Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
mBio ; : e0144524, 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38953638

RESUMEN

Neurotropic alphaherpesviruses, including herpes simplex virus type 1 and pseudorabies virus, establish a lifelong presence within the peripheral nervous system of their mammalian hosts. Upon entering cells, two conserved tegument proteins, pUL36 and pUL37, traffic DNA-containing capsids to nuclei. These proteins support long-distance retrograde axonal transport and invasion of the nervous system in vivo. To better understand how pUL36 and pUL37 function, recombinant viral particles carrying BioID2 fused to these proteins were produced to biotinylate cellular proteins in their proximity (<10 nm) during infection. Eighty-six high-confidence host proteins were identified by mass spectrometry and subsequently targeted by CRISPR-Cas9 gene editing to assess their contributions to early infection. Proteins were identified that both supported and antagonized infection in immortalized human epithelial cells. The latter included zyxin, a protein that localizes to focal adhesions and regulates actin cytoskeletal dynamics. Zyxin knockout cells were hyper-permissive to infection and could be rescued with even modest expression of GFP-zyxin. These results provide a resource for studies of the virus-cell interface and identify zyxin as a novel deterrent to alphaherpesvirus infection.IMPORTANCENeuroinvasive alphaherpesviruses are highly prevalent with many members found across mammals [e.g., herpes simplex virus type 1 (HSV-1) in humans and pseudorabies virus in pigs]. HSV-1 causes a range of clinical manifestations from cold sores to blindness and encephalitis. There are no vaccines or curative therapies available for HSV-1. A fundamental feature of these viruses is their establishment of lifelong infection of the nervous system in their respective hosts. This outcome is possible due to a potent neuroinvasive property that is coordinated by two proteins: pUL36 and pUL37. In this study, we explore the cellular protein network in proximity to pUL36 and pUL37 during infection and examine the impact of knocking down the expression of these proteins upon infection.

2.
Proc Natl Acad Sci U S A ; 121(19): e2401341121, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38696466

RESUMEN

Neurotropic alphaherpesviruses, including herpes simplex virus type 1 (HSV-1), recruit microtubule motor proteins to invade cells. The incoming viral particle traffics to nuclei in a two-step process. First, the particle uses the dynein-dynactin motor to sustain transport to the centrosome. In neurons, this step is responsible for long-distance retrograde axonal transport and is an important component of the neuroinvasive property shared by these viruses. Second, a kinesin-dependent mechanism redirects the particle from the centrosome to the nucleus. We have reported that the kinesin motor used during the second step of invasion is assimilated into nascent virions during the previous round of infection. Here, we report that the HSV-1 pUL37 tegument protein suppresses the assimilated kinesin-1 motor during retrograde axonal transport. Region 2 (R2) of pUL37 was required for suppression and functioned independently of the autoinhibitory mechanism native to kinesin-1. Furthermore, the motor domain and proximal coiled coil of kinesin-1 were sufficient for HSV-1 assimilation, pUL37 suppression, and nuclear trafficking. pUL37 localized to the centrosome, the site of assimilated kinesin-1 activation during infection, when expressed in cells in the absence of other viral proteins; however, pUL37 did not suppress kinesin-1 in this context. These results indicate that the pUL37 tegument protein spatially and temporally regulates kinesin-1 via the amino-terminal motor region in the context of the incoming viral particle.


Asunto(s)
Herpesvirus Humano 1 , Cinesinas , Proteínas Estructurales Virales , Cinesinas/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/metabolismo , Humanos , Animales , Transporte Axonal/fisiología , Chlorocebus aethiops , Centrosoma/metabolismo , Neuronas/metabolismo , Neuronas/virología , Células Vero , Núcleo Celular/metabolismo , Núcleo Celular/virología
3.
J Spec Oper Med ; 23(4): 47-56, 2023 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-37851859

RESUMEN

United States Special Operations Forces (SOF) personnel are frequently exposed to explosive blasts in training and combat. However, the effects of repeated blast exposure on the human brain are incompletely understood. Moreover, there is currently no diagnostic test to detect repeated blast brain injury (rBBI). In this "Human Performance Optimization" article, we discuss how the development and implementation of a reliable diagnostic test for rBBI has the potential to promote SOF brain health, combat readiness, and quality of life.


Asunto(s)
Traumatismos por Explosión , Personal Militar , Humanos , Estados Unidos , Calidad de Vida , Encéfalo/diagnóstico por imagen , Traumatismos por Explosión/diagnóstico , Traumatismos por Explosión/terapia , Explosiones
4.
Sci Immunol ; 8(82): eade2860, 2023 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-37083451

RESUMEN

Inborn errors of TLR3-dependent type I IFN immunity in cortical neurons underlie forebrain herpes simplex virus-1 (HSV-1) encephalitis (HSE) due to uncontrolled viral growth and subsequent cell death. We report an otherwise healthy patient with HSE who was compound heterozygous for nonsense (R422*) and frameshift (P493fs9*) RIPK3 variants. Receptor-interacting protein kinase 3 (RIPK3) is a ubiquitous cytoplasmic kinase regulating cell death outcomes, including apoptosis and necroptosis. In vitro, the R422* and P493fs9* RIPK3 proteins impaired cellular apoptosis and necroptosis upon TLR3, TLR4, or TNFR1 stimulation and ZBP1/DAI-mediated necroptotic cell death after HSV-1 infection. The patient's fibroblasts displayed no detectable RIPK3 expression. After TNFR1 or TLR3 stimulation, the patient's cells did not undergo apoptosis or necroptosis. After HSV-1 infection, the cells supported excessive viral growth despite normal induction of antiviral IFN-ß and IFN-stimulated genes (ISGs). This phenotype was, nevertheless, rescued by application of exogenous type I IFN. The patient's human pluripotent stem cell (hPSC)-derived cortical neurons displayed impaired cell death and enhanced viral growth after HSV-1 infection, as did isogenic RIPK3-knockout hPSC-derived cortical neurons. Inherited RIPK3 deficiency therefore confers a predisposition to HSE by impairing the cell death-dependent control of HSV-1 in cortical neurons but not their production of or response to type I IFNs.


Asunto(s)
Encefalitis por Herpes Simple , Herpes Simple , Herpesvirus Humano 1 , Humanos , Muerte Celular , Encefalitis por Herpes Simple/genética , Herpesvirus Humano 1/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Receptores Tipo I de Factores de Necrosis Tumoral , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo
5.
J Virol ; 96(9): e0148621, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35420461

RESUMEN

Following exposure and replication at mucosal surfaces, most alphaherpesviruses invade the peripheral nervous system by retrograde axonal transport and establish lifelong latent infections in the peripheral ganglia. Reactivation of ganglionic infections is followed by anterograde axonal transport of virions back to body surfaces where viral replication results in disease that can range from moderate to severe in presentation. In the case of bovine herpesvirus 1 (BoHV-1), replication in the epithelial mucosa presents as infectious bovine rhinotracheitis (IBR), a respiratory disease of significant economic impact. In this study, we provide a live-cell analysis of BoHV-1 retrograde axonal transport relative to the model alphaherpesvirus pathogen pseudorabies virus (PRV) and demonstrate that this critical neuroinvasive step is conserved between the two viruses. In addition, we report that the BoHV-1 pUL37 tegument protein supports processive retrograde motion in infected axons and invasion of the calf peripheral nervous system. IMPORTANCE A molecular and cellular understanding of the retrograde axonal transport process that underlies the neuroinvasive properties of the alphaherpesviruses is established from studies of herpes simplex virus and pseudorabies virus. The degree to which this phenotype is conserved in other related viruses has largely not been examined. We provide a time-lapse analysis of the retrograde axonal transport kinetics of bovine herpesvirus 1 and demonstrate that mutation of the pUL37 region 2 effector affords a strategy to produce live-attenuated vaccines for enhanced protection of cattle.


Asunto(s)
Transporte Axonal , Herpesvirus Bovino 1 , Células Receptoras Sensoriales , Proteínas Virales , Animales , Axones , Bovinos , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/patogenicidad , Células Receptoras Sensoriales/virología , Proteínas Virales/genética
6.
Integr Org Biol ; 4(1): obab037, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35112052

RESUMEN

For many animals, the juvenile stage of life can be particularly perilous. Once independent, immature animals must often complete the same basic survival functions as adults despite smaller body size and other growth-related limits on performance. Because, by definition, juveniles have yet to reproduce, we should expect strong selection for mechanisms to offset these ontogenetic limitations, allowing individuals to reach reproductive adulthood and maintain Darwinian fitness. We use an integrated ontogenetic dataset on morphology, locomotor performance, and longevity in wild cottontail rabbits (Sylvilagus floridanus, Allen 1848) to test the hypothesis that prey animals are under selective pressure to maximize juvenile performance. We predicted that (1) juveniles would accelerate more quickly than adults, allowing them to reach adult-like escape speeds, and (2) juveniles with greater levels of performance should survive for longer durations in the wild, thus increasing their reproductive potential. Using high-speed video and force platform measurements, we quantified burst acceleration, escape speed, and mechanical power production in 38 wild-caught S. floridanus (26 juveniles, 12 adults; all rabbits >1 kg in body mass were designated to be adults, based on published growth curves and evidence of epiphyseal fusion). A subsample of 22 rabbits (15 juveniles, 7 adults) was fitted with radio-telemetry collars for documenting survivorship in the wild. We found that acceleration and escape speed peaked in the late juvenile period in S. floridanus, at an age range that coincides with a period of pronounced demographic attrition in wild populations. Differences in mass-specific mechanical power production explained ∼75% of the variation in acceleration across the dataset, indicating that juvenile rabbits outpace adults by producing more power per unit body mass. We found a positive, though non-significant, association between peak escape speed and survivorship duration in the wild, suggesting a complex relationship between locomotor performance and fitness in growing S. floridanus.

7.
Nature ; 599(7886): 662-666, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34789877

RESUMEN

Neurotropic alphaherpesviruses initiate infection in exposed mucosal tissues and, unlike most viruses, spread rapidly to sensory and autonomic nerves where life-long latency is established1. Recurrent infections arise sporadically from the peripheral nervous system throughout the life of the host, and invasion of the central nervous system may occur, with severe outcomes2. These viruses directly recruit cellular motors for transport along microtubules in nerve axons, but how the motors are manipulated to deliver the virus to neuronal nuclei is not understood. Here, using herpes simplex virus type I and pseudorabies virus as model alphaherpesviruses, we show that a cellular kinesin motor is captured by virions in epithelial cells, carried between cells, and subsequently used in neurons to traffic to nuclei. Viruses assembled in the absence of kinesin are not neuroinvasive. The findings explain a critical component of the alphaherpesvirus neuroinvasive mechanism and demonstrate that these viruses assimilate a cellular protein as an essential proviral structural component. This principle of viral assimilation may prove relevant to other virus families and offers new strategies to combat infection.


Asunto(s)
Herpesvirus Humano 1/metabolismo , Herpesvirus Suido 1/metabolismo , Cinesinas/metabolismo , Movimiento , Virión/metabolismo , Ensamble de Virus , Animales , Transporte Biológico , Cápside/metabolismo , Línea Celular , Núcleo Celular/virología , Chlorocebus aethiops , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Neuronas/metabolismo , Neuronas/virología , Conejos , Porcinos
8.
Elife ; 102021 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-33851917

RESUMEN

Sensory-guided limb control relies on communication across sensorimotor loops. For active touch with the hand, the longest loop is the transcortical continuation of ascending pathways, particularly the lemnisco-cortical and corticocortical pathways carrying tactile signals via the cuneate nucleus, ventral posterior lateral (VPL) thalamus, and primary somatosensory (S1) and motor (M1) cortices to reach corticospinal neurons and influence descending activity. We characterized excitatory connectivity along this pathway in the mouse. In the lemnisco-cortical leg, disynaptic cuneate→VPL→S1 connections excited mainly layer (L) 4 neurons. In the corticocortical leg, S1→M1 connections from L2/3 and L5A neurons mainly excited downstream L2/3 neurons, which excite corticospinal neurons. The findings provide a detailed new wiring diagram for the hand/forelimb-related transcortical circuit, delineating a basic but complex set of cell-type-specific feedforward excitatory connections that selectively and extensively engage diverse intratelencephalic projection neurons, thereby polysynaptically linking subcortical somatosensory input to cortical motor output to spinal cord.


Asunto(s)
Miembro Anterior/inervación , Corteza Motora/fisiología , Corteza Somatosensorial/fisiología , Núcleos Talámicos Ventrales/fisiología , Animales , Femenino , Masculino , Ratones
9.
J Clin Invest ; 131(1)2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33393505

RESUMEN

Human herpes simplex virus 1 (HSV-1) encephalitis can be caused by inborn errors of the TLR3 pathway, resulting in impairment of CNS cell-intrinsic antiviral immunity. Deficiencies of the TLR3 pathway impair cell-intrinsic immunity to vesicular stomatitis virus (VSV) and HSV-1 in fibroblasts, and to HSV-1 in cortical but not trigeminal neurons. The underlying molecular mechanism is thought to involve impaired IFN-α/ß induction by the TLR3 recognition of dsRNA viral intermediates or by-products. However, we show here that human TLR3 controls constitutive levels of IFNB mRNA and secreted bioactive IFN-ß protein, and thereby also controls constitutive mRNA levels for IFN-stimulated genes (ISGs) in fibroblasts. Tlr3-/- mouse embryonic fibroblasts also have lower basal ISG levels. Moreover, human TLR3 controls basal levels of IFN-ß secretion and ISG mRNA in induced pluripotent stem cell-derived cortical neurons. Consistently, TLR3-deficient human fibroblasts and cortical neurons are vulnerable not only to both VSV and HSV-1, but also to several other families of viruses. The mechanism by which TLR3 restricts viral growth in human fibroblasts and cortical neurons in vitro and, by inference, by which the human CNS prevents infection by HSV-1 in vivo, is therefore based on the control of early viral infection by basal IFN-ß immunity.


Asunto(s)
Corteza Cerebral/inmunología , Fibroblastos/inmunología , Herpesvirus Humano 1/inmunología , Interferón beta/inmunología , Neuronas/inmunología , Receptor Toll-Like 3/inmunología , Vesiculovirus/inmunología , Animales , Línea Celular , Corteza Cerebral/patología , Corteza Cerebral/virología , Fibroblastos/patología , Fibroblastos/virología , Humanos , Interferón beta/genética , Ratones , Ratones Noqueados , Neuronas/patología , Neuronas/virología , Receptor Toll-Like 3/genética
10.
Curr Issues Mol Biol ; 41: 171-220, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32807747

RESUMEN

Herpesviruses virions are large and complex structures that deliver their genetic content to nuclei upon entering cells. This property is not unusual as many other viruses including the adenoviruses, orthomyxoviruses, papillomaviruses, polyomaviruses, and retroviruses, do likewise. However, the means by which viruses in the alphaherpesvirinae subfamily accomplish this fundamental stage of the infectious cycle is tied to their defining ability to efficiently invade the nervous system. Fusion of the viral envelope with a cell membrane results in the deposition of the capsid, along with an assortment of tegument proteins, into the cytosol. Establishment of infection requires that the capsid traverse the cytosol, dock at a nuclear pore, and inject its genome into the nucleoplasm. Accumulating evidence indicates that the capsid is not the effector of this delivery process, but is instead shepherded by tegument proteins that remain capsid bound. At the same time, tegument proteins that are released from the capsid upon entry act to increase the susceptibility of the cell to the ensuing infection. Mucosal epithelial cells and neurons are both susceptible to alphaherpesvirus infection and, together, provide the niche to which these viruses have adapted. Although much has been revealed about the functions of de novo expressed tegument proteins during the late stages of assembly and egress, this review will specifically address the roles of tegument proteins brought into the cell with the incoming virion, and our current understanding of alphaherpesvirus genome delivery to nuclei.


Asunto(s)
Alphaherpesvirinae/genética , Alphaherpesvirinae/patogenicidad , Citoplasma/virología , Genoma Viral/genética , Infecciones por Herpesviridae/virología , Animales , Proteínas de la Cápside/genética , Núcleo Celular/virología , Humanos , Virión/genética , Ensamble de Virus/genética , Internalización del Virus
11.
NPJ Vaccines ; 5(1): 104, 2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33298966

RESUMEN

Herpes simplex virus (HSV) infections are common and can cause severe illness but no vaccine is currently available. The recent failure of subunit HSV vaccines has highlighted the need for vaccines that present a diverse array of antigens, including the development of next-generation live-attenuated vaccines. However, most attenuated HSV strains propagate poorly, limiting their ability to elicit protective immune responses. A live-attenuated vaccine that replicates in non-neural tissue but is ablated for transmission into the nervous system may elicit protective immune responses without evoking neurologic complications or establishing life-long infections. Initial studies of R2, a live-attenuated vaccine that is engineered to be unable to invade the nervous system, used the guinea pig genital HSV model to evaluate the ability of R2 to replicate at the site of inoculation, cause disease and infect neural tissues. R2 was then evaluated as a vaccine using three routes of inoculation: intramuscular (IM), intradermal (ID) and intravaginal (IVag) and compared to IM administered gD2+MPL/Alum vaccine in the same model. R2 replicated in the genital tract but did not produce acute or recurrent disease and did not infect the neural tissue. The R2 vaccine-induced neutralizing antibody and decreased the severity of acute and recurrent HSV-2 disease as well as recurrent shedding. The ID route was the most effective. ID administered R2 was more effective than gD2+MPL/Alum at inducing neutralizing antibody, suppressing acute disease, and acute vaginal virus replication. R2 was especially more effective at reducing recurrent virus shedding, the most common source of HSV transmission. The live-attenuated prophylactic HSV vaccine, R2, was effective in the guinea pig model of genital HSV-2 especially when administered by the ID route. The use of live-attenuated HSV vaccines that robustly replicate in mucosal tissues but are ablated for neuroinvasion offers a promising approach for HSV vaccines.

12.
Vaccine ; 38(29): 4524-4528, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32448623

RESUMEN

Neurotropic alpha-herpesviruses that infect mammals establish life-long latent infections in the peripheral nervous system after initial infection of exposed mucosal tissues. The neuroinvasive properties can lead to severe complications both with clinical and veterinary alpha-herpesviruses, and vaccines are often unavailable or provide limited protection. Here we assess the properties and efficacy of an R2 vaccine derived from the alpha-herpesvirus, pseudorabies virus (PRV), in pigs. We demonstrate that the PRV R2 vaccine does not invade the porcine peripheral nervous system within the limits of detection. Furthermore, after a single intranasal vaccination, R2 conferred protection to pigs subsequently challenged with a virulent PRV field strain (NIA-3). These findings support that the R2 vaccine design is non-neuroinvasive and is an effective vaccine in the context of a natural host.


Asunto(s)
Herpesvirus Suido 1 , Seudorrabia , Enfermedades de los Porcinos , Vacunas , Vacunas Virales , Animales , Anticuerpos Antivirales , Seudorrabia/prevención & control , Vacunas contra la Seudorrabia , Porcinos , Enfermedades de los Porcinos/prevención & control
13.
PLoS One ; 14(12): e0227028, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31887177

RESUMEN

Urban landscapes can present ecological challenges for wildlife species, yet many species survive, and even thrive, near dense human populations. Coyotes (Canis latrans), for example, have expanded their geographic range across North America and, as a result of their adaptability and behavioral flexibility, are now a common occupant of many urban areas in the United States. We investigated the spatial ecology of 27 coyotes fitted with Global Positioning System (GPS) telemetry collars radio-collared in the Cuyahoga Valley, Ohio. Our objectives were to quantify coyote space use, evaluate resource selection, and investigate coyote movement and activity patterns. To measure space use, we estimated home range (95%) and core area (50%) size of coyotes using the adaptive local convex hull (a-LoCoH) method. We found the mean (± SE) home range size of resident coyotes (4.7 ± 1.8 km2) was significantly smaller than ranges of transient coyotes (67.7 ± 89.6 km2). Similarly, mean (± SE) core area size of resident coyotes (0.9 ± 0.6 km2) was significantly smaller than core areas of transient coyotes (11.9 ± 16.7 km2). Home range and core area size of both resident and transient coyotes did not vary by sex, age, or season. For all coyotes, use of natural land cover was significantly greater than use of altered and developed land. When coyotes were using altered and developed land, GPS fixes were most common at night. Coyote movement patterns differed with respect to status, time period, and season; peaking during nighttime hours. A better understanding of coyote space use and movement within anthropogenic landscapes aids management of people, parks, and wildlife by providing the data necessary for research-based management decisions.


Asunto(s)
Migración Animal/fisiología , Coyotes/fisiología , Fenómenos de Retorno al Lugar Habitual/fisiología , Análisis Espacio-Temporal , Urbanización , Factores de Edad , Animales , Femenino , Sistemas de Información Geográfica , Masculino , Ohio , Fotoperiodo , Tecnología de Sensores Remotos , Estaciones del Año , Factores Sexuales
14.
Nat Med ; 25(12): 1873-1884, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31806906

RESUMEN

Herpes simplex virus-1 (HSV-1) encephalitis (HSE) is typically sporadic. Inborn errors of TLR3- and DBR1-mediated central nervous system cell-intrinsic immunity can account for forebrain and brainstem HSE, respectively. We report five unrelated patients with forebrain HSE, each heterozygous for one of four rare variants of SNORA31, encoding a small nucleolar RNA of the H/ACA class that are predicted to direct the isomerization of uridine residues to pseudouridine in small nuclear RNA and ribosomal RNA. We show that CRISPR/Cas9-introduced bi- and monoallelic SNORA31 deletions render human pluripotent stem cell (hPSC)-derived cortical neurons susceptible to HSV-1. Accordingly, SNORA31-mutated patient hPSC-derived cortical neurons are susceptible to HSV-1, like those from TLR3- or STAT1-deficient patients. Exogenous interferon (IFN)-ß renders SNORA31- and TLR3- but not STAT1-mutated neurons resistant to HSV-1. Finally, transcriptome analysis of SNORA31-mutated neurons revealed normal responses to TLR3 and IFN-α/ß stimulation but abnormal responses to HSV-1. Human SNORA31 thus controls central nervous system neuron-intrinsic immunity to HSV-1 by a distinctive mechanism.


Asunto(s)
Encefalitis por Herpes Simple/genética , Herpesvirus Humano 1/genética , Neuronas/inmunología , ARN Nucleolar Pequeño/genética , Adulto , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/virología , Preescolar , Encefalitis por Herpes Simple/inmunología , Encefalitis por Herpes Simple/patología , Encefalitis por Herpes Simple/virología , Femenino , Predisposición Genética a la Enfermedad , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Inmunidad/genética , Lactante , Masculino , Metagenoma/genética , Metagenoma/inmunología , Persona de Mediana Edad , Neuronas/virología , ARN Nucleolar Pequeño/inmunología
15.
Sci Rep ; 9(1): 19731, 2019 12 24.
Artículo en Inglés | MEDLINE | ID: mdl-31874983

RESUMEN

Few insect species are as popular as periodical cicadas (Magicicada spp.). Despite representing an enormous biomass and numbers that exceed 370/m2 during mass emergences, the extended time period of the underground nymphal stages (up to 17 years) complicates investigations of their life history traits and ecology. Upon emergence, female cicadas mate and then use their ovipositors to cut through wood to lay their eggs. Given the ability to penetrate into wood, we hypothesized that the ovipositor cuticle is augmented with inorganic elements, which could increase hardness and reduce ovipositor fracturing. We used scanning electron microscopy and energy dispersive x-ray spectroscopy to evaluate the material properties of ovipositors of four cicada species, including three species of periodical cicadas. We found 14 inorganic elements of the cuticle, of which P, Ca, Si, Mg, Na, Fe, Zn, Mn, Cl, K, and S show the highest concentrations (%wt) near the apex of the ovipositor, where other structural modifications for penetrating wood are present. To the best of our knowledge, this is the first report of metal deposits in the cuticle of true bugs (Hemiptera, >80,000 described species).


Asunto(s)
Hemípteros/fisiología , Metales/metabolismo , Oviposición/fisiología , Madera , Animales , Femenino
16.
J Virol ; 93(22)2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31462572

RESUMEN

Upon replication in mucosal epithelia and transmission to nerve endings, capsids of herpes simplex virus 1 (HSV-1) travel retrogradely within axons to peripheral ganglia, where life-long latent infections are established. A capsid-bound tegument protein, pUL37, is an essential effector of retrograde axonal transport and also houses a deamidase activity that antagonizes innate immune signaling. In this report, we examined whether the deamidase of HSV-1 pUL37 contributes to the neuroinvasive retrograde axonal transport mechanism. We conclude that neuroinvasion is enhanced by the deamidase, but the critical contribution of pUL37 to retrograde axonal transport functions independently of this activity.IMPORTANCE Herpes simplex virus 1 invades the nervous system by entering nerve endings and sustaining long-distance retrograde axonal transport to reach neuronal nuclei in ganglia of the peripheral nervous system. The incoming viral particle carries a deamidase activity on its surface that antagonizes antiviral responses. We examined the contribution of the deamidase to the hallmark neuroinvasive property of this virus.


Asunto(s)
Proteínas de la Cápside/metabolismo , Proteínas Estructurales Virales/metabolismo , Animales , Transporte Axonal/fisiología , Axones/virología , Cápside/metabolismo , Línea Celular , Chlorocebus aethiops , Ganglios/metabolismo , Ganglios/virología , Herpes Simple/virología , Herpesvirus Humano 1/metabolismo , Humanos , Mucosa Intestinal , Neuronas/virología , Células Vero , Proteínas Estructurales Virales/genética , Virión/metabolismo
17.
J Exp Biol ; 222(Pt 16)2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31350298

RESUMEN

Juvenile animals must survive in the same environment as adults despite smaller sizes, immature musculoskeletal tissues, general ecological naïveté and other limits of performance. Developmental changes in muscle leverage could constitute one mechanism to promote increased performance in juveniles despite ontogenetic limitations. We tested this hypothesis using a holistic dataset on growth and locomotor development in wild eastern cottontail rabbits (Sylvilagus floridanus) to examine ontogenetic changes in hindlimb muscle effective mechanical advantage (EMA). EMA is a dimensionless index of muscle leverage, equal to the quotient of average muscle lever length and the load arm length of the ground reaction force (GRF), effectively representing the magnitude of output force arising from a given muscle force. We found that EMA at the hip and ankle joints, as well as overall hindlimb EMA, significantly declined across ontogeny in S. floridanus, whereas EMA at the knee joint remained unchanged. Ontogenetic decreases in EMA were due to isometric scaling of muscle lever arm lengths alongside positive ontogenetic allometry of GRF load arm lengths - which in turn was primarily related to positive allometry of hindlimb segment lengths. Greater EMA limits the estimated volume of hindlimb extensor muscle that has to be activated in young rabbits, likely mitigating the energetic cost of locomotion and saving metabolic resources for other physiological functions, such as growth and tissue differentiation. An additional examination of limb growth allometry across a diverse sample of mammalian taxa suggests that ontogenetic decreases in limb joint EMA may be a common mammalian trend.


Asunto(s)
Lagomorpha/fisiología , Locomoción , Animales , Fenómenos Biomecánicos , Lagomorpha/crecimiento & desarrollo
18.
Proc Natl Acad Sci U S A ; 115(37): E8775-E8782, 2018 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-30154162

RESUMEN

Herpes simplex virus type 1 (HSV-1) encephalitis (HSE) is the most common sporadic viral encephalitis in Western countries. Some HSE children carry inborn errors of the Toll-like receptor 3 (TLR3)-dependent IFN-α/ß- and -λ-inducing pathway. Induced pluripotent stem cell (iPSC)-derived cortical neurons with TLR3 pathway mutations are highly susceptible to HSV-1, due to impairment of cell-intrinsic TLR3-IFN immunity. In contrast, the contribution of cell-intrinsic immunity of human trigeminal ganglion (TG) neurons remains unclear. Here, we describe efficient in vitro derivation and purification of TG neurons from human iPSCs via a cranial placode intermediate. The resulting TG neurons are of sensory identity and exhibit robust responses to heat (capsaicin), cold (icilin), and inflammatory pain (ATP). Unlike control cortical neurons, both control and TLR3-deficient TG neurons were highly susceptible to HSV-1. However, pretreatment of control TG neurons with poly(I:C) induced the cells into an anti-HSV-1 state. Moreover, both control and TLR3-deficient TG neurons developed resistance to HSV-1 following pretreatment with IFN-ß but not IFN-λ. These data indicate that TG neurons are vulnerable to HSV-1 because they require preemptive stimulation of the TLR3 or IFN-α/ß receptors to induce antiviral immunity, whereas cortical neurons possess a TLR3-dependent constitutive resistance that is sufficient to block incoming HSV-1 in the absence of prior antiviral signals. The lack of constitutive resistance in TG neurons in vitro is consistent with their exploitation as a latent virus reservoir in vivo. Our results incriminate deficiencies in the constitutive TLR3-dependent response of cortical neurons in the pathogenesis of HSE.


Asunto(s)
Inmunidad/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Neuronas/metabolismo , Receptor Toll-Like 3/metabolismo , Antivirales/farmacología , Diferenciación Celular/genética , Células Cultivadas , Corteza Cerebral/citología , Niño , Herpesvirus Humano 1/inmunología , Herpesvirus Humano 1/fisiología , Humanos , Inmunidad/genética , Células Madre Pluripotentes Inducidas/citología , Interferón beta/farmacología , Mutación , Neuronas/efectos de los fármacos , Neuronas/virología , Poli I-C/farmacología , Receptor Toll-Like 3/genética , Ganglio del Trigémino/citología
19.
J Virol ; 92(18)2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29976665

RESUMEN

The herpesvirus capsid assembles in the nucleus as an immature procapsid precursor built around viral scaffold proteins. The event that initiates procapsid maturation is unknown, but it is dependent upon activation of the VP24 internal protease. Scaffold cleavage triggers angularization of the shell and its decoration with the VP26 and pUL25 capsid-surface proteins. In both the procapsid and mature angularized capsid, the apical region of the major capsid protein (VP5) is surface exposed. We investigated whether the VP5 apical region contributes to intracellular transport dynamics following entry into primary sensory neurons and also tested the hypothesis that conserved negatively charged amino acids in the apical region contribute to VP26 acquisition. To our surprise, neither hypothesis proved true. Instead, mutation of glutamic acid residues in the apical region delayed viral propagation and induced focal capsid accumulations in nuclei. Examination of capsid morphogenesis based on epitope unmasking, capsid composition, and ultrastructural analysis indicated that these clusters consisted of procapsids. The results demonstrate that, in addition to established events that occur inside the capsid, the exterior capsid shell promotes capsid morphogenesis and maturation.IMPORTANCE Herpesviruses assemble capsids and encapsidate their genomes by a process that is unlike those of other mammalian viruses but is similar to those of some bacteriophage. Many important aspects of herpesvirus morphogenesis remain enigmatic, including how the capsid shell matures into a stable angularized configuration. Capsid maturation is triggered by activation of a protease that cleaves an internal protein scaffold. We report on the fortuitous discovery that a region of the major capsid protein that is exposed on the outer surface of the capsid also contributes to capsid maturation, demonstrating that the morphogenesis of the capsid shell from its procapsid precursor to the mature angularized form is dependent upon internal and external components of the megastructure.


Asunto(s)
Proteínas de la Cápside/genética , Cápside/metabolismo , Herpesvirus Humano 1/fisiología , Proteínas Virales/metabolismo , Animales , Proteínas de la Cápside/metabolismo , Chlorocebus aethiops , Epítopos/química , Epítopos/genética , Epítopos/metabolismo , Herpesvirus Humano 1/química , Mutación , Células Vero , Proteínas Virales/genética , Virión/metabolismo , Ensamble de Virus/fisiología
20.
J Virol ; 92(17)2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29899099

RESUMEN

Herpesvirus particles have a complex architecture consisting of an icosahedral capsid that is surrounded by a lipid envelope. Connecting these two components is a layer of tegument that consists of various amounts of 20 or more proteins. The arrangement of proteins within the tegument cannot easily be assessed and instead is inferred from tegument interactions identified in reductionist models. To better understand the tegument architecture, we have developed an approach to probe capsid-tegument interactions of extracellular viral particles by encoding tobacco etch virus (TEV) protease sites in viral structural proteins, along with distinct fluorescent tags in capsid and tegument components. In this study, TEV sites were engineered within the pUL36 large tegument protein, a critical structural element that is anchored directly on the capsid surface. Purified pseudorabies virus extracellular particles were permeabilized, and TEV protease was added to selectively cleave the exposed pUL36 backbone. Interactions with the capsid were assessed in situ by monitoring the fate of the fluorescent signals following cleavage. Although several regions of pUL36 are proposed to bind capsids, pUL36 was found stably anchored to the capsid exclusively at its carboxyl terminus. Two additional tegument proteins, pUL37 and pUS3, were tethered to the capsid via pUL36, whereas the pUL16, pUL47, pUL48, and pUL49 tegument proteins were not stably bound to the capsid.IMPORTANCE Neuroinvasive alphaherpesviruses produce diseases of clinical and economic significance in humans and veterinary animals but are predominantly associated with less serious recurrent disease. Like all viruses, herpesviruses assemble a metastable particle that selectively dismantles during initial infection. This process is made more complex by the presence of a tegument layer that resides between the capsid surface and envelope. Components of the tegument are essential for particle assembly and also serve as critical effectors that promote infection upon entry into cells. How this dynamic network of protein interactions is arranged within virions is largely unknown. We present a molecular approach to dissect the tegument, and with it we begin to tease apart the protein interactions that underlie this complex layer of the virion architecture.


Asunto(s)
Proteínas de la Cápside/metabolismo , Herpesvirus Suido 1/ultraestructura , Proteínas Estructurales Virales/metabolismo , Estructuras Virales , Animales , Línea Celular , Unión Proteica , Proteolisis , Porcinos , Proteínas Estructurales Virales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA