Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Más filtros










Intervalo de año de publicación
1.
J Neurovirol ; 30(1): 39-51, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38172412

RESUMEN

Sarbecoviruses such as SARS and SARS-CoV-2 have been responsible for two major outbreaks in humans, the latter resulting in a global pandemic. While sarbecoviruses primarily cause an acute respiratory infection, they have been shown to infect the nervous system. However, mechanisms of sarbecovirus neuroinvasion and neuropathogenesis remain unclear. In this study, we examined the infectivity and trans-synaptic transmission potential of the sarbecoviruses SARS and SARS-CoV-2 in human stem cell-derived neural model systems. We demonstrated limited ability of sarbecoviruses to infect and replicate in human stem cell-derived neurons. Furthermore, we demonstrated an inability of sarbecoviruses to transmit between synaptically connected human stem cell-derived neurons. Finally, we determined an absence of SARS-CoV-2 infection in olfactory neurons in experimentally infected ferrets. Collectively, this study indicates that sarbecoviruses exhibit low potential to infect human stem cell-derived neurons, lack an ability to infect ferret olfactory neurons, and lack an inbuilt molecular mechanism to utilise retrograde axonal trafficking and trans-synaptic transmission to spread within the human nervous system.


Asunto(s)
Axones , COVID-19 , Hurones , SARS-CoV-2 , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , Humanos , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Animales , COVID-19/virología , COVID-19/transmisión , Axones/virología , Hurones/virología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Neuronas/virología , Replicación Viral , Chlorocebus aethiops , Células-Madre Neurales/virología , Células Vero
2.
Nat Commun ; 14(1): 4485, 2023 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-37495586

RESUMEN

Anosmia was identified as a hallmark of COVID-19 early in the pandemic, however, with the emergence of variants of concern, the clinical profile induced by SARS-CoV-2 infection has changed, with anosmia being less frequent. Here, we assessed the clinical, olfactory and neuroinflammatory conditions of golden hamsters infected with the original Wuhan SARS-CoV-2 strain, its isogenic ORF7-deletion mutant and three variants: Gamma, Delta, and Omicron/BA.1. We show that infected animals develop a variant-dependent clinical disease including anosmia, and that the ORF7 of SARS-CoV-2 contributes to the induction of olfactory dysfunction. Conversely, all SARS-CoV-2 variants are neuroinvasive, regardless of the clinical presentation they induce. Taken together, this confirms that neuroinvasion and anosmia are independent phenomena upon SARS-CoV-2 infection. Using newly generated nanoluciferase-expressing SARS-CoV-2, we validate the olfactory pathway as a major entry point into the brain in vivo and demonstrate in vitro that SARS-CoV-2 travels retrogradely and anterogradely along axons in microfluidic neuron-epithelial networks.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Cricetinae , COVID-19/virología , SARS-CoV-2/genética , Genoma Viral , Axones/virología , Bulbo Olfatorio/virología , Internalización del Virus , Carga Viral , Variación Genética
3.
J Virol ; 96(6): e0217821, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35045266

RESUMEN

The assembly and egress of alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), within neurons is poorly understood. A key unresolved question is the structure of the viral particle that moves by anterograde transport along the axon, and two alternative mechanisms have been described. In the "married" model, capsids acquire their envelopes in the cell body and then traffic along axons as enveloped virions within a bounding organelle. In the "separate" model, nonenveloped capsids travel from the cell body into and along the axon, eventually encountering their envelopment organelles at a distal site, such as the nerve cell terminal. Here, we describe an "envelopment trap" to test these models using the dominant negative terminal endosomal sorting complex required for transport (ESCRT) component VPS4-EQ. Green fluorescent protein (GFP)-tagged VPS4-EQ was used to arrest HSV-1 or PRV capsid envelopment, inhibit downstream trafficking, and GFP-label envelopment intermediates. We found that GFP-VPS4-EQ inhibited trafficking of HSV-1 capsids into and along the neurites and axons of mouse CAD cells and rat embryonic primary cortical neurons, consistent with egress via the married pathway. In contrast, transport of HSV-1 capsids was unaffected in the neurites of human SK-N-SH neuroblastoma cells, consistent with the separate mechanism. Unexpectedly, PRV (generally thought to utilize the married pathway) also appeared to employ the separate mechanism in SK-N-SH cells. We propose that apparent differences in the methods of HSV-1 and PRV egress are more likely a reflection of the host neuron in which transport is studied rather than true biological differences between the viruses themselves. IMPORTANCE Alphaherpesviruses, including herpes simplex virus 1 (HSV-1) and pseudorabies virus (PRV), are pathogens of the nervous system. They replicate in the nerve cell body and then travel great distances along axons to reach nerve termini and spread to adjacent epithelial cells; however, key aspects of how these viruses travel along axons remain controversial. Here, we test two alternative mechanisms for transport, the married and separate models, by blocking envelope assembly, a critical step in viral egress. When we arrest formation of the viral envelope using a mutated component of the cellular ESCRT apparatus, we find that entry of viral particles into axons is blocked in some types of neurons but not others. This approach allows us to determine whether envelope assembly occurs prior to entry of viruses into axons or afterwards and, thus, to distinguish between the alternative models for viral transport.


Asunto(s)
Alphaherpesvirinae , Complejos de Clasificación Endosomal Requeridos para el Transporte , Herpesvirus Humano 1 , Herpesvirus Suido 1 , Neuronas , Alphaherpesvirinae/metabolismo , Animales , Axones/virología , Línea Celular Tumoral , Células Cultivadas , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Suido 1/fisiología , Humanos , Ratones , Neuronas/virología , Ratas , Ensamble de Virus/fisiología , Internalización del Virus
4.
Viruses ; 13(9)2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34578430

RESUMEN

Herpes simplex virus type-1 (HSV-1) and type-2 (HSV-2) are prototypical alphaherpesviruses that are characterized by their unique properties to infect trigeminal and dorsal root ganglionic neurons, respectively, and establish life-long latent infections. These viruses initially infect mucosal epithelial tissues and subsequently spread to neurons. They are associated with a significant disease spectrum, including orofacial and ocular infections for HSV-1 and genital and neonatal infections for HSV-2. Viral glycoproteins within the virion envelope bind to specific cellular receptors to mediate virus entry into cells. This is achieved by the fusion of the viral envelope with the plasma membrane. Similarly, viral glycoproteins expressed on cell surfaces mediate cell-to-cell fusion and facilitate virus spread. An interactive complex of viral glycoproteins gB, gD/gH/gL, and gK and other proteins mediate these membrane fusion phenomena with glycoprotein B (gB), the principal membrane fusogen. The requirement for the virion to enter neuronal axons suggests that the heterodimeric protein complex of gK and membrane protein UL20, found only in alphaherpesviruses, constitute a critical determinant for neuronal entry. This hypothesis was substantiated by the observation that a small deletion in the amino terminus of gK prevents entry into neuronal axons while allowing entry into other cells via endocytosis. Cellular receptors and receptor-mediated signaling synergize with the viral membrane fusion machinery to facilitate virus entry and intercellular spread. Unraveling the underlying interactions among viral glycoproteins, envelope proteins, and cellular receptors will provide new innovative approaches for antiviral therapy against herpesviruses and other neurotropic viruses.


Asunto(s)
Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Fusión de Membrana , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus , Axones/virología , Fusión Celular , Humanos , Neuronas/virología , Proteínas del Envoltorio Viral/química , Tropismo Viral
5.
Immunol Res ; 69(6): 553-557, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34363587

RESUMEN

The persistence of neurological symptoms after SARS-CoV-2 infection, as well as the presence of late axonal damage, is still unknown. We performed extensive systemic and neurological follow-up evaluations in 107 out of 193 consecutive patients admitted to the COVID-19 medical unit, University Hospital of Verona, Italy between March and June 2020. We analysed serum neurofilament light chain (NfL) levels in all cases including a subgroup (n = 29) of patients with available onset samples. Comparisons between clinical and biomarker data were then performed. Neurological symptoms were still present in a significant number (n = 49) of patients over the follow-up. The most common reported symptoms were hyposmia (n = 11), fatigue (n = 28), myalgia (n = 14), and impaired memory (n = 11) and were more common in cases with severe acute COVID-19. Follow-up serum NfL values (15.2 pg/mL, range 2.4-62.4) were within normal range in all except 5 patients and did not differentiate patients with vs without persistent neurological symptoms. In patients with available onset and follow-up samples, a significant (p < 0.001) decrease of NfL levels was observed and was more evident in patients with a severe acute disease. Despite the common persistence of neurological symptoms, COVID-19 survivors do not show active axonal damage, which seems a peculiar feature of acute SARS-CoV-2 infection.


Asunto(s)
Axones/patología , COVID-19/patología , Enfermedades del Sistema Nervioso/patología , Adulto , Anciano , Anciano de 80 o más Años , Ageusia/patología , Ageusia/virología , Anosmia/patología , Anosmia/virología , Axones/virología , Progresión de la Enfermedad , Fatiga/patología , Fatiga/virología , Femenino , Humanos , Italia , Masculino , Trastornos de la Memoria/patología , Trastornos de la Memoria/virología , Persona de Mediana Edad , Mialgia/patología , Mialgia/virología , Enfermedades del Sistema Nervioso/virología , Proteínas de Neurofilamentos/sangre , SARS-CoV-2
6.
Neuropharmacology ; 198: 108766, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34454912

RESUMEN

The coronavirus disease 2019 (Covid-19) pandemic intensified the already catastrophic drug overdose and substance use disorder (SUD) epidemic, signaling a syndemic as social isolation, economic and mental health distress, and disrupted treatment services disproportionally impacted this vulnerable population. Along with these social and societal factors, biological factors triggered by intense stress intertwined with incumbent overactivity of the immune system and the resulting inflammatory outcomes may impact the functional status of the central nervous system (CNS). We review the literature concerning SARS-CoV2 infiltration and infection in the CNS and the prospects of synergy between stress, inflammation, and kynurenine pathway function during illness and recovery from Covid-19. Taken together, inflammation and neuroimmune signaling, a consequence of Covid-19 infection, may dysregulate critical pathways and underlie maladaptive changes in the CNS, to exacerbate the development of neuropsychiatric symptoms and in the vulnerability to develop SUD. This article is part of the special Issue on 'Vulnerabilities to Substance Abuse'.


Asunto(s)
COVID-19/epidemiología , Abuso de Medicamentos/estadística & datos numéricos , SARS-CoV-2 , Trastornos Relacionados con Sustancias/epidemiología , Adaptación Psicológica , Enzima Convertidora de Angiotensina 2/fisiología , Animales , Axones/virología , COVID-19/inmunología , COVID-19/fisiopatología , COVID-19/psicología , Comorbilidad , Susceptibilidad a Enfermedades , Células Endoteliales/virología , Humanos , Inmunidad Innata , Inflamación/etiología , Quinurenina/metabolismo , Neuronas/virología , Neurotransmisores/metabolismo , Mucosa Olfatoria/virología , Pandemias , SARS-CoV-2/fisiología , Aislamiento Social , Estrés Psicológico , Trastornos Relacionados con Sustancias/etiología , Trastornos Relacionados con Sustancias/fisiopatología , Triptófano/metabolismo , Tropismo Viral
7.
Virology ; 559: 65-73, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33836340

RESUMEN

α-herpesviruses have been very successful, principally because they establish lifelong latency in sensory ganglia. An essential piece of the lifecycle of α-herpesviruses involves the capacity to travel from sensory neurons to epithelial tissues following virus reactivation from latency, a process known as anterograde transport. Virus particles formed in neuron cell bodies hitchhike on kinesin motors that run along microtubules, the length of axons. Herpes simplex virus (HSV) and pseudorabies virus (PRV) have been intensely studied to elucidate anterograde axonal transport. Both viruses use similar strategies for anterograde transport, although there are significant differences in the form of virus particles transported in axons, the identity of the kinesins that transport viruses, and how certain viral membrane proteins, gE/gI and US9, participate in this process. This review compares the older models for HSV and PRV anterograde transport with recent results, which are casting a new light on several aspects of this process.


Asunto(s)
Transporte Axonal , Axones/virología , Herpesviridae/fisiología , Neuronas/virología , Proteínas de la Matriz Viral/metabolismo , Epitelio/virología , Herpesviridae/genética , Proteínas de la Matriz Viral/genética , Virión
8.
Viruses ; 13(1)2021 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-33440758

RESUMEN

Understanding how Zika virus (Flaviviridae; ZIKV) affects neural cells is paramount in comprehending pathologies associated with infection. Whilst the effects of ZIKV in neural development are well documented, impact on the adult nervous system remains obscure. Here, we investigated the effects of ZIKV infection in established mature myelinated central nervous system (CNS) cultures. Infection incurred damage to myelinated fibers, with ZIKV-positive cells appearing when myelin damage was first detected as well as axonal pathology, suggesting the latter was a consequence of oligodendroglia infection. Transcriptome analysis revealed host factors that were upregulated during ZIKV infection. One such factor, CCL5, was validated in vitro as inhibiting myelination. Transferred UV-inactivated media from infected cultures did not damage myelin and axons, suggesting that viral replication is necessary to induce the observed effects. These data show that ZIKV infection affects CNS cells even after myelination-which is critical for saltatory conduction and neuronal function-has taken place. Understanding the targets of this virus across developmental stages including the mature CNS, and the subsequent effects of infection of cell types, is necessary to understand effective time frames for therapeutic intervention.


Asunto(s)
Axones/virología , Enfermedades Desmielinizantes/etiología , Infección por el Virus Zika/complicaciones , Infección por el Virus Zika/virología , Virus Zika/fisiología , Animales , Biomarcadores , Traumatismos del Nervio Craneal/etiología , Traumatismos del Nervio Craneal/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones , Ratas , Transcriptoma
9.
Acta Neuropathol Commun ; 8(1): 199, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33228789

RESUMEN

The highly neurotropic rabies virus (RABV) enters peripheral neurons at axon termini and requires long distance axonal transport and trans-synaptic spread between neurons for the infection of the central nervous system (CNS). Recent 3D imaging of field RABV-infected brains revealed a remarkably high proportion of infected astroglia, indicating that highly virulent field viruses are able to suppress astrocyte-mediated innate immune responses and virus elimination pathways. While fundamental for CNS invasion, in vivo field RABV spread and tropism in peripheral tissues is understudied. Here, we used three-dimensional light sheet and confocal laser scanning microscopy to investigate the in vivo distribution patterns of a field RABV clone in cleared high-volume tissue samples after infection via a natural (intramuscular; hind leg) and an artificial (intracranial) inoculation route. Immunostaining of virus and host markers provided a comprehensive overview of RABV infection in the CNS and peripheral nerves after centripetal and centrifugal virus spread. Importantly, we identified non-neuronal, axon-ensheathing neuroglia (Schwann cells, SCs) in peripheral nerves of the hind leg and facial regions as a target cell population of field RABV. This suggests that virus release from axons and infected SCs is part of the RABV in vivo cycle and may affect RABV-related demyelination of peripheral neurons and local innate immune responses. Detection of RABV in axon-surrounding myelinating SCs after i.c. infection further provided evidence for anterograde spread of RABV, highlighting that RABV axonal transport and spread of infectious virus in peripheral nerves is not exclusively retrograde. Our data support a new model in which, comparable to CNS neuroglia, SC infection in peripheral nerves suppresses glia-mediated innate immunity and delays antiviral host responses required for successful transport from the peripheral infection sites to the brain.


Asunto(s)
Transporte Axonal , Encéfalo/virología , Inmunidad Innata/inmunología , Neuroglía/virología , Neuronas/virología , Nervios Periféricos/virología , Virus de la Rabia/patogenicidad , Tropismo Viral , Animales , Axones/metabolismo , Axones/patología , Axones/virología , Encéfalo/inmunología , Encéfalo/patología , Imagenología Tridimensional , Ratones , Microscopía Confocal , Neuroglía/inmunología , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Nervios Periféricos/inmunología , Nervios Periféricos/patología , ARN Viral , Rabia , Células de Schwann/inmunología , Células de Schwann/patología , Células de Schwann/virología
10.
J Virol ; 94(23)2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32938770

RESUMEN

The herpes simplex virus (HSV) heterodimer gE/gI and another membrane protein, US9, which has neuron-specific effects, promote the anterograde transport of virus particles in neuronal axons. Deletion of both HSV gE and US9 blocks the assembly of enveloped particles in the neuronal cytoplasm, which explains why HSV virions do not enter axons. Cytoplasmic envelopment depends upon interactions between viral membrane proteins and tegument proteins that encrust capsids. We report that tegument protein UL16 is unstable, i.e., rapidly degraded, in neurons infected with a gE-/US9- double mutant. Immunoprecipitation experiments with lysates of HSV-infected neurons showed that UL16 and three other tegument proteins, namely, VP22, UL11, and UL21, bound either to gE or gI. All four of these tegument proteins were also pulled down with US9. In neurons transfected with tegument proteins and gE/gI or US9, there was good evidence that VP22 and UL16 bound directly to US9 and gE/gI. However, there were lower quantities of these tegument proteins that coprecipitated with gE/gI and US9 from transfected cells than those of infected cells. This apparently relates to a matrix of several different tegument proteins formed in infected cells that bind to gE/gI and US9. In cells transfected with individual tegument proteins, this matrix is less prevalent. Similarly, coprecipitation of gE/gI and US9 was observed in HSV-infected cells but not in transfected cells, which argued against direct US9-gE/gI interactions. These studies suggest that gE/gI and US9 binding to these tegument proteins has neuron-specific effects on virus HSV assembly, a process required for axonal transport of enveloped particles.IMPORTANCE Herpes simplex viruses 1 and 2 and varicella-zoster virus cause significant morbidity and mortality. One basic property of these viruses is the capacity to establish latency in the sensory neurons and to reactivate from latency and then cause disease in peripheral tissues, such as skin and mucosal epithelia. The transport of nascent HSV particles from neuron cell bodies into axons and along axons to axon tips in the periphery is an important component of this reactivation and reinfection. Two HSV membrane proteins, gE/gI and US9, play an essential role in these processes. Our studies help elucidate how HSV gE/gI and US9 promote the assembly of virus particles and sorting of these virions into neuronal axons.


Asunto(s)
Axones/virología , Herpes Simple/virología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipoproteínas/metabolismo , Simplexvirus/metabolismo , Proteínas Virales/metabolismo , Transporte Axonal/fisiología , Cápside/metabolismo , Línea Celular , Citoplasma/virología , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 2 , Transporte de Proteínas , Proteínas Reguladoras y Accesorias Virales/metabolismo , Proteínas Estructurales Virales/metabolismo , Virión/metabolismo , Ensamble de Virus
11.
ACS Chem Neurosci ; 11(13): 1868-1870, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32605374
12.
Crit Rev Microbiol ; 46(4): 381-396, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32715819

RESUMEN

Alzheimer's disease (AD) is a multifactorial disease triggered by environmental factors in combination with genetic predisposition. Infectious agents, in particular herpes simplex virus type 1 (HSV-1), are gradually being recognised as important factors affecting the development of AD. However, the mechanism linking HSV-1 and AD remains unknown. Of note, HSV-1 manipulates the activity of cofilin-1 to ensure their efficient infection in neuron cells. Cofilin-1, the main regulator of actin cytoskeleton reorganization, is implicating for the plastic of dendritic spines and axon regeneration of neuronal cells. Moreover, dysfunction of cofilin-1 is observed in most AD patients, as well as in mice with AD and ageing. Further, inhibition of cofilin-1 activity ameliorates the host cognitive impairment in an animal model of AD. Together, dysregulation of cofilin-1 led by HSV-1 infection is a potential link between HSV-1 and AD. Herein, we critically summarize the role of cofilin-1-mediated actin dynamics in both HSV-1 infection and AD, respectively. We also propose several hypotheses regarding the connecting roles of cofilin-1 dysregulation in HSV-1 infection and AD. Our review provides a foundation for future studies targeting individuals carrying HSV-1 in combination with cofilin-1 to promote a more individualised approach for treatment and prevention of AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/virología , Cofilina 1/metabolismo , Herpes Simple/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Enfermedad de Alzheimer/genética , Animales , Axones/metabolismo , Axones/virología , Cofilina 1/genética , Herpes Simple/genética , Herpesvirus Humano 1/genética , Humanos , Neuronas/metabolismo , Neuronas/virología
13.
J Virol ; 94(14)2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32376626

RESUMEN

Expression of viral genes and activation of innate antiviral responses during infection result in an increase in reactive oxygen species (ROS) and toxic by-products of energy metabolism which can lead to cell death. The mitochondrion and its associated proteins are crucial regulators of these responses and related pathways such as autophagy and apoptosis. Through a mass spectrometry approach, we have shown that the herpes simplex virus 1 (HSV-1) neurovirulence- and autophagy-modulating protein ICP34.5 interacts with numerous mitochondrion-associated factors. Specifically, we showed that amino acids 68 to 87 of ICP34.5, the domain that binds beclin1 and controls neurovirulence, are necessary for interactions with PGAM5, KEAP1, and other regulators of the antioxidant response, mitochondrial trafficking, and programmed cell death. We further show that while this domain interacts with multiple cellular stress response factors, it does not alter apoptosis or antioxidant gene expression. That said, the attenuated replication of a recombinant virus lacking residues 68 to 87 (termed Δ68-87) in primary human fibroblasts was restored by addition of ferric nitrate. Furthermore, in primary mouse neurons, the perinuclear localization of mitochondria that follows infection with HSV-1 was notably absent following Δ68-87 infection. Through this 20-amino-acid domain, ICP34.5 significantly reduces mitochondrial motility in axons of neurons. We propose the hypothesis that ICP34.5 promotes perinuclear mitochondrial localization by modulating transport of mitochondria through interaction with PGAM5. These data expand upon previous observations of altered mitochondrial dynamics following alphaherpesvirus infections and identify a key determinant of this activity during HSV-1 infections.IMPORTANCE Herpes simplex virus persists lifelong in neurons and can reactivate to cause recurrent lesions in mucosal tissues. A key determinant of virulence is the viral protein ICP34.5, of which residues 68 to 87 significantly contribute to neurovirulence through an unknown mechanism. Our report provides evidence that residues 68 to 87 of ICP34.5 are required for binding mitochondrion-associated factors. These interactions alter mitochondrial dynamics in neurons, thereby facilitating viral replication and pathogenesis.


Asunto(s)
Axones/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Mitocondrias/metabolismo , Proteínas Virales/metabolismo , Axones/patología , Axones/virología , Células HEK293 , Herpes Simple/patología , Herpesvirus Humano 1/genética , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/genética , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Mitocondrias/genética , Mitocondrias/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Dominios Proteicos , Transporte de Proteínas , Proteínas Virales/genética
14.
J Biol Chem ; 295(20): 6926-6935, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32249210

RESUMEN

Mouse hepatitis virus (MHV; murine coronavirus) causes meningoencephalitis, myelitis, and optic neuritis followed by axonal loss and demyelination. This murine virus is used as a common model to study acute and chronic virus-induced demyelination in the central nervous system. Studies with recombinant MHV strains that differ in the gene encoding the spike protein have demonstrated that the spike has a role in MHV pathogenesis and retrograde axonal transport. Fusion peptides (FPs) in the spike protein play a key role in MHV pathogenesis. In a previous study of the effect of deleting a single proline residue in the FP of a demyelinating MHV strain, we found that two central, consecutive prolines are important for cell-cell fusion and pathogenesis. The dihedral fluctuation of the FP was shown to be repressed whenever two consecutive prolines were present, in contrast to the presence of a single proline in the chain. Using this proline-deleted MHV strain, here we investigated whether intracranial injection of this strain can induce optic neuritis by retrograde axonal transport from the brain to the retina through the optic nerve. We observed that the proline-deleted recombinant MHV strain is restricted to the optic nerve, is unable to translocate to the retina, and causes only minimal demyelination and no neuronal death. We conclude that an intact proline dyad in the FP of the recombinant demyelinating MHV strain plays a crucial role in translocation of the virus through axons and subsequent neurodegeneration.


Asunto(s)
Transporte Axonal/genética , Virus de la Hepatitis Murina/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Animales , Transporte Axonal/fisiología , Axones/metabolismo , Axones/virología , Encéfalo/metabolismo , Infecciones por Coronavirus/patología , Enfermedades Desmielinizantes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Virus de la Hepatitis Murina/metabolismo , Nervio Óptico/metabolismo , Nervio Óptico/virología , Péptidos/metabolismo , Prolina/metabolismo , Eliminación de Secuencia/genética , Glicoproteína de la Espiga del Coronavirus/genética , Proteínas del Envoltorio Viral/metabolismo
15.
Viruses ; 12(4)2020 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-32218146

RESUMEN

Rabies is a zoonotic neurological infection caused by lyssavirus that continues to result in devastating loss of human life. Many aspects of rabies pathogenesis in human neurons are not well understood. Lack of appropriate ex-vivo models for studying rabies infection in human neurons has contributed to this knowledge gap. In this study, we utilize advances in stem cell technology to characterize rabies infection in human stem cell-derived neurons. We show key cellular features of rabies infection in our human neural cultures, including upregulation of inflammatory chemokines, lack of neuronal apoptosis, and axonal transmission of viruses in neuronal networks. In addition, we highlight specific differences in cellular pathogenesis between laboratory-adapted and field strain lyssavirus. This study therefore defines the first stem cell-derived ex-vivo model system to study rabies pathogenesis in human neurons. This new model system demonstrates the potential for enabling an increased understanding of molecular mechanisms in human rabies, which could lead to improved control methods.


Asunto(s)
Lyssavirus/fisiología , Neuronas/virología , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis , Axones/metabolismo , Axones/virología , Biomarcadores , Calcio/metabolismo , Supervivencia Celular , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Ratones , Imagen Molecular , Virus de la Rabia/fisiología , Infecciones por Rhabdoviridae/virología
16.
PLoS Pathog ; 16(2): e1008380, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32109948

RESUMEN

Several barriers protect the central nervous system (CNS) from pathogen invasion. Yet viral infections of the CNS are common and often debilitating. Understanding how neurotropic viruses co-opt host machinery to overcome challenges to neuronal entry and transmission is important to combat these infections. Neurotropic reovirus disseminates through neural routes and invades the CNS to cause lethal encephalitis in newborn animals. To define mechanisms of reovirus neuronal entry and directional transport, we used primary neuron cultures, which reproduce in vivo infection patterns displayed by different reovirus serotypes. Treatment of neurons with small-molecule inhibitors of different endocytic uptake pathways allowed us to discover that the cellular machinery mediating macropinocytosis is required for reovirus neuronal entry. This mechanism of reovirus entry differs from clathrin-mediated endocytosis, which is used by reovirus to invade non-neuronal cells. Analysis of reovirus transport and release from isolated soma or axonal termini of neurons cultivated in microfluidic devices indicates that reovirus is capable of retrograde but only limited anterograde neuronal transmission. The dynamics of retrograde reovirus movement are consistent with fast axonal transport coordinated by dynein along microtubules. Further analysis of viral transport revealed that multiple virions are transported together in axons within non-acidified vesicles. Reovirus-containing vesicles acidify after reaching the soma, where disassembly of virions and release of the viral core into the cytoplasm initiates replication. These results define mechanisms of reovirus neuronal entry and transport and establish a foundation to identify common host factors used by neuroinvasive viruses. Furthermore, our findings emphasize consideration of cell type-specific entry mechanisms in the tailored design of neurotropic viruses as tracers, oncolytic agents, and delivery vectors.


Asunto(s)
Transporte Axonal/fisiología , Infecciones por Reoviridae/metabolismo , Reoviridae/metabolismo , Animales , Axones/virología , Línea Celular , Sistema Nervioso Central , Citoplasma/metabolismo , Endocitosis , Masculino , Ratones , Microtúbulos/metabolismo , Neuronas/metabolismo , Neuronas/virología , Pinocitosis/fisiología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Reoviridae/genética , Virión/metabolismo , Internalización del Virus
17.
J Virol ; 94(9)2020 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-32075931

RESUMEN

Alphaherpesviruses, including pseudorabies virus (PRV), are neuroinvasive pathogens that establish lifelong latency in peripheral ganglia following the initial infection at mucosal surfaces. The establishment of latent infection and subsequent reactivations, during which newly assembled virions are sorted into and transported anterogradely inside axons to the initial mucosal site of infection, rely on axonal bidirectional transport mediated by microtubule-based motors. Previous studies using cultured peripheral nervous system (PNS) neurons have demonstrated that KIF1A, a kinesin-3 motor, mediates the efficient axonal sorting and transport of newly assembled PRV virions. Here we report that KIF1A, unlike other axonal kinesins, is an intrinsically unstable protein prone to proteasomal degradation. Interestingly, PRV infection of neuronal cells leads not only to a nonspecific depletion of KIF1A mRNA but also to an accelerated proteasomal degradation of KIF1A proteins, leading to a near depletion of KIF1A protein late in infection. Using a series of PRV mutants deficient in axonal sorting and anterograde spread, we identified the PRV US9/gE/gI protein complex as a viral factor facilitating the proteasomal degradation of KIF1A proteins. Moreover, by using compartmented neuronal cultures that fluidically and physically separate axons from cell bodies, we found that the proteasomal degradation of KIF1A occurs in axons during infection. We propose that the PRV anterograde sorting complex, gE/gI/US9, recruits KIF1A to viral transport vesicles for axonal sorting and transport and eventually accelerates the proteasomal degradation of KIF1A in axons.IMPORTANCE Pseudorabies virus (PRV) is an alphaherpesvirus related to human pathogens herpes simplex viruses 1 and 2 and varicella-zoster virus. Alphaherpesviruses are neuroinvasive pathogens that establish lifelong latent infections in the host peripheral nervous system (PNS). Following reactivation from latency, infection spreads from the PNS back via axons to the peripheral mucosal tissues, a process mediated by kinesin motors. Here, we unveil and characterize the underlying mechanisms for a PRV-induced, accelerated degradation of KIF1A, a kinesin-3 motor promoting the sorting and transport of PRV virions in axons. We show that PRV infection disrupts the synthesis of KIF1A and simultaneously promotes the degradation of intrinsically unstable KIF1A proteins by proteasomes in axons. Our work implies that the timing of motor reduction after reactivation would be critical because progeny particles would have a limited time window for sorting into and transport in axons for further host-to-host spread.


Asunto(s)
Herpesvirus Suido 1/metabolismo , Cinesinas/metabolismo , Seudorrabia/metabolismo , Animales , Transporte Axonal/fisiología , Axones/virología , Línea Celular , Células Cultivadas , Interacciones Huésped-Patógeno , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinesinas/fisiología , Masculino , Microtúbulos/metabolismo , Neuronas/virología , Cultivo Primario de Células , Ratas , Ratas Sprague-Dawley , Proteínas del Envoltorio Viral/genética , Virión/metabolismo
18.
J Virol ; 94(8)2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-31969440

RESUMEN

The features of herpes simplex virus 1 (HSV-1) strain 129 (H129), including natural neurotropism and anterograde transneuronal trafficking, make it a potential tool for anterograde neural circuitry tracing. Recently anterograde polysynaptic and monosynaptic tracers were developed from H129 and have been applied for the identification of novel connections and functions of different neural circuitries. However, how H129 viral particles are transported in neurons, especially those of the central nervous system, remains unclear. In this study, we constructed recombinant H129 variants with mCherry-labeled capsids and/or green fluorescent protein (GFP)-labeled envelopes and infected the cortical neurons to study axonal transport of H129 viral particles. We found that different types of viral particles were unevenly distributed in the nucleus, cytoplasm of the cell body, and axon. Most H129 progeny particles were unenveloped capsids and were transported as capsids rather than virions in the axon. Notably, capsids acquired envelopes at axonal varicosities and terminals where the sites forming synapses are connected with other neurons. Moreover, viral capsids moved more frequently in the anterograde direction in axons, with an average velocity of 0.62 ± 0.18 µm/s and maximal velocity of 1.80 ± 0.15 µm/s. We also provided evidence that axonal transport of capsids requires the kinesin-1 molecular motor. These findings support that H129-derived tracers map the neural circuit anterogradely and possibly transsynaptically. These data will guide future modifications and improvements of H129-based anterograde viral tracers.IMPORTANCE Anterograde transneuronal tracers derived from herpes simplex virus 1 (HSV-1) strain 129 (H129) are important tools for mapping neural circuit anatomic and functional connections. It is, therefore, critical to elucidate the transport pattern of H129 within neurons and between neurons. We constructed recombinant H129 variants with genetically encoded fluorescence-labeled capsid protein and/or glycoprotein to visualize viral particle movement in neurons. Both electron microscopy and light microscopy data show that H129 capsids and envelopes move separately, and notably, capsids are enveloped at axonal varicosity and terminals, which are the sites forming synapses to connect with other neurons. Superresolution microscopy-based colocalization analysis and inhibition of H129 particle movement by inhibitors of molecular motors support that kinesin-1 contributes to the anterograde transport of capsids. These results shed light into the mechanisms for anterograde transport of H129-derived tracer in axons and transmission between neurons via synapses, explaining the anterograde labeling of neural circuits by H129-derived tracers.


Asunto(s)
Cápside/metabolismo , Herpes Simple/virología , Herpesvirus Humano 1/fisiología , Neuronas/virología , Animales , Transporte Axonal , Axones/patología , Axones/virología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Glicoproteínas/metabolismo , Proteínas Fluorescentes Verdes , Herpes Simple/patología , Herpesvirus Humano 1/genética , Cinesinas/metabolismo , Ratones , Ratones Endogámicos C57BL/embriología , Neuronas/patología , Células Vero , Virión/metabolismo
20.
PLoS Pathog ; 16(1): e1007985, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31995633

RESUMEN

Axonal sorting, the controlled passage of specific cargoes from the cell soma into the axon compartment, is critical for establishing and maintaining the polarity of mature neurons. To delineate axonal sorting events, we took advantage of two neuroinvasive alpha-herpesviruses. Human herpes simplex virus 1 (HSV-1) and pseudorabies virus of swine (PRV; suid herpesvirus 1) have evolved as robust cargo of axonal sorting and transport mechanisms. For efficient axonal sorting and subsequent egress from axons and presynaptic termini, progeny capsids depend on three viral membrane proteins (Us7 (gI), Us8 (gE), and Us9), which engage axon-directed kinesin motors. We present evidence that Us7-9 of the veterinary pathogen pseudorabies virus (PRV) form a tripartite complex to recruit Kif1a, a kinesin-3 motor. Based on multi-channel super-resolution and live TIRF microscopy, complex formation and motor recruitment occurs at the trans-Golgi network. Subsequently, progeny virus particles enter axons as enveloped capsids in a transport vesicle. Artificial recruitment of Kif1a using a drug-inducible heterodimerization system was sufficient to rescue axonal sorting and anterograde spread of PRV mutants devoid of Us7-9. Importantly, biophysical evidence suggests that Us9 is able to increase the velocity of Kif1a, a previously undescribed phenomenon. In addition to elucidating mechanisms governing axonal sorting, our results provide further insight into the composition of neuronal transport systems used by alpha-herpesviruses, which will be critical for both inhibiting the spread of infection and the safety of herpesvirus-based oncolytic therapies.


Asunto(s)
Axones/virología , Cápside/metabolismo , Herpes Simple/metabolismo , Herpesvirus Humano 1/metabolismo , Herpesvirus Suido 1/metabolismo , Cinesinas/metabolismo , Seudorrabia/metabolismo , Animales , Transporte Axonal , Axones/metabolismo , Herpes Simple/genética , Herpes Simple/virología , Herpesvirus Humano 1/genética , Herpesvirus Suido 1/genética , Interacciones Huésped-Patógeno , Humanos , Cinesinas/genética , Unión Proteica , Seudorrabia/genética , Seudorrabia/virología , Porcinos , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Red trans-Golgi/metabolismo , Red trans-Golgi/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...