Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 176
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; : e0039724, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38869283

RESUMEN

Enterovirus D68 (EV-D68) is an emerging pathogen that can cause severe respiratory and neurologic disease [acute flaccid myelitis (AFM)]. Intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with US/IL/14-18952 (IL52), a clinical isolate from the 2014 EV-D68 epidemic, results in many of the pathogenic features of human AFM, including viral infection of the spinal cord, death of motor neurons, and resultant progressive paralysis. In distinction, CA/14-4231 (CA4231), another clinical isolate from the 2014 EV-D68 outbreak, does not cause paralysis in mice, does not grow in the spinal cord, and does not cause motor neuron loss following IM injection. A panel of chimeric viruses containing sequences from IL52 and CA4231 was used to demonstrate that VP1 is the main determinant of EV-D68 neurovirulence following IM injection of neonatal SW mice. VP1 contains four amino acid differences between IL52 and CA4231. Mutations resulting in substituting these four amino acids (CA4231 residues into the IL52 polyprotein) completely abolished neurovirulence. Conversely, mutations resulting in substituting VP1 IL52 amino acid residues into the CA4231 polyprotein created a virus that induced paralysis to the same degree as IL52. Neurovirulence following infection of neonatal SW mice with parental and chimeric viruses was associated with viral growth in the spinal cord. IMPORTANCE: Emerging viruses allow us to investigate mutations leading to increased disease severity. Enterovirus D68 (EV-D68), once the cause of rare cases of respiratory illness, recently acquired the ability to cause severe respiratory and neurologic disease. Chimeric viruses were used to demonstrate that viral structural protein VP1 determines growth in the spinal cord, motor neuron loss, and paralysis following intramuscular (IM) injection of neonatal Swiss Webster (SW) mice with EV-D68. These results have relevance for predicting the clinical outcome of future EV-D68 epidemics as well as targeting retrograde transport as a potential strategy for treating virus-induced neurologic disease.

2.
J Virol ; 97(3): e0180522, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36802227

RESUMEN

West Nile virus (WNV) is the leading cause of epidemic arboviral encephalitis in the United States. As there are currently no proven antiviral therapies or licensed human vaccines, understanding the neuropathogenesis of WNV is critical for rational therapeutic design. In WNV-infected mice, the depletion of microglia leads to enhanced viral replication, increased central nervous system (CNS) tissue injury, and increased mortality, suggesting that microglia play a critical role in protection against WNV neuroinvasive disease. To determine if augmenting microglial activation would provide a potential therapeutic strategy, we administered granulocyte-macrophage colony-stimulating factor (GM-CSF) to WNV-infected mice. Recombinant human GM-CSF (rHuGMCSF) (sargramostim [Leukine]) is an FDA-approved drug used to increase white blood cells following leukopenia-inducing chemotherapy or bone marrow transplantation. Daily treatment of both uninfected and WNV-infected mice with subcutaneous injections of GM-CSF resulted in microglial proliferation and activation as indicated by the enhanced expression of the microglia activation marker ionized calcium binding adaptor molecule 1 (Iba1) and several microglia-associated inflammatory cytokines, including CCL2 (C-C motif chemokine ligand 2), interleukin 6 (IL-6), and IL-10. In addition, more microglia adopted an activated morphology as demonstrated by increased sizes and more pronounced processes. GM-CSF-induced microglial activation in WNV-infected mice was associated with reduced viral titers and apoptotic activity (caspase 3) in the brains of WNV-infected mice and significantly increased survival. WNV-infected ex vivo brain slice cultures (BSCs) treated with GM-CSF also showed reduced viral titers and caspase 3 apoptotic cell death, indicating that GM-CSF specifically targets the CNS and that its actions are not dependent on peripheral immune activity. Our studies suggest that stimulation of microglial activation may be a viable therapeutic approach for the treatment of WNV neuroinvasive disease. IMPORTANCE Although rare, WNV encephalitis poses a devastating health concern, with few treatment options and frequent long-term neurological sequelae. Currently, there are no human vaccines or specific antivirals against WNV infections, so further research into potential new therapeutic agents is critical. This study presents a novel treatment option for WNV infections using GM-CSF and lays the foundation for further studies into the use of GM-CSF as a treatment for WNV encephalitis as well as a potential treatment for other viral infections.


Asunto(s)
Encéfalo , Fiebre del Nilo Occidental , Animales , Ratones , Encéfalo/virología , Caspasa 3/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Fiebre del Nilo Occidental/terapia , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/fisiología , Carga Viral/fisiología , Microglía/citología , Microglía/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas Recombinantes/farmacología
3.
J Virol ; 97(5): e0015623, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37154751

RESUMEN

In 2014, 2016, and 2018, the United States experienced unprecedented spikes in pediatric cases of acute flaccid myelitis (AFM), which is a poliomyelitis-like paralytic illness. Accumulating clinical, immunological, and epidemiological evidence has identified enterovirus D68 (EV-D68) as a major causative agent of these biennial AFM outbreaks. There are currently no available FDA-approved antivirals that are effective against EV-D68, and the treatment for EV-D68-associated AFM is primarily supportive. Telaprevir is an food and drug administration (FDA)-approved protease inhibitor that irreversibly binds the EV-D68 2A protease and inhibits EV-D68 replication in vitro. Here, we utilize a murine model of EV-D68 associated AFM to show that early telaprevir treatment improves paralysis outcomes in Swiss Webster (SW) mice. Telaprevir reduces both viral titer and apoptotic activity in both muscles and spinal cords at early disease time points, which results in improved AFM outcomes in infected mice. Following intramuscular inoculation in mice, EV-D68 infection results in a stereotypic pattern of weakness that is reflected by the loss of the innervating motor neuron population, in sequential order, of the ipsilateral (injected) hindlimb, the contralateral hindlimb, and then the forelimbs. Telaprevir treatment preserved motor neuron populations and reduced weakness in limbs beyond the injected hindlimb. The effects of telaprevir were not seen when the treatment was delayed, and toxicity limited doses beyond 35 mg/kg. These studies are a proof of principle, provide the first evidence of benefit of an FDA-approved antiviral drug with which to treat AFM, and emphasize both the need to develop better tolerated therapies that remain efficacious when administered after viral infections and the development of clinical symptoms. IMPORTANCE Recent outbreaks of EV-D68 in 2014, 2016, and 2018 have resulted in over 600 cases of a paralytic illness that is known as AFM. AFM is a predominantly pediatric disease with no FDA-approved treatment, and many patients show minimal recovery from limb weakness. Telaprevir is an FDA-approved antiviral that has been shown to inhibit EV-D68 in vitro. Here, we demonstrate that a telaprevir treatment that is given concurrently with an EV-D68 infection improves AFM outcomes in mice by reducing apoptosis and viral titers at early time points. Telaprevir also protected motor neurons and improved paralysis outcomes in limbs beyond the site of viral inoculation. This study improves understanding of EV-D68 pathogenesis in the mouse model of AFM. This study serves as a proof of principle for the first FDA-approved drug that has been shown to improve AFM outcomes and have in vivo efficacy against EV-D68 as well as underlines the importance of the continued development of EV-D68 antivirals.


Asunto(s)
Enfermedades Virales del Sistema Nervioso Central , Enterovirus Humano D , Infecciones por Enterovirus , Animales , Estados Unidos , Ratones , Enterovirus Humano D/fisiología , Modelos Animales de Enfermedad , Parálisis/tratamiento farmacológico , Parálisis/etiología , Infecciones por Enterovirus/patología , Antivirales/farmacología , Antivirales/uso terapéutico
4.
Ann Neurol ; 93(5): 893-905, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36602053

RESUMEN

OBJECTIVE: Monkeypox virus (MPXV) disease has been declared a public health emergency by the World Health Organization, creating an urgent need for neurologists to be able to recognize, diagnosis, and treat MPXV-associated neurologic disease. METHODS: Three cases of MPXV-associated central nervous system (CNS) disease occurring during the 2022 outbreak, and their associated imaging findings are presented, with 2 cases previously published in a limited capacity in a public health bulletin. RESULTS: Three previously healthy immunocompetent gay men in their 30s developed a febrile illness followed by progressive neurologic symptoms with presence of a vesiculopustular rash. MPXV nucleic acid was detected by polymerase chain reaction (PCR) from skin lesions of 2 patients, with the third patient having indeterminate testing but an epidemiologic link to a confirmed MPXV disease case. Cerebrospinal fluid demonstrated a lymphocytic pleocytosis, elevated protein, and negative MPXV-specific PCR. In 2 patients, magnetic resonance imaging of the brain and spine demonstrated partially enhancing, longitudinally extensive central spinal cord lesions with multifocal subcortical, basal ganglia, thalamic, cerebellar, and/or brainstem lesions. The third patient had thalamic and basal ganglia lesions. All patients received 14 days of tecovirimat, and 2 patients also received multiple forms of immunotherapy, including intravenous immunoglobulin, pulsed high-dose steroids, plasmapheresis, and/or rituximab. Good neurologic recovery was observed in all cases. INTERPRETATION: MPXV can be associated with CNS disease. It is unclear whether this is from a parainfectious immune-mediated injury or direct CNS viral invasion. ANN NEUROL 2023;93:893-905.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Mpox , Humanos , Masculino , Enfermedades del Sistema Nervioso Central/virología , Imagen por Resonancia Magnética , Mpox/diagnóstico , Mpox/patología , Monkeypox virus/fisiología
5.
Emerg Infect Dis ; 29(8): 1655-1658, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37486227

RESUMEN

Mycobacterium abscessus infections have been reported as adverse events related to medical tourism. We report M. abscessus meningitis in a patient who traveled from Colorado, USA, to Mexico to receive intrathecal stem cell injections as treatment for multiple sclerosis. We also review the management of this challenging central nervous system infection.


Asunto(s)
Turismo Médico , Meningitis , Infecciones por Mycobacterium no Tuberculosas , Mycobacterium abscessus , Humanos , Meningitis/tratamiento farmacológico , Mycobacterium abscessus/fisiología , Infecciones por Mycobacterium no Tuberculosas/diagnóstico , Infecciones por Mycobacterium no Tuberculosas/etiología , Infecciones por Mycobacterium no Tuberculosas/tratamiento farmacológico , Células Madre
6.
Ann Neurol ; 92(4): 527-531, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35932225

RESUMEN

Monkeypox virus (MPV) is an orthopox virus in the Poxviridae family that is currently of international concern. It is endemic to Central and Western Africa with two known viral clades. Various African rodents and primates are likely the natural reservoirs. Zoonotic transmission occurs by direct contact with infected animals (e.g., bites, scratches, slaughtering). Human to human transmission occurs through close contact with infected persons (e.g., respiratory droplets, skin-on-skin, or sexual contact) or fomites. Classically, human MPV disease first has a febrile prodrome with lymphadenopathy followed by a diffuse maculopapular to vesiculopustular skin/mucosal lesion eruption. In the current 2022 outbreak, which is primarily affecting men who have sex with men (MSM) currently, the febrile prodrome may be absent and skin/mucosal lesions may be isolated to the genital and anal regions. Rarely, MPV likely has the potential to be neuroinvasive based on animal models, previous case series, and preliminary reports currently under investigation. Even though neurologic manifestations of human MPV infection are rare, given the sheer numbers of increasing cases throughout the world, neurologists should be prepared to recognize, diagnose, and treat potential neuroinvasive disease or other neurologic symptoms. ANN NEUROL 2022;92:527-531.


Asunto(s)
Monkeypox virus , Minorías Sexuales y de Género , Animales , Brotes de Enfermedades , Homosexualidad Masculina , Humanos , Masculino , Piel
7.
Antimicrob Agents Chemother ; 66(8): e0022722, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35894595

RESUMEN

Enterovirus D68 (EV-D68) can cause mild to severe respiratory illness and is associated with a poliomyelitis-like paralytic syndrome called acute flaccid myelitis (AFM). Most cases of EV-D68-associated AFM occur in young children who are brought to the clinic after the onset of neurologic symptoms. There are currently no known antiviral therapies for AFM, and it is unknown whether antiviral treatments will be effective if initiated after the onset of neurologic symptoms (when patients are likely to present for medical care). We developed a "clinical treatment model" for AFM, in which individual EV-D68-infected mice are tracked and treated with an EV-D68-specific human-mouse chimeric monoclonal antibody after the onset of moderate paralysis. Mice treated with antibody had significantly better paralysis outcomes compared to nonspecific antibody-treated controls. Treatment did not reverse paralysis that was present at the time of treatment initiation but did slow the further loss of function, including progression of weakness to other limbs, as well as reducing viral titer in the muscle and spinal cords of treated animals. We observed the greatest therapeutic effect in EV-D68 isolates which were neutralized by low concentrations of antibody, and diminishing therapeutic effect in EV-D68 isolates which required higher doses of antibody for neutralization. This work supports the use of virus-specific immunotherapy for the treatment of AFM. It also suggests that patients who present with AFM should be treated as soon as possible if recent infection with EV-D68 is suspected.


Asunto(s)
Enterovirus Humano D , Infecciones por Enterovirus , Animales , Anticuerpos Neutralizantes/uso terapéutico , Antivirales , Enfermedades Virales del Sistema Nervioso Central , Niño , Preescolar , Modelos Animales de Enfermedad , Enterovirus Humano D/fisiología , Infecciones por Enterovirus/tratamiento farmacológico , Humanos , Ratones , Mielitis , Enfermedades Neuromusculares , Parálisis/complicaciones , Parálisis/tratamiento farmacológico
8.
J Virol ; 95(18): e0083521, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34190599

RESUMEN

Recruitment of immune cells from the periphery is critical for controlling West Nile virus (WNV) growth in the central nervous system (CNS) and preventing subsequent WNV-induced CNS disease. Neuroinflammatory responses, including the release of proinflammatory cytokines and chemokines by CNS cells, influence the entry and function of peripheral immune cells that infiltrate the CNS. However, these same cytokines and chemokines contribute to tissue damage in other models of CNS injury. Rosiglitazone is a peroxisome proliferator-activated receptor gamma (PPARγ) agonist that inhibits neuroinflammation. We used rosiglitazone in WNV-infected ex vivo brain slice cultures (BSC) to investigate the role of neuroinflammation within the CNS in the absence of peripheral immune cells. Rosiglitazone treatment inhibited WNV-induced expression of proinflammatory chemokines and cytokines, interferon beta (IFN-ß), and IFN-stimulated genes (ISG) and also decreased WNV-induced activation of microglia. These decreased neuroinflammatory responses were associated with activation of astrocytes, robust viral growth, increased activation of caspase 3, and increased neuronal loss. Rosiglitazone had a similar effect on in vivo WNV infection, causing increased viral growth, tissue damage, and disease severity in infected mice, even though the number of infiltrating peripheral immune cells was higher in rosiglitazone-treated, WNV-infected mice than in untreated, infected controls. These results indicate that local neuroinflammatory responses are capable of controlling viral growth within the CNS and limiting neuronal loss and may function to keep the virus in check prior to the infiltration of peripheral immune cells, limiting both virus- and immune-mediated neuronal damage. IMPORTANCE West Nile virus is the most common cause of epidemic encephalitis in the United States and can result in debilitating CNS disease. There are no effective vaccines or treatments for WNV-induced CNS disease in humans. The peripheral immune response is critical for protection against WNV CNS infections. We now demonstrate that intrinsic immune responses also control viral growth and limit neuronal loss. These findings have important implications for developing new therapies for WNV-induced CNS disease.


Asunto(s)
Encéfalo/inmunología , Muerte Celular , Enfermedades del Sistema Nervioso Central/prevención & control , Inmunidad Innata/inmunología , Neuronas/inmunología , Fiebre del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Animales , Encéfalo/patología , Encéfalo/virología , Enfermedades del Sistema Nervioso Central/etiología , Enfermedades del Sistema Nervioso Central/patología , Quimiocinas/metabolismo , Citocinas/metabolismo , Femenino , Masculino , Ratones , Neuronas/patología , Neuronas/virología , Fiebre del Nilo Occidental/complicaciones , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/virología
9.
J Pediatr ; 244: 161-168.e1, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35150729

RESUMEN

OBJECTIVE: To investigate the optimal implementation and clinical and financial impacts of the FilmArray Meningitis Encephalitis Panel (MEP) multiplex polymerase chain reaction testing of cerebrospinal fluid (CSF) in children with suspected central nervous system infection. STUDY DESIGN: A pre-post quasiexperimental cohort study to investigate the impact of implementing MEP using a rapid CSF diagnostic stewardship program was conducted at Children's Hospital Colorado (CHCO). MEP was implemented with electronic medical record indication selection to guide testing to children meeting approved use criteria: infants <2 months, immunocompromised, encephalitis, and ≥5 white blood cells/µL of CSF. Positive results were communicated with antimicrobial stewardship real-time decision support. All cases with CSF obtained by lumbar puncture sent to the CHCO microbiology laboratory meeting any of the 4 aforementioned criteria were included with preimplementation controls (2015-2016) compared with postimplementation cases (2017-2018). Primary outcome was time-to-optimal antimicrobials compared using log-rank test with Kaplan-Meier analysis. RESULTS: Time-to-optimal antimicrobials decreased from 28 hours among 1124 preimplementation controls to 18 hours (P < .0001) among 1127 postimplementation cases (72% with MEP testing conducted). Postimplementation, time-to-positive CSF results was faster (4.8 vs 9.6 hours, P < .0001), intravenous antimicrobial duration was shorter (24 vs 36 hours, P = .004), with infectious neurologic diagnoses more frequently identified (15% vs 10%, P = .03). There were no differences in time-to-effective antimicrobials, hospital admissions, antimicrobial starts, or length of stay. Costs of microbiologic testing increased, but total hospital costs were unchanged. CONCLUSIONS: Implementation of MEP with a rapid central nervous system diagnostic stewardship program improved antimicrobial use with faster results shortening empiric therapy. Routine MEP testing for high-yield indications enables antimicrobial optimization with unchanged overall costs.


Asunto(s)
Antiinfecciosos , Infecciones del Sistema Nervioso Central , Encefalitis , Meningitis , Malformaciones del Sistema Nervioso , Antibacterianos , Antiinfecciosos/uso terapéutico , Infecciones del Sistema Nervioso Central/líquido cefalorraquídeo , Infecciones del Sistema Nervioso Central/diagnóstico , Infecciones del Sistema Nervioso Central/tratamiento farmacológico , Niño , Estudios de Cohortes , Encefalitis/diagnóstico , Humanos , Lactante , Meningitis/diagnóstico , Estudios Retrospectivos
10.
MMWR Morb Mortal Wkly Rep ; 71(38): 1212-1215, 2022 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-36136957

RESUMEN

Monkeypox virus (MPXV) is an orthopoxvirus in the Poxviridae family. The current multinational monkeypox outbreak has now spread to 96 countries that have not historically reported monkeypox, with most cases occurring among gay, bisexual, and other men who have sex with men (1,2). The first monkeypox case in the United States associated with this outbreak was identified in May 2022 in Massachusetts (1); monkeypox has now been reported in all 50 states, the District of Columbia (DC), and one U.S. territory. MPXV is transmitted by close contact with infected persons or animals; infection results in a febrile illness followed by a diffuse vesiculopustular rash and lymphadenopathy. However, illness in the MPXV current Clade II outbreak has differed: the febrile prodrome is frequently absent or mild, and the rash often involves genital, anal, or oral regions (3,4). Although neuroinvasive disease has been previously reported with MPXV infection (5,6), it appears to be rare. This report describes two cases of encephalomyelitis in patients with monkeypox disease that occurred during the current U.S. outbreak. Although neurologic complications of acute MPXV infections are rare, suspected cases should be reported to state, tribal, local, or territorial health departments to improve understanding of the range of clinical manifestations of and treatment options for MPXV infections during the current outbreak.


Asunto(s)
Encefalomielitis , Exantema , Mpox , Minorías Sexuales y de Género , Colorado/epidemiología , District of Columbia , Homosexualidad Masculina , Humanos , Masculino , Mpox/epidemiología , Monkeypox virus , Estados Unidos
11.
Nature ; 534(7608): 538-43, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27337340

RESUMEN

Over 50% of patients who survive neuroinvasive infection with West Nile virus (WNV) exhibit chronic cognitive sequelae. Although thousands of cases of WNV-mediated memory dysfunction accrue annually, the mechanisms responsible for these impairments are unknown. The classical complement cascade, a key component of innate immune pathogen defence, mediates synaptic pruning by microglia during early postnatal development. Here we show that viral infection of adult hippocampal neurons induces complement-mediated elimination of presynaptic terminals in a murine WNV neuroinvasive disease model. Inoculation of WNV-NS5-E218A, a WNV with a mutant NS5(E218A) protein leads to survival rates and cognitive dysfunction that mirror human WNV neuroinvasive disease. WNV-NS5-E218A-recovered mice (recovery defined as survival after acute infection) display impaired spatial learning and persistence of phagocytic microglia without loss of hippocampal neurons or volume. Hippocampi from WNV-NS5-E218A-recovered mice with poor spatial learning show increased expression of genes that drive synaptic remodelling by microglia via complement. C1QA was upregulated and localized to microglia, infected neurons and presynaptic terminals during WNV neuroinvasive disease. Murine and human WNV neuroinvasive disease post-mortem samples exhibit loss of hippocampal CA3 presynaptic terminals, and murine studies revealed microglial engulfment of presynaptic terminals during acute infection and after recovery. Mice with fewer microglia (Il34(-/-) mice with a deficiency in IL-34 production) or deficiency in complement C3 or C3a receptor were protected from WNV-induced synaptic terminal loss. Our study provides a new murine model of WNV-induced spatial memory impairment, and identifies a potential mechanism underlying neurocognitive impairment in patients recovering from WNV neuroinvasive disease.


Asunto(s)
Proteínas del Sistema Complemento/inmunología , Trastornos de la Memoria/patología , Trastornos de la Memoria/virología , Microglía/inmunología , Plasticidad Neuronal , Terminales Presinápticos/patología , Virus del Nilo Occidental/patogenicidad , Animales , Región CA3 Hipocampal/inmunología , Región CA3 Hipocampal/patología , Región CA3 Hipocampal/virología , Activación de Complemento , Vía Clásica del Complemento/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Trastornos de la Memoria/inmunología , Trastornos de la Memoria/fisiopatología , Ratones , Neuronas/inmunología , Neuronas/patología , Neuronas/virología , Terminales Presinápticos/inmunología , Memoria Espacial , Fiebre del Nilo Occidental/patología , Fiebre del Nilo Occidental/fisiopatología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/inmunología
12.
Ann Neurol ; 88(1): 1-11, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32506549

RESUMEN

In less than 6 months, the severe acute respiratory syndrome-coronavirus type 2 (SARS-CoV-2) has spread worldwide infecting nearly 6 million people and killing over 350,000. Initially thought to be restricted to the respiratory system, we now understand that coronavirus disease 2019 (COVID-19) also involves multiple other organs, including the central and peripheral nervous system. The number of recognized neurologic manifestations of SARS-CoV-2 infection is rapidly accumulating. These may result from a variety of mechanisms, including virus-induced hyperinflammatory and hypercoagulable states, direct virus infection of the central nervous system (CNS), and postinfectious immune mediated processes. Example of COVID-19 CNS disease include encephalopathy, encephalitis, acute disseminated encephalomyelitis, meningitis, ischemic and hemorrhagic stroke, venous sinus thrombosis, and endothelialitis. In the peripheral nervous system, COVID-19 is associated with dysfunction of smell and taste, muscle injury, the Guillain-Barre syndrome, and its variants. Due to its worldwide distribution and multifactorial pathogenic mechanisms, COVID-19 poses a global threat to the entire nervous system. Although our understanding of SARS-CoV-2 neuropathogenesis is still incomplete and our knowledge is evolving rapidly, we hope that this review will provide a useful framework and help neurologists in understanding the many neurologic facets of COVID-19. ANN NEUROL 2020;88:1-11 ANN NEUROL 2020;88:1-11.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/fisiopatología , Enfermedades del Sistema Nervioso/fisiopatología , Neumonía Viral/fisiopatología , Encefalopatías/etiología , Encefalopatías/fisiopatología , Isquemia Encefálica/etiología , Isquemia Encefálica/fisiopatología , COVID-19 , Infecciones por Coronavirus/complicaciones , Coagulación Intravascular Diseminada/etiología , Coagulación Intravascular Diseminada/fisiopatología , Encefalitis/etiología , Encefalitis/fisiopatología , Encefalomielitis Aguda Diseminada/etiología , Encefalomielitis Aguda Diseminada/fisiopatología , Síndrome de Guillain-Barré/etiología , Síndrome de Guillain-Barré/fisiopatología , Humanos , Inflamación , Hemorragias Intracraneales/etiología , Hemorragias Intracraneales/fisiopatología , Leucoencefalitis Hemorrágica Aguda/etiología , Leucoencefalitis Hemorrágica Aguda/fisiopatología , Meningitis Viral/etiología , Meningitis Viral/fisiopatología , Enfermedades del Sistema Nervioso/etiología , Trastornos del Olfato/etiología , Trastornos del Olfato/fisiopatología , Pandemias , Neumonía Viral/complicaciones , SARS-CoV-2 , Trombosis de los Senos Intracraneales/etiología , Trombosis de los Senos Intracraneales/fisiopatología , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología , Trombofilia/etiología , Trombofilia/fisiopatología
13.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31167912

RESUMEN

Enterovirus D68 (EV-D68) is an emerging virus that has been identified as a cause of recent outbreaks of acute flaccid myelitis (AFM), a poliomyelitis-like spinal cord syndrome that can result in permanent paralysis and disability. In experimental mouse models, EV-D68 spreads to, infects, and kills spinal motor neurons following infection by various routes of inoculation. The topography of virus-induced motor neuron loss correlates with the pattern of paralysis. The mechanism(s) by which EV-D68 spreads to target motor neurons remains unclear. We sought to determine the capacity of EV-D68 to spread by the neuronal route and to determine the role of known EV-D68 receptors, sialic acid and intracellular adhesion molecule 5 (ICAM-5), in neuronal infection. To do this, we utilized a microfluidic chamber culture system in which human induced pluripotent stem cell (iPSC) motor neuron cell bodies and axons can be compartmentalized for independent experimental manipulation. We found that EV-D68 can infect motor neurons via their distal axons and spread by retrograde axonal transport to the neuronal cell bodies. Virus was not released from the axons via anterograde axonal transport after infection of the cell bodies. Prototypic strains of EV-D68 depended on sialic acid for axonal infection and transport, while contemporary circulating strains isolated during the 2014 EV-D68 outbreak did not. The pattern of infection did not correspond with the ICAM-5 distribution and expression in either human tissue, the mouse model, or the iPSC motor neurons.IMPORTANCE Enterovirus D68 (EV-D68) infections are on the rise worldwide. Since 2014, the United States has experienced biennial spikes in EV-D68-associated acute flaccid myelitis (AFM) that have left hundreds of children paralyzed. Much remains to be learned about the pathogenesis of EV-D68 in the central nervous system (CNS). Herein we investigated the mechanisms of EV-D68 CNS invasion through neuronal pathways. A better understanding of EV-D68 infection in experimental models may allow for better prevention and treatment strategies of EV-D68 CNS disease.


Asunto(s)
Transporte Axonal , Enterovirus Humano D/fisiología , Infecciones por Enterovirus/metabolismo , Infecciones por Enterovirus/virología , Interacciones Huésped-Patógeno , Neuronas Motoras/metabolismo , Neuronas Motoras/virología , Ácido N-Acetilneuramínico/metabolismo , Animales , Moléculas de Adhesión Celular/metabolismo , Enfermedades Virales del Sistema Nervioso Central/metabolismo , Enfermedades Virales del Sistema Nervioso Central/virología , Modelos Animales de Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Neuronas Motoras/citología , Mielitis/metabolismo , Mielitis/virología , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neuromusculares/metabolismo , Enfermedades Neuromusculares/virología , Parálisis/etiología
14.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30814290

RESUMEN

Reovirus encephalitis in mice was used as a model system to investigate astrocyte activation (astrogliosis) following viral infection of the brain. Reovirus infection resulted in astrogliosis, as evidenced by increased expression of glial fibrillary acidic protein (GFAP), and the upregulation of genes that have been previously associated with astrocyte activation. Astrocyte activation occurred in regions of the brain that are targeted by reovirus but extended beyond areas of active infection. Astrogliosis also occurred following reovirus infection of ex vivo brain slice cultures (BSCs), demonstrating that factors intrinsic to the brain are sufficient to activate astrocytes and that this process can occur in the absence of any contribution from the peripheral immune response. In agreement with previous reports, reovirus antigen did not colocalize with GFAP in infected brains, suggesting that reovirus does not infect astrocytes. Reovirus-infected neurons produce interferon beta (IFN-ß). IFN-ß treatment of primary astrocytes resulted in both the upregulation of GFAP and cytokines that are associated with astrocyte activation. In addition, the ability of media from reovirus-infected BSCs to activate primary astrocytes was blocked by anti-IFN-ß antibodies. These results suggest that IFN-ß, likely released from reovirus-infected neurons, results in the activation of astrocytes during reovirus encephalitis. In areas where infection and injury were pronounced, an absence of GFAP staining was consistent with activation-induced cell death as a mechanism of inflammation control. In support of this, activated Bak and cleaved caspase 3 were detected in astrocytes within reovirus-infected brains, indicating that activated astrocytes undergo apoptosis.IMPORTANCE Viral encephalitis is a significant cause of worldwide morbidity and mortality, and specific treatments are extremely limited. Virus infection of the brain triggers neuroinflammation; however, the role of neuroinflammation in the pathogenesis of viral encephalitis is unclear. Initial neuroinflammatory responses likely contribute to viral clearance, but prolonged exposure to proinflammatory cytokines released during neuroinflammation may be deleterious and contribute to neuronal death and tissue injury. Activation of astrocytes is a hallmark of neuroinflammation. Here, we show that reovirus infection of the brain results in the activation of astrocytes via an IFN-ß-mediated process and that these astrocytes later die by Bak-mediated apoptosis. A better understanding of neuroinflammatory responses during viral encephalitis may facilitate the development of new treatment strategies for these diseases.


Asunto(s)
Astrocitos/inmunología , Interferón beta/metabolismo , Infecciones por Reoviridae/inmunología , Animales , Apoptosis , Astrocitos/metabolismo , Astrocitos/virología , Encéfalo/inmunología , Encéfalo/virología , Muerte Celular , Modelos Animales de Enfermedad , Encefalitis Viral/virología , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Gliosis , Inflamación/metabolismo , Interferón beta/inmunología , Ratones , Neurogénesis , Neuronas/virología , Reoviridae/metabolismo , Infecciones por Reoviridae/metabolismo , Transducción de Señal/inmunología
15.
J Neurovirol ; 26(2): 149-154, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31858483

RESUMEN

Usutu virus is an emerging mosquito-borne flavivirus initially identified in South Africa in 1959 that is now circulating throughout parts of Africa, Europe, and the Middle East. It is closely related to West Nile virus, and has similar vectors, amplifying bird hosts, and epidemiology. Usutu virus infection can occur in humans and may be asymptomatic or cause systemic (e.g., fever, rash, and hepatitis) or neuroinvasive (e.g., meningitis and encephalitis) disease. Given few reported cases, the full clinical spectrum is not known. No anti-viral treatment is available, but it can be largely prevented by avoiding mosquito bites. Because of similar mosquitoes, birds, and climate to Europe, the potential for introduction to North America is possible.


Asunto(s)
Infecciones por Flavivirus/epidemiología , Infecciones por Flavivirus/virología , Flavivirus , Animales , Infecciones por Flavivirus/transmisión , Humanos , América del Norte
16.
J Virol ; 92(16)2018 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-29899084

RESUMEN

Flaviviruses account for most arthropod-borne cases of human encephalitis in the world. However, the exact mechanisms of injury to the central nervous system (CNS) during flavivirus infections remain poorly understood. Microglia are the resident immune cells of the CNS and are important for multiple functions, including control of viral pathogenesis. Utilizing a pharmacologic method of microglia depletion (PLX5622 [Plexxikon Inc.], an inhibitor of colony-stimulating factor 1 receptor), we sought to determine the role of microglia in flaviviral pathogenesis. Depletion of microglia resulted in increased mortality and viral titer in the brain following infection with either West Nile virus (WNV) or Japanese encephalitis virus (JEV). Interestingly, microglial depletion did not prevent virus-induced increases in the expression of relevant cytokines and chemokines at the mRNA level. In fact, the expression of several proinflammatory genes was increased in virus-infected, microglia-depleted mice compared to virus-infected, untreated controls. In contrast, and as expected, expression of the macrophage marker triggering receptor expressed on myeloid cells 2 (TREM2) was decreased in virus-infected, PLX5622-treated mice compared to virus-infected controls.IMPORTANCE As CNS invasion by flaviviruses is a rare but life-threatening event, it is critical to understand how brain-resident immune cells elicit protection or injury during disease progression. Microglia have been shown to be important in viral clearance but may also contribute to CNS injury as part of the neuroinflammatory process. By utilizing a microglial depletion model, we can begin to parse out the exact roles of microglia during flaviviral pathogenesis with hopes of understanding specific mechanisms as potential targets for therapeutics.


Asunto(s)
Encéfalo/patología , Encéfalo/virología , Encefalitis Japonesa/patología , Microglía/inmunología , Carga Viral , Fiebre del Nilo Occidental/patología , Animales , Encéfalo/inmunología , Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Encefalitis Japonesa/virología , Ratones , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Análisis de Supervivencia , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/virología , Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/aislamiento & purificación
17.
PLoS Pathog ; 13(2): e1006199, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28231269

RESUMEN

In 2014, the United States experienced an epidemic of acute flaccid myelitis (AFM) cases in children coincident with a nationwide outbreak of enterovirus D68 (EV-D68) respiratory disease. Up to half of the 2014 AFM patients had EV-D68 RNA detected by RT-PCR in their respiratory secretions, although EV-D68 was only detected in cerebrospinal fluid (CSF) from one 2014 AFM patient. Given previously described molecular and epidemiologic associations between EV-D68 and AFM, we sought to develop an animal model by screening seven EV-D68 strains for the ability to induce neurological disease in neonatal mice. We found that four EV-D68 strains from the 2014 outbreak (out of five tested) produced a paralytic disease in mice resembling human AFM. The remaining 2014 strain, as well as 1962 prototype EV-D68 strains Fermon and Rhyne, did not produce, or rarely produced, paralysis in mice. In-depth examination of the paralysis caused by a representative 2014 strain, MO/14-18947, revealed infectious virus, virion particles, and viral genome in the spinal cords of paralyzed mice. Paralysis was elicited in mice following intramuscular, intracerebral, intraperitoneal, and intranasal infection, in descending frequency, and was associated with infection and loss of motor neurons in the anterior horns of spinal cord segments corresponding to paralyzed limbs. Virus isolated from spinal cords of infected mice transmitted disease when injected into naïve mice, fulfilling Koch's postulates in this model. Finally, we found that EV-D68 immune sera, but not normal mouse sera, protected mice from development of paralysis and death when administered prior to viral challenge. These studies establish an experimental model to study EV-D68-induced myelitis and to better understand disease pathogenesis and develop potential therapies.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Enterovirus/patología , Mielitis/virología , Animales , Enterovirus Humano D , Infecciones por Enterovirus/complicaciones , Femenino , Inmunohistoquímica , Masculino , Ratones , Microscopía Electrónica de Transmisión , Mielitis/patología , Parálisis/virología , Reacción en Cadena de la Polimerasa , Médula Espinal/patología , Médula Espinal/virología
18.
Ann Neurol ; 93(1): 1-12, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36573470
19.
Ann Neurol ; 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38065680
20.
Semin Neurol ; 39(4): 419-427, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31533182

RESUMEN

There are many arthropod-borne viruses (arboviruses) capable of neuroinvasion, with West Nile virus being one of the most well known. In this review, we highlight five rarer emerging or reemerging arboviruses capable of neuroinvasion: Cache Valley, eastern equine encephalitis, Jamestown Canyon, Powassan, and Usutu viruses. Cache Valley and Jamestown Canyon viruses likely circulate throughout most of North America, while eastern equine encephalitis and Powassan viruses typically circulate in the eastern half. Usutu virus is not currently circulating in North America, but has the potential to be introduced in the future given similar climate, vectors, and host species to Europe (where it has been circulating). Health care providers should contact their state or local health departments with any questions regarding arboviral disease surveillance, diagnosis, treatment, or prevention. To prevent neuroinvasive arboviral diseases, use of insect repellent and other mosquito and tick bite prevention strategies are key.


Asunto(s)
Infecciones por Arbovirus/epidemiología , Infecciones por Bunyaviridae/epidemiología , Encefalitis de California/epidemiología , Encefalitis Transmitida por Garrapatas/epidemiología , Encefalomielitis Equina Oriental/epidemiología , Infecciones por Flavivirus/epidemiología , Animales , Infecciones por Arbovirus/diagnóstico , Infecciones por Arbovirus/terapia , Virus Bunyamwera/aislamiento & purificación , Infecciones por Bunyaviridae/diagnóstico , Infecciones por Bunyaviridae/terapia , Virus de la Encefalitis de California/aislamiento & purificación , Encefalitis de California/diagnóstico , Encefalitis de California/terapia , Encefalitis Transmitida por Garrapatas/diagnóstico , Encefalitis Transmitida por Garrapatas/terapia , Encefalomielitis Equina Oriental/diagnóstico , Encefalomielitis Equina Oriental/terapia , Flavivirus/aislamiento & purificación , Infecciones por Flavivirus/diagnóstico , Infecciones por Flavivirus/terapia , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA