Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.733
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 8822, 2024 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-39394194

RESUMO

Zika virus (ZIKV), an emerging mosquito-borne flavivirus, is associated with congenital neurological complications. Here, we investigate potential pathological correlates of virus gene expression in representative ZIKV strains through RNA sequencing and ribosome profiling. In addition to the single long polyprotein found in all flaviviruses, we identify the translation of unrecognised upstream open reading frames (uORFs) in the genomic 5' region. In Asian/American strains, ribosomes translate uORF1 and uORF2, whereas in African strains, the two uORFs are fused into one (African uORF). We use reverse genetics to examine the impact on ZIKV fitness of different uORFs mutant viruses. We find that expression of the African uORF and the Asian/American uORF1 modulates virus growth and tropism in human cortical neurons and cerebral organoids, suggesting a potential role in neurotropism. Although the uORFs are expressed in mosquito cells, we do not see a measurable effect on transmission by the mosquito vector in vivo. The discovery of ZIKV uORFs sheds new light on the infection of the human brain cells by this virus and raises the question of their existence in other neurotropic flaviviruses.


Assuntos
Encéfalo , Neurônios , Fases de Leitura Aberta , Infecção por Zika virus , Zika virus , Zika virus/genética , Zika virus/fisiologia , Humanos , Fases de Leitura Aberta/genética , Infecção por Zika virus/virologia , Animais , Encéfalo/virologia , Neurônios/virologia , Neurônios/metabolismo , Replicação Viral , Organoides/virologia , Chlorocebus aethiops , Tropismo Viral , Células Vero , Mosquitos Vetores/virologia , Ribossomos/metabolismo
2.
Front Immunol ; 15: 1450576, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39372409

RESUMO

Our case demonstrated unique cytomegalovirus (CMV) encephalitis post-haploidentical donor hematopoietic stem cell transplantation (HID-HSCT), with early findings on diffusion-weighted brain magnetic resonance imaging (MRI) in the absence of any neurologic symptoms. A 54-year-old Chinese man with acute lymphoblastic leukemia (Philadelphia chromosome-negative) underwent HID-HSCT. After HSCT, the patient developed CMV viremia and severe acute graft-versus-host disease. Recurrence of CMV viremia was observed. On day 129, brain MRI was performed to determine the cause for the intermittent fever. Diffusion-weighted imaging (DWI) revealed several bright spots in the cortex of the frontal lobes and anterior angle of the left lateral ventricle. Subsequently, he developed transplant-associated thrombotic microangiopathy, posterior reversible encephalopathy syndrome, and enlargement of lesions alongside the ventricular wall on a brain MRI series. Metagenomic next-generation sequencing (NGS) of the cerebrospinal fluid (CSF) led to the final diagnosis of CMV encephalitis. Although ganciclovir combined with foscarnet was administered, the patient's consciousness deteriorated, followed by respiratory failure. The patient died on day 198. Additionally, we performed a systematic review to comprehensively analyze this disease. Regarding treatment, immunological therapies, including virus-specific T cells from a third donor and CMV-cytotoxic T lymphocytes, may be more effective. This case report and systematic review underscores the complexities of managing CMV ventriculoencephalitis in HSCT recipients and emphasizes the importance of early diagnosis by brain MRI and CSF polymerase chain reaction or NGS and ongoing research in improving outcomes.


Assuntos
Infecções por Citomegalovirus , Transplante de Células-Tronco Hematopoéticas , Imageamento por Ressonância Magnética , Humanos , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Masculino , Infecções por Citomegalovirus/diagnóstico por imagem , Pessoa de Meia-Idade , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Encéfalo/virologia , Citomegalovirus/genética , Encefalite Viral/etiologia , Encefalite Viral/diagnóstico por imagem , Transplante Haploidêntico , Antivirais/uso terapêutico , Doença Enxerto-Hospedeiro/etiologia , Ventriculite Cerebral/diagnóstico por imagem , Ventriculite Cerebral/etiologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Evolução Fatal
3.
Virol J ; 21(1): 217, 2024 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-39277738

RESUMO

Japanese encephalitis is an acute infectious disease of the central nervous system caused by neurotropic Japanese encephalitis virus (JEV). As a member of TAM (Tyro3, Axl and Mertk) family, Mertk has involved in multiple biological processes by engaging with its bridging ligands Gas6 and Protein S, including invasion of pathogens, phagocytosis of apoptotic cells, inflammatory response regulation, and the maintenance of blood brain barrier (BBB) integrity. However, its role in encephalitis caused by JEV infection has not been studied in detail. Here, we found that Mertk-/- mice exhibited higher mortality and more rapid disease progression than wild-type mice after JEV challenge. There were no significant differences in viral load and cytokines expression level in peripheral tissues between Wild type and Mertk-/- mice. Furthermore, the absence of Mertk had little effect on the inflammatory response and immunopathological damage while it can cause an increased viral load in the brain. For the in vitro model of BBB, Mertk was shown to maintain the integrity of the BBB. In vivo, Mertk-/- mice exhibited higher BBB permeability and lower BBB integrity. Taken together, our findings demonstrate that Mertk acts as a protective factor in the development of encephalitis induced by JEV infection, which is mainly associated with its beneficial effect on BBB integrity, rather than its regulation of inflammatory response.


Assuntos
Barreira Hematoencefálica , Vírus da Encefalite Japonesa (Espécie) , Encefalite Japonesa , c-Mer Tirosina Quinase , Animais , Camundongos , Barreira Hematoencefálica/metabolismo , Encéfalo/virologia , Encéfalo/patologia , c-Mer Tirosina Quinase/metabolismo , c-Mer Tirosina Quinase/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Carga Viral
4.
Cells ; 13(17)2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39272987

RESUMO

Blood-brain barrier (BBB) injury and dysfunction following infection with the human immunodeficiency virus (HIV) enables viral entry into the brain, infection of resident brain cells, neuronal injury and subsequent neurodegeneration leading to HIV-associated neurocognitive disorders (HAND). Although combination antiretroviral therapy has significantly reduced the incidence and prevalence of acquired immunodeficiency syndrome and increased the life expectancy of people living with HIV, the prevalence of HAND remains high. With aging of people living with HIV associated with increased comorbidities, the prevalence of HIV-related central nervous system (CNS) complications is expected to remain high. Considering the principal role of the brain endothelium in HIV infection of the CNS and HAND, the purpose of this manuscript is to review the current literature on the pathobiology of the brain endothelium structural and functional dysregulation in HIV infection, including in the presence of HIV-1 and viral proteins (gp120, Tat, Nef, and Vpr). We summarize evidence from human and animal studies, in vitro studies, and associated mechanisms. We further summarize evidence of synergy or lack thereof between commonly abused substances (cocaine, methamphetamine, alcohol, tobacco, opioids, and cannabinoids) and HIV- or viral protein-induced BBB injury and dysfunction.


Assuntos
Barreira Hematoencefálica , Encéfalo , Infecções por HIV , Transtornos Relacionados ao Uso de Substâncias , Humanos , Infecções por HIV/patologia , Infecções por HIV/complicações , Transtornos Relacionados ao Uso de Substâncias/patologia , Transtornos Relacionados ao Uso de Substâncias/complicações , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Encéfalo/virologia , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/metabolismo , Animais , Endotélio/patologia , Endotélio/metabolismo , HIV-1
5.
PLoS Pathog ; 20(9): e1012168, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39283947

RESUMO

Human Immunodeficiency Virus (HIV) is widely acknowledged for its profound impact on the immune system. Although HIV primarily affects peripheral CD4 T cells, its influence on the central nervous system (CNS) cannot be overlooked. Within the brain, microglia and CNS-associated macrophages (CAMs) serve as the primary targets for HIV and the simian immunodeficiency virus (SIV) in nonhuman primates. This infection can lead to neurological effects and establish a viral reservoir. Given the gaps in our understanding of how these cells respond in vivo to acute CNS infection, we conducted single-cell RNA sequencing (scRNA-seq) on myeloid cells from the brains of three rhesus macaques 12 days after SIV infection, along with three uninfected controls. Our analysis revealed six distinct microglial clusters including homeostatic microglia, preactivated microglia, and activated microglia expressing high levels of inflammatory and disease-related molecules. In response to acute SIV infection, the homeostatic and preactivated microglia population decreased, while the activated and disease-related microglia increased. All microglial clusters exhibited upregulation of MHC class I molecules and interferon-related genes, indicating their crucial roles in defending against SIV during the acute phase. All microglia clusters also upregulated genes linked to cellular senescence. Additionally, we identified two distinct CAM populations: CD14lowCD16hi and CD14hiCD16low CAMs. Interestingly, during acute SIV infection, the dominant CAM population changed to one with an inflammatory phenotype. Specific upregulated genes within one microglia and one macrophage cluster were associated with neurodegenerative pathways, suggesting potential links to neurocognitive disorders. This research sheds light on the intricate interactions between viral infection, innate immune responses, and the CNS, providing valuable insights for future investigations.


Assuntos
Macaca mulatta , Macrófagos , Microglia , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Análise de Célula Única , Animais , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Microglia/imunologia , Microglia/virologia , Vírus da Imunodeficiência Símia/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Sistema Nervoso Central/virologia , Sistema Nervoso Central/imunologia , Encéfalo/virologia , Encéfalo/imunologia , Encéfalo/patologia
6.
mBio ; 15(10): e0152224, 2024 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-39189744

RESUMO

Extracellular vesicles (EV), key players in cell-to-cell communication, may contribute to disease propagation in several neurodegenerative diseases, including Alzheimer's disease (AD), by favoring the dissemination of neurotoxic proteins within the brain. Interestingly, growing evidence supports the role of herpes simplex virus type 1 (HSV-1) infection in the pathogenesis of AD. Here, we investigated whether HSV-1 infection could promote the spread of phosphorylated tau (ptau) among neurons via EV. We analyzed the ptau species that were secreted via EV following HSV-1 infection in neuroblastoma cells and primary neurons, focusing particularly on T205, T181, and T217, the phosphorylation sites mainly associated with AD. Moreover, by overexpressing human tau tagged with GFP (htauGFP), we found that recipient tau knockout (KO) neurons uptook EV that are loaded with HSV-1-induced phtauGFP. Finally, we exploited an in vivo model of acute infection and assessed that cerebral HSV-1 infection promotes the release of ptau via EV in the brain of infected mice. Overall, our data suggest that, following HSV-1 infection, EV play a role in tau spreading within the brain, thus contributing to neurodegeneration.IMPORTANCEHerpes simplex virus type 1 (HSV-1) infection that reaches the brain has been repeatedly linked with the appearance of the pathognomonic markers of Alzheimer's disease (AD), including accumulation of amyloid beta and hyperphosphorylated tau proteins, and cognitive deficits. AD is a multifactorial neurodegenerative disease representing the most common form of dementia in the elderly, and no cure is currently available, thus prompting additional investigation on potential risk factors and pathological mechanisms. Here, we demonstrate that the virus exploits the extracellular vesicles (EV) to disseminate phosphorylated tau (ptau) among brain cells. Importantly, we provide evidence that the HSV-1-induced EV-bearing ptau can be undertaken by recipient neurons, thus likely contributing to misfolding and aggregation of native tau, as reported for other AD models. Hence, our data highlight a novel mechanism exploited by HSV-1 to propagate tau-related damage in the brain.


Assuntos
Vesículas Extracelulares , Herpes Simples , Herpesvirus Humano 1 , Neurônios , Proteínas tau , Proteínas tau/metabolismo , Proteínas tau/genética , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/virologia , Neurônios/virologia , Neurônios/metabolismo , Animais , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/genética , Camundongos , Fosforilação , Humanos , Herpes Simples/virologia , Herpes Simples/metabolismo , Encéfalo/virologia , Encéfalo/metabolismo , Encéfalo/patologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/virologia , Camundongos Endogâmicos C57BL
7.
J Neurovirol ; 30(4): 434-440, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39155353

RESUMO

Demyelinating central nervous system (CNS) disorders are a diverse group of conditions characterised by damage to the myelin sheath. These include not only primary autoimmune disorders such as multiple sclerosis (MS) or neuromyelitis optica spectrum disorder (NMOSD), but secondary demyelinating conditions caused by infection and neoplasm, where immunosuppressive therapy may worsen the condition or delay definitive treatment. We describe a young man with an unusual presentation of CNS demyelinating disease associated with HIV infection and positive syphilis serology. MRI brain and spine showed a demyelinating tumefactive lesion accompanied by longitudinal extensive transverse myelitis, and we initially suspected NMOSD. However anti-aquaporin 4 antibodies were negative, going against a diagnosis of NMOSD and he then tested positive for HIV which led us to consider TB myelitis, neurosyphilis and HIV vacuolar myelopathy. He was commenced on highly active retroviral therapy and treated with steroids and immunosuppression. He did not respond to treatment as expected so a brain biopsy was required to narrow the differential. Brain biopsy initially raised the possibility of progressive multifocal leukoencephalopathy which is associated with infection with the John Cunningham (JC) virus. Ultimately JC Virus PCR on the biopsy was negative, the final report suggesting nonspecific active chronic inflammation. We detail his clinical course and the diagnostic challenges along the way.


Assuntos
Infecções por HIV , Imageamento por Ressonância Magnética , Humanos , Masculino , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , Infecções por HIV/imunologia , Adulto , Encéfalo/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/virologia , Terapia Antirretroviral de Alta Atividade , Neurossífilis/tratamento farmacológico , Neurossífilis/complicações , Neurossífilis/virologia , Neurossífilis/diagnóstico , Neurossífilis/patologia , Neurossífilis/diagnóstico por imagem , Mielite Transversa/virologia , Mielite Transversa/tratamento farmacológico , Mielite Transversa/diagnóstico por imagem , Mielite Transversa/patologia , Doenças Desmielinizantes/virologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/diagnóstico por imagem , Doenças Desmielinizantes/tratamento farmacológico , Doenças Desmielinizantes/imunologia
8.
EMBO Rep ; 25(8): 3276-3299, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39039298

RESUMO

Transmigration of circulating monocytes from the bloodstream to tissues represents an early hallmark of inflammation. This process plays a pivotal role during viral neuroinvasion, encephalitis, and HIV-associated neurocognitive disorders. How monocytes locally unzip endothelial tight junction-associated proteins (TJAPs), without perturbing impermeability, to reach the central nervous system remains poorly understood. Here, we show that human circulating monocytes express the TJAP Occludin (OCLN) to promote transmigration through endothelial cells. We found that human monocytic OCLN (hmOCLN) clusters at monocyte-endothelium interface, while modulation of hmOCLN expression significantly impacts monocyte transmigration. Furthermore, we designed OCLN-derived peptides targeting its extracellular loops (EL) and show that transmigration of treated monocytes is inhibited in vitro and in zebrafish embryos, while preserving vascular integrity. Monocyte transmigration toward the brain is an important process for HIV neuroinvasion and we found that the OCLN-derived peptides significantly inhibit HIV dissemination to cerebral organoids. In conclusion, our study identifies an important role for monocytic OCLN during transmigration and provides a proof-of-concept for the development of mitigation strategies to prevent monocyte infiltration and viral neuroinvasion.


Assuntos
Células Endoteliais , Monócitos , Ocludina , Migração Transendotelial e Transepitelial , Peixe-Zebra , Ocludina/metabolismo , Ocludina/genética , Humanos , Monócitos/metabolismo , Monócitos/efeitos dos fármacos , Monócitos/virologia , Animais , Migração Transendotelial e Transepitelial/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Células Endoteliais/efeitos dos fármacos , Infecções por HIV/virologia , Infecções por HIV/tratamento farmacológico , HIV-1/fisiologia , HIV-1/efeitos dos fármacos , Peptídeos/farmacologia , Peptídeos/metabolismo , Encéfalo/metabolismo , Encéfalo/virologia
9.
Brain ; 147(9): 3059-3069, 2024 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-39049445

RESUMO

Perivascular macrophages (PVMs) and, to a lesser degree, microglia are targets and reservoirs of HIV and simian immunodeficiency virus (SIV) in the brain. Previously, we demonstrated that colony-stimulating factor 1 receptor (CSF1R) in PVMs was upregulated and activated in chronically SIV-infected rhesus macaques with encephalitis, correlating with SIV infection of PVMs. Herein, we investigated the role of CSF1R in the brain during acute SIV infection using BLZ945, a brain-penetrant CSF1R kinase inhibitor. Apart from three uninfected historic controls, nine Indian rhesus macaques were infected acutely with SIVmac251 and divided into three groups (n = 3 each): an untreated control and two groups treated for 20-30 days with low- (10 mg/kg/day) or high- (30 mg/kg/day) dose BLZ945. With the high-dose BLZ945 treatment, there was a significant reduction in cells expressing CD163 and CD206 across all four brain areas examined, compared with the low-dose treatment and control groups. In 9 of 11 tested regions, tissue viral DNA (vDNA) loads were reduced by 95%-99% following at least one of the two doses, and even to undetectable levels in some instances. Decreased numbers of CD163+ and CD206+ cells correlated significantly with lower levels of vDNA in all four corresponding brain areas. In contrast, BLZ945 treatment did not significantly affect the number of microglia. Our results indicate that doses as low as 10 mg/kg/day of BLZ945 are sufficient to reduce the tissue vDNA loads in the brain with no apparent adverse effect. This study provides evidence that infected PVMs are highly sensitive to CSF1R inhibition, opening new possibilities to achieve viral clearance.


Assuntos
Encéfalo , Macaca mulatta , Macrófagos , Síndrome de Imunodeficiência Adquirida dos Símios , Vírus da Imunodeficiência Símia , Animais , Síndrome de Imunodeficiência Adquirida dos Símios/tratamento farmacológico , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/efeitos dos fármacos , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/virologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Carga Viral/efeitos dos fármacos , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Antígenos CD/metabolismo , Masculino , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/virologia , Antígenos de Diferenciação Mielomonocítica/metabolismo , Receptores de Superfície Celular/metabolismo , Anisóis
10.
mBio ; 15(8): e0104924, 2024 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-38953354

RESUMO

Polyomaviruses are species-specific DNA viruses that can cause disease in immunocompromised individuals. Despite their role as the causative agents for several diseases, there are no currently approved antivirals for treating polyomavirus infection. Brincidofovir (BCV) is an antiviral approved for the treatment of poxvirus infections and has shown activity against other double-stranded DNA viruses. In this study, we tested the efficacy of BCV against polyomavirus infection in vitro and in vivo using mouse polyomavirus (MuPyV). BCV inhibited virus production in primary mouse kidney cells and brain cortical cells. BCV treatment of cells transfected with MuPyV genomic DNA resulted in a reduction in virus levels, indicating that viral inhibition occurs post-entry. Although in vitro BCV treatment had a limited effect on viral DNA and RNA levels, drug treatment was associated with a reduction in viral protein, raising the possibility that BCV acts post-transcriptionally to inhibit MuPyV infection. In mice, BCV treatment was well tolerated, and prophylactic treatment resulted in a reduction in viral DNA levels and a potent suppression of infectious virus production in the kidney and brain. In mice with chronic polyomavirus infection, therapeutic administration of BCV decreased viremia and reduced infection in the kidney. These data demonstrate that BCV exerts antiviral activity against polyomavirus infection in vivo, supporting further investigation into the use of BCV to treat clinical polyomavirus infections. IMPORTANCE: Widespread in the human population and able to persist asymptomatically for the life of an individual, polyomavirus infections cause a significant disease burden in the immunocompromised. Individuals undergoing immune suppression, such as kidney transplant patients or those treated for autoimmune diseases, are particularly at high risk for polyomavirus-associated diseases. Because no antiviral agent exists for treating polyomavirus infections, management of polyomavirus-associated diseases typically involves reducing or discontinuing immunomodulatory therapy. This can be perilous due to the risk of transplant rejection and the potential development of adverse immune reactions. Thus, there is a pressing need for the development of antivirals targeting polyomaviruses. Here, we investigate the effects of brincidofovir, an FDA-approved antiviral, on polyomavirus infection in vivo using mouse polyomavirus. We show that the drug is well-tolerated in mice, reduces infectious viral titers, and limits viral pathology, indicating the potential of brincidofovir as an anti-polyomavirus therapeutic.


Assuntos
Antivirais , Citosina , Organofosfonatos , Infecções por Polyomavirus , Polyomavirus , Animais , Citosina/análogos & derivados , Citosina/farmacologia , Citosina/uso terapêutico , Infecções por Polyomavirus/tratamento farmacológico , Infecções por Polyomavirus/virologia , Polyomavirus/efeitos dos fármacos , Camundongos , Antivirais/farmacologia , Antivirais/uso terapêutico , Organofosfonatos/farmacologia , Organofosfonatos/uso terapêutico , Replicação Viral/efeitos dos fármacos , Rim/virologia , Rim/efeitos dos fármacos , Feminino , DNA Viral/genética , Células Cultivadas , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Encéfalo/virologia
11.
J Gen Virol ; 105(7)2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38995681

RESUMO

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection is associated with neurological sequelae including haemorrhage, thrombosis and ischaemic necrosis and encephalitis. However, the mechanism by which this occurs is unclear. Neurological disease associated with COVID-19 has been proposed to occur following direct infection of the central nervous system and/or indirectly by local or systemic immune activation. We evaluated the expression of angiotensin-converting enzyme-2 and transmembrane protease, serine 2 (TMPRSS2) in brain tissue from five healthy human donors and observed low-level expression of these proteins in cells morphologically consistent with astrocytes, neurons and choroidal ependymal cells within the frontal cortex and medulla oblongata. Primary human astrocytes, neurons, choroid plexus epithelial cells and pericytes supported productive SARS-CoV-2 infection with ancestral, Alpha, Delta and Omicron variants. Infected cells supported the full viral life cycle, releasing infectious virus particles. In contrast, primary brain microvascular endothelial cells and microglia were refractory to SARS-CoV-2 infection. These data support a model whereby SARS-CoV-2 can infect human brain cells, and the mechanism of viral entry warrants further investigation.


Assuntos
Enzima de Conversão de Angiotensina 2 , Astrócitos , COVID-19 , Plexo Corióideo , Células Epiteliais , Neurônios , Pericitos , SARS-CoV-2 , Serina Endopeptidases , Humanos , Pericitos/virologia , SARS-CoV-2/fisiologia , Astrócitos/virologia , Plexo Corióideo/virologia , Enzima de Conversão de Angiotensina 2/metabolismo , Enzima de Conversão de Angiotensina 2/genética , Neurônios/virologia , COVID-19/virologia , COVID-19/patologia , Células Epiteliais/virologia , Serina Endopeptidases/metabolismo , Serina Endopeptidases/genética , Células Cultivadas , Encéfalo/virologia , Encéfalo/patologia , Sistema Nervoso Central/virologia
12.
Cells ; 13(13)2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38994979

RESUMO

HIV-associated neurocognitive disorders (HAND) persist under antiretroviral therapy as a complex pathology that has been difficult to study in cellular and animal models. Therefore, we generated an ex vivo human brain slice model of HIV-1 infection from surgically resected adult brain tissue. Brain slice cultures processed for flow cytometry showed >90% viability of dissociated cells within the first three weeks in vitro, with parallel detection of astrocyte, myeloid, and neuronal populations. Neurons within brain slices showed stable dendritic spine density and mature spine morphologies in the first weeks in culture, and they generated detectable activity in multi-electrode arrays. We infected cultured brain slices using patient-matched CD4+ T-cells or monocyte-derived macrophages (MDMs) that were exposed to a GFP-expressing R5-tropic HIV-1 in vitro. Infected slice cultures expressed viral RNA and developed a spreading infection up to 9 days post-infection, which were significantly decreased by antiretrovirals. We also detected infected myeloid cells and astrocytes within slices and observed minimal effect on cellular viability over time. Overall, this human-centered model offers a promising resource to study the cellular mechanisms contributing to HAND (including antiretroviral toxicity, substance use, and aging), infection of resident brain cells, and new neuroprotective therapeutics.


Assuntos
Encéfalo , Infecções por HIV , HIV-1 , Humanos , Encéfalo/virologia , Encéfalo/patologia , HIV-1/fisiologia , Infecções por HIV/virologia , Infecções por HIV/patologia , Adulto , Neurônios/virologia , Neurônios/metabolismo , Macrófagos/virologia , Macrófagos/metabolismo , Astrócitos/virologia , Linfócitos T CD4-Positivos/virologia , Técnicas de Cultura de Tecidos
13.
CNS Neurosci Ther ; 30(6): e14822, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38923860

RESUMO

INTRODUCTION: Despite the extensive neurological symptoms induced by COVID-19 and the identification of SARS-CoV-2 in post-mortem brain samples from COVID-19 patients months after death, the precise mechanisms of SARS-CoV-2 invasion into the central nervous system remain unclear due to the lack of research models. METHODS: We collected glioma tissue samples from glioma patients who had a recent history of COVID-19 and examined the presence of the SARS-CoV-2 spike protein. Subsequently, spatial transcriptomic analyses were conducted on normal brain tissues, glioma tissues, and glioma tissues from glioma patients with recent COVID-19 history. Additionally, single-cell sequencing data from both glioma tissues and glioma organoids were collected and analyzed. Glioma organoids were utilized to evaluate the efficacy of potential COVID-19 blocking agents. RESULTS: Glioma tissues from glioma patients with recent COVID-19 history exhibited the presence of the SARS-CoV-2 spike protein. Differences between glioma tissues from glioma patients who had a recent history of COVID-19 and healthy brain tissues primarily manifested in neuronal cells. Notably, neuronal cells within glioma tissues of COVID-19 history demonstrated heightened susceptibility to Alzheimer's disease, depression, and synaptic dysfunction, indicative of neuronal aberrations. Expressions of SARS-CoV-2 entry factors were confirmed in both glioma tissues and glioma organoids. Moreover, glioma organoids were susceptible to pseudo-SARS-CoV-2 infection and the infections could be partly blocked by the potential COVID-19 drugs. CONCLUSIONS: Gliomas had inherent traits that render them susceptible to SARS-CoV-2 infection, leading to their representability of COVID-19 neurological symptoms. This established a biological foundation for the rationality and feasibility of utilization of glioma organoids as research and blocking drug testing model in SARS-CoV-2 infection within the central nervous system.


Assuntos
Neoplasias Encefálicas , COVID-19 , Glioma , Organoides , SARS-CoV-2 , Humanos , Glioma/patologia , Glioma/virologia , COVID-19/complicações , COVID-19/patologia , Organoides/virologia , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/virologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Masculino , Feminino , Pessoa de Meia-Idade , Encéfalo/patologia , Encéfalo/virologia , Encéfalo/metabolismo
14.
Front Cell Infect Microbiol ; 14: 1423394, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38887492

RESUMO

Extracellular vesicles (EVs) are membrane-bound vesicles secreted by all cell types that play a central role in cell-to-cell communication. Since these vesicles serve as vehicles of cellular content (nucleic acids, proteins and lipids) with the potential to cross biological barriers, they represent a novel attractive window into an otherwise inaccessible organ, such as the brain. The composition of EVs is cell-type specific and mirrors the physiological condition of the cell-of-origin. Consequently, during viral infection, EVs undergo significant changes in their content and morphology, thereby reflecting alterations in the cellular state. Here, we briefly summarize the potential of brain-derived EVs as a lens into viral infection in the central nervous system, thereby: 1) uncovering underlying pathophysiological processes at play and 2) serving as liquid biopsies of the brain, representing a non-invasive source of biomarkers for monitoring disease activity. Although translating the potential of EVs from research to diagnosis poses complexities, characterizing brain-derived EVs in the context of viral infections holds promise to enhance diagnostic and therapeutic strategies, offering new avenues for managing infectious neurological diseases.


Assuntos
Biomarcadores , Encéfalo , Vesículas Extracelulares , Viroses , Vesículas Extracelulares/metabolismo , Humanos , Biomarcadores/metabolismo , Encéfalo/patologia , Encéfalo/metabolismo , Encéfalo/virologia , Viroses/metabolismo , Animais , Comunicação Celular
15.
Int J Mol Sci ; 25(11)2024 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-38891803

RESUMO

Rabies virus (RABV) is a neurotropic virus that causes fatal neurological disease, raising serious public health issues and attracting extensive attention in society. To elucidate the molecular mechanism of RABV-induced neuronal damage, we used hematoxylin-eosin staining, transmission electron microscopy, transcriptomics analysis, and immune response factor testing to investigate RABV-infected neurons. We successfully isolated the neurons from murine brains. The specificity of the isolated neurons was identified by a monoclonal antibody, and the viability of the neurons was 83.53-95.0%. We confirmed that RABV infection induced serious damage to the neurons according to histochemistry and transmission electron microscope (TEM) scanning. In addition, the transcriptomics analysis suggested that multiple genes related to the pyroptosis pathway were significantly upregulated, including gasdermin D (Gsdmd), Nlrp3, caspase-1, and IL-1ß, as well as the chemokine genes Ccl2, Ccl3, Ccl4, Ccl5, Ccl7, Ccl12, and Cxcl10. We next verified this finding in the brains of mice infected with the rRC-HL, GX074, and challenge virus standard strain-24 (CVS-24) strains of RABV. Importantly, we found that the expression level of the Gsdmd protein was significantly upregulated in the neurons infected with different RABV strains and ranged from 691.1 to 5764.96 pg/mL, while the basal level of mock-infected neurons was less than 100 pg/mL. Taken together, our findings suggest that Gsdmd-induced pyroptosis is involved in the neuron damage caused by RABV infection.


Assuntos
Neurônios , Proteínas de Ligação a Fosfato , Piroptose , Vírus da Raiva , Raiva , Animais , Neurônios/virologia , Neurônios/metabolismo , Neurônios/patologia , Vírus da Raiva/patogenicidade , Vírus da Raiva/fisiologia , Raiva/virologia , Raiva/patologia , Raiva/metabolismo , Camundongos , Proteínas de Ligação a Fosfato/metabolismo , Proteínas de Ligação a Fosfato/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Encéfalo/virologia , Encéfalo/patologia , Encéfalo/metabolismo , Gasderminas
16.
J Neuroinflammation ; 21(1): 163, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918792

RESUMO

BACKGROUND: The SARS-CoV-2 virus activates maternal and placental immune responses. Such activation in the setting of other infections during pregnancy is known to impact fetal brain development. The effects of maternal immune activation on neurodevelopment are mediated at least in part by fetal brain microglia. However, microglia are inaccessible for direct analysis, and there are no validated non-invasive surrogate models to evaluate in utero microglial priming and function. We have previously demonstrated shared transcriptional programs between microglia and Hofbauer cells (HBCs, or fetal placental macrophages) in mouse models. METHODS AND RESULTS: We assessed the impact of maternal SARS-CoV-2 on HBCs isolated from 24 term placentas (N = 10 SARS-CoV-2 positive cases, 14 negative controls). Using single-cell RNA-sequencing, we demonstrated that HBC subpopulations exhibit distinct cellular programs, with specific subpopulations differentially impacted by SARS-CoV-2. Assessment of differentially expressed genes implied impaired phagocytosis, a key function of both HBCs and microglia, in some subclusters. Leveraging previously validated models of microglial synaptic pruning, we showed that HBCs isolated from placentas of SARS-CoV-2 positive pregnancies can be transdifferentiated into microglia-like cells (HBC-iMGs), with impaired synaptic pruning behavior compared to HBC models from negative controls. CONCLUSION: These findings suggest that HBCs isolated at birth can be used to create personalized cellular models of offspring microglial programming.


Assuntos
COVID-19 , Macrófagos , Microglia , Placenta , Complicações Infecciosas na Gravidez , SARS-CoV-2 , Feminino , Gravidez , Microglia/virologia , Humanos , Placenta/virologia , COVID-19/imunologia , Macrófagos/virologia , Complicações Infecciosas na Gravidez/virologia , Complicações Infecciosas na Gravidez/patologia , SARS-CoV-2/patogenicidade , Feto , Adulto , Encéfalo/virologia , Encéfalo/patologia , Camundongos , Animais
17.
Vet Res ; 55(1): 71, 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38822398

RESUMO

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.


Assuntos
Vacina BCG , Encéfalo , COVID-19 , SARS-CoV-2 , Animais , Camundongos , Vacina BCG/administração & dosagem , COVID-19/imunologia , COVID-19/virologia , SARS-CoV-2/fisiologia , Encéfalo/patologia , Encéfalo/virologia , Carga Viral , Pulmão/patologia , Pulmão/virologia , Pulmão/imunologia , Enzima de Conversão de Angiotensina 2/metabolismo , Camundongos Transgênicos , Feminino
18.
J Neurovirol ; 30(3): 215-228, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38922550

RESUMO

The cellular prion protein (PrPC) is an extracellular cell membrane protein. Due to its diversified roles, a definite role of PrPC has been difficult to establish. During viral infection, PrPC has been reported to play a pleiotropic role. Here, we have attempted to envision the function of PrPC in the neurotropic m-CoV-MHV-RSA59-induced model of neuroinflammation in C57BL/6 mice. A significant upregulation of PrPC at protein and mRNA levels was evident in infected mouse brains during the acute phase of neuroinflammation. Furthermore, investigation of the effect of MHV-RSA59 infection on PrPC expression in specific neuronal, microglial, and astrocytoma cell lines, revealed a differential expression of prion protein during neuroinflammation. Additionally, siRNA-mediated downregulation of prnp transcripts reduced the expression of viral antigen and viral infectivity in these cell lines. Cumulatively, our results suggest that PrPC expression significantly increases during acute MHV-RSA59 infection and that PrPC also assists in viral infectivity and viral replication.


Assuntos
Camundongos Endogâmicos C57BL , Microglia , Vírus da Hepatite Murina , Doenças Neuroinflamatórias , Proteínas PrPC , Animais , Vírus da Hepatite Murina/patogenicidade , Camundongos , Proteínas PrPC/metabolismo , Proteínas PrPC/genética , Doenças Neuroinflamatórias/virologia , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/imunologia , Doenças Neuroinflamatórias/genética , Doenças Neuroinflamatórias/patologia , Microglia/metabolismo , Microglia/virologia , Microglia/patologia , Encéfalo/virologia , Encéfalo/metabolismo , Encéfalo/patologia , Neurônios/virologia , Neurônios/metabolismo , Neurônios/patologia , Replicação Viral , Linhagem Celular Tumoral , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Regulação para Cima , Linhagem Celular , Humanos , Modelos Animais de Doenças , Proteínas Priônicas
19.
J Neurovirol ; 30(2): 202-207, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38778005

RESUMO

Tumefactive demyelinating lesions (TDL) are a rare occurrence among inflammatory demyelinating diseases of the central nervous system, distinguished by tumor-like lesions exceeding 2 cm in diameter. While various etiologies have been associated with TDL, only a limited number of case reports document the coexistence of acute disseminated encephalomyelitis (ADEM) and TDL. Here, we present the case of a female diagnosed with dengue fever two weeks prior, who subsequently developed left hemiparesis and encephalopathy. Both her brain magnetic resonance imaging (MRI) and clinical course align with the characteristics of tumefactive ADEM.


Assuntos
Vírus da Dengue , Dengue , Encefalomielite Aguda Disseminada , Imageamento por Ressonância Magnética , Humanos , Encefalomielite Aguda Disseminada/diagnóstico por imagem , Encefalomielite Aguda Disseminada/virologia , Encefalomielite Aguda Disseminada/patologia , Feminino , Dengue/complicações , Dengue/virologia , Dengue/diagnóstico por imagem , Dengue/patologia , Vírus da Dengue/patogenicidade , Vírus da Dengue/genética , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Encéfalo/virologia
20.
Viruses ; 16(5)2024 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-38793666

RESUMO

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.


Assuntos
Enzima de Conversão de Angiotensina 2 , Astrócitos , Barreira Hematoencefálica , COVID-19 , Dipeptidil Peptidase 4 , Pericitos , SARS-CoV-2 , Transcitose , Internalização do Vírus , Barreira Hematoencefálica/virologia , Barreira Hematoencefálica/metabolismo , Humanos , SARS-CoV-2/fisiologia , Enzima de Conversão de Angiotensina 2/metabolismo , Pericitos/virologia , Pericitos/metabolismo , COVID-19/virologia , COVID-19/metabolismo , Astrócitos/virologia , Astrócitos/metabolismo , Dipeptidil Peptidase 4/metabolismo , Encéfalo/virologia , Encéfalo/metabolismo , Endocitose , Células Endoteliais da Veia Umbilical Humana/virologia , Permeabilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA