding of the potential long-term effects of SARS-CoV-2 infection on the progression of PD.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Modelos Animales de Enfermedad , Neuronas Dopaminérgicas , Ratones Transgénicos , Enfermedad de Parkinson , SARS-CoV-2 , Animales , Neuronas Dopaminérgicas/patología , Neuronas Dopaminérgicas/metabolismo , Neuronas Dopaminérgicas/virología , Humanos , COVID-19/patología , COVID-19/virología , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/virología , Ratones , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , Microglía/patología , Microglía/metabolismo , Microglía/virología , Células Madre Embrionarias Humanas/metabolismo , Astrocitos/patología , Astrocitos/virología , Astrocitos/metabolismo , Encéfalo/patología , Encéfalo/virología
16.
Nat Commun ; 15(1): 3803, 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38778015

RESUMEN

Human endogenous retroviruses (HERVs) are repetitive elements previously implicated in major psychiatric conditions, but their role in aetiology remains unclear. Here, we perform specialised transcriptome-wide association studies that consider HERV expression quantified to precise genomic locations, using RNA sequencing and genetic data from 792 post-mortem brain samples. In Europeans, we identify 1238 HERVs with expression regulated in cis, of which 26 represent expression signals associated with psychiatric disorders, with ten being conditionally independent from neighbouring expression signals. Of these, five are additionally significant in fine-mapping analyses and thus are considered high confidence risk HERVs. These include two HERV expression signatures specific to schizophrenia risk, one shared between schizophrenia and bipolar disorder, and one specific to major depressive disorder. No robust signatures are identified for autism spectrum conditions or attention deficit hyperactivity disorder in Europeans, or for any psychiatric trait in other ancestries, although this is likely a result of relatively limited statistical power. Ultimately, our study highlights extensive HERV expression and regulation in the adult cortex, including in association with psychiatric disorder risk, therefore providing a rationale for exploring neurological HERV expression in complex neuropsychiatric traits.


Asunto(s)
Trastorno Bipolar , Trastorno Depresivo Mayor , Retrovirus Endógenos , Estudio de Asociación del Genoma Completo , Esquizofrenia , Transcriptoma , Humanos , Retrovirus Endógenos/genética , Esquizofrenia/genética , Esquizofrenia/virología , Trastorno Bipolar/genética , Factores de Riesgo , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/virología , Trastornos Mentales/genética , Encéfalo/metabolismo , Encéfalo/virología , Femenino , Masculino , Predisposición Genética a la Enfermedad , Trastorno por Déficit de Atención con Hiperactividad/genética , Adulto
17.
Virulence ; 15(1): 2355971, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38745468

RESUMEN

The vertebrate central nervous system (CNS) is the most complex system of the body. The CNS, especially the brain, is generally regarded as immune-privileged. However, the specialized immune strategies in the brain and how immune cells, specifically macrophages in the brain, respond to virus invasion remain poorly understood. Therefore, this study aimed to examine the potential immune response of macrophages in the brain of orange-spotted groupers (Epinephelus coioides) following red-spotted grouper nervous necrosis virus (RGNNV) infection. We observed that RGNNV induced macrophages to produce an inflammatory response in the brain of orange-spotted grouper, and the macrophages exhibited M1-type polarization after RGNNV infection. In addition, we found RGNNV-induced macrophage M1 polarization via the CXCR3.2- CXCL11 pathway. Furthermore, we observed that RGNNV triggered M1 polarization in macrophages, resulting in substantial proinflammatory cytokine production and subsequent damage to brain tissue. These findings reveal a unique mechanism for brain macrophage polarization, emphasizing their role in contributing to nervous tissue damage following viral infection in the CNS.


Asunto(s)
Encéfalo , Enfermedades de los Peces , Macrófagos , Nodaviridae , Infecciones por Virus ARN , Animales , Macrófagos/inmunología , Macrófagos/virología , Enfermedades de los Peces/virología , Enfermedades de los Peces/inmunología , Encéfalo/virología , Encéfalo/inmunología , Encéfalo/patología , Nodaviridae/fisiología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Quimiocina CXCL11 , Receptores CXCR3/metabolismo , Lubina/inmunología , Lubina/virología , Transducción de Señal , Citocinas/metabolismo , Citocinas/inmunología , Proteínas de Peces/inmunología , Proteínas de Peces/genética
18.
Viruses ; 16(5)2024 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-38793575

RESUMEN

BACKGROUND: EcoHIV is a chimeric HIV that replicates in mice in CD4+ T cells, macrophages, and microglia (but not in neurons), causing lasting neurocognitive impairment resembling neurocognitive disease in people living with HIV. The present study was designed to develop EcoHIV-susceptible primary mouse brain cultures to investigate the indirect effects of HIV infection on neuronal integrity. RESULTS: We used two EcoHIV clones encoding EGFP and mouse bone marrow-derived macrophages (BMM), mixed mouse brain cells, or enriched mouse glial cells from two wild-type mouse strains to test EcoHIV replication efficiency, the identity of productively infected cells, and neuronal apoptosis and integrity. EcoHIV replicated efficiently in BMM. In mixed brain cell cultures, EcoHIV targeted microglia but did not cause neuronal apoptosis. Instead, the productive infection of the microglia activated them and impaired synaptophysin expression, dendritic density, and axonal structure in the neurons. EcoHIV replication in the microglia and neuronal structural changes during infection were prevented by culture with an antiretroviral. CONCLUSIONS: In murine brain cell cultures, EcoHIV replication in the microglia is largely responsible for the aspects of neuronal dysfunction relevant to cognitive disease in infected mice and people living with HIV. These cultures provide a tool for further study of HIV neuropathogenesis and its control.


Asunto(s)
Encéfalo , Microglía , Neuronas , Replicación Viral , Animales , Ratones , Encéfalo/virología , Encéfalo/patología , Neuronas/virología , Neuronas/patología , Microglía/virología , Células Cultivadas , Infecciones por VIH/virología , Macrófagos/virología , Modelos Animales de Enfermedad , Apoptosis , Humanos , VIH-1/fisiología , Cultivo Primario de Células , Ratones Endogámicos C57BL
19.
Viruses ; 16(5)2024 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-38793581

RESUMEN

Rabies is a fatal encephalitic infectious disease caused by the rabies virus (RABV). RABV is highly neurotropic and replicates in neuronal cell lines in vitro. The RABV fixed strain, HEP-Flury, was produced via passaging in primary chicken embryonic fibroblast cells. HEP-Flury showed rapid adaptation when propagated in mouse neuroblastoma (MNA) cells. In this study, we compared the growth of our previously constructed recombinant HEP (rHEP) strain-based on the sequence of the HEP (HEP-Flury) strain-with that of the original HEP strain. The original HEP strain exhibited higher titer than rHEP and a single substitution at position 80 in the matrix (M) protein M(D80N) after incubation in MNA cells, which was absent in rHEP. In vivo, intracerebral inoculation of the rHEP-M(D80N) strain with this substitution resulted in enhanced viral growth in the mouse brain and a significant loss of body weight in the adult mice. The number of viral antigen-positive cells in the brains of adult mice inoculated with the rHEP-M(D80N) strain was significantly higher than that with the rHEP strain at 5 days post-inoculation. Our findings demonstrate that a single amino acid substitution in the M protein M(D80N) is associated with neurovirulence in mice owing to adaptation to mouse neuronal cells.


Asunto(s)
Sustitución de Aminoácidos , Encéfalo , Virus de la Rabia , Rabia , Proteínas de la Matriz Viral , Animales , Virus de la Rabia/genética , Virus de la Rabia/patogenicidad , Ratones , Virulencia , Encéfalo/virología , Encéfalo/patología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/metabolismo , Rabia/virología , Neuronas/virología , Neuronas/patología , Replicación Viral , Línea Celular
20.
Microbiol Spectr ; 12(6): e0069024, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38752731

RESUMEN

Enterovirus A71 (EV-A71) is associated with neurological conditions such as acute meningitis and encephalitis. The virus is detected in the bloodstream, and high blood viral loads are associated with central nervous system (CNS) manifestations. We used an in vitro blood-brain barrier (BBB) model made up of human brain-like endothelial cells (hBLECs) and brain pericytes grown in transwell systems to investigate whether three genetically distinct EV-A71 strains (subgenogroups C1, C1-like, and C4) can cross the human BBB. EV-A71 poorly replicated in hBLECs, which released moderate amounts of infectious viruses from their luminal side and trace amounts of infectious viruses from their basolateral side. The barrier properties of hBLECs were not impaired by EV-A71 infection. We investigated the passage through hBLECs of EV-A71-infected white blood cells. EV-A71 strains efficiently replicated in immune cells, including monocytes, neutrophils, and NK/T cells. Attachment to hBLECs of immune cells infected with the C1-like virus was higher than attachment of cells infected with C1-06. EV-A71 infection did not impair the transmigration of immune cells through hBLECs. Overall, EV-A71 targets different white blood cell populations that have the potential to be used as a Trojan horse to cross hBLECs more efficiently than cell-free EV-A71 particles.IMPORTANCEEnterovirus A71 (EV-A71) was first reported in the USA, and numerous outbreaks have since occurred in Asia and Europe. EV-A71 re-emerged as a new multirecombinant strain in 2015 in Europe and is now widespread. The virus causes hand-foot-and-mouth disease in young children and is involved in nervous system infections. How the virus spreads to the nervous system is unclear. We investigated whether white blood cells could be infected by EV-A71 and transmit it across human endothelial cells mimicking the blood-brain barrier protecting the brain from adverse effects. We found that endothelial cells provide a strong roadblock to prevent the passage of free virus particles but allow the migration of infected immune cells, including monocytes, neutrophils, and NK/T cells. Our data are consistent with the potential role of immune cells in the pathogenesis of EV-A71 infections by spreading the virus in the blood and across the human blood-brain barrier.


Asunto(s)
Barrera Hematoencefálica , Células Endoteliales , Enterovirus Humano A , Infecciones por Enterovirus , Barrera Hematoencefálica/virología , Humanos , Enterovirus Humano A/genética , Enterovirus Humano A/fisiología , Infecciones por Enterovirus/virología , Infecciones por Enterovirus/inmunología , Células Endoteliales/virología , Replicación Viral , Monocitos/virología , Monocitos/inmunología , Pericitos/virología , Leucocitos/virología , Leucocitos/inmunología , Encéfalo/virología , Células Asesinas Naturales/inmunología , Neutrófilos/inmunología , Neutrófilos/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA