Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Viruses ; 15(6)2023 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-37376543

RESUMO

Minute Virus of Mice (MVM) is an autonomous parvovirus of the Parvoviridae family that replicates in mouse cells and transformed human cells. MVM genomes localize to cellular sites of DNA damage with the help of their essential non-structural phosphoprotein NS1 to establish viral replication centers. MVM replication induces a cellular DNA damage response that is mediated by signaling through the ATM kinase pathway, while inhibiting induction of the ATR kinase signaling pathway. However, the cellular signals regulating virus localization to cellular DNA damage response sites has remained unknown. Using chemical inhibitors to DNA damage response proteins, we have discovered that NS1 localization to cellular DDR sites is independent of ATM or DNA-PK signaling but is dependent on ATR signaling. Pulsing cells with an ATR inhibitor after S-phase entry leads to attenuated MVM replication. These observations suggest that the initial localization of MVM to cellular DDR sites depends on ATR signaling before it is inactivated by vigorous virus replication.


Assuntos
Vírus Miúdo do Camundongo , Infecções por Parvoviridae , Parvovirus , Humanos , Animais , Camundongos , Vírus Miúdo do Camundongo/fisiologia , Linhagem Celular , Parvovirus/fisiologia , Transdução de Sinais , Dano ao DNA , Replicação Viral/fisiologia , Replicação do DNA , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo
2.
Viruses ; 13(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-34372512

RESUMO

Parvoviruses are small single-stranded (ss) DNA viruses, which replicate in the nucleoplasm and affect both the structure and function of the nucleus. The nuclear stage of the parvovirus life cycle starts at the nuclear entry of incoming capsids and culminates in the successful passage of progeny capsids out of the nucleus. In this review, we will present past, current, and future microscopy and biochemical techniques and demonstrate their potential in revealing the dynamics and molecular interactions in the intranuclear processes of parvovirus infection. In particular, a number of advanced techniques will be presented for the detection of infection-induced changes, such as DNA modification and damage, as well as protein-chromatin interactions.


Assuntos
Núcleo Celular/virologia , Interações entre Hospedeiro e Microrganismos/genética , Parvovirus/genética , Parvovirus/fisiologia , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Interações entre Hospedeiro e Microrganismos/fisiologia , Humanos , Camundongos , Infecções por Parvoviridae/virologia , Replicação Viral/genética , Replicação Viral/fisiologia
3.
Viruses ; 12(6)2020 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-32560452

RESUMO

Several members of the Protoparvovirus genus, capable of infecting humans, have been recently discovered, including cutavirus (CuV) and tusavirus (TuV). To begin the characterization of these viruses, we have used cryo-electron microscopy and image reconstruction to determine their capsid structures to ~2.9 Å resolution, and glycan array and cell-based assays to identify glycans utilized for cellular entry. Structural comparisons show that the CuV and TuV capsids share common features with other parvoviruses, including an eight-stranded anti-parallel ß-barrel, depressions at the icosahedral 2-fold and surrounding the 5-fold axes, and a channel at the 5-fold axes. However, the viruses exhibit significant topological differences in their viral protein surface loops. These result in three separated 3-fold protrusions, similar to the bufaviruses also infecting humans, suggesting a host-driven structure evolution. The surface loops contain residues involved in receptor binding, cellular trafficking, and antigenic reactivity in other parvoviruses. In addition, terminal sialic acid was identified as the glycan potentially utilized by both CuV and TuV for cellular entry, with TuV showing additional recognition of poly-sialic acid and sialylated Lewis X (sLeXLeXLeX) motifs reported to be upregulated in neurotropic and cancer cells, respectively. These structures provide a platform for annotating the cellular interactions of these human pathogens.


Assuntos
Proteínas do Capsídeo/metabolismo , Capsídeo/ultraestrutura , Parvovirus/fisiologia , Receptores Virais/metabolismo , Ligação Viral , Adulto , Sequência de Aminoácidos , Animais , Criança , Microscopia Crioeletrônica , Humanos , Ácido N-Acetilneuramínico/metabolismo , Infecções por Parvoviridae/patologia , Parvovirus/genética , Polissacarídeos/metabolismo , Conformação Proteica , Análise de Sequência de DNA
4.
Viruses ; 12(3)2020 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-32204363

RESUMO

Ostrich diseases characterized by paralysis have been breaking out in broad areas of China since 2015, causing major damage to the ostrich breeding industry in China. This report describes a parvovirus detected in ostriches from four different regions. The entire genomes of four parvovirus strains were sequenced following amplification by PCR, and we conducted comprehensive analysis of the ostrich parvovirus genome. Results showed that the length genomes of the parvovirus contained two open reading frames. Ostrich parvovirus (OsPV) is a branch of goose parvovirus (GPV). Genetic distance analysis revealed a close relationship between the parvovirus and goose parvovirus strains from China, with the closest being the 2016 goose parvovirus RC16 strain from Chongqing. This is the first report of a parvovirus in ostriches. However, whether OsPV is the pathogen of ostrich paralysis remains uncertain. This study contributes new information about the evolution and epidemiology of parvovirus in China, which provides a new way for the study of paralysis in ostriches.


Assuntos
Evolução Molecular , Genoma Viral , Infecções por Parvoviridae/virologia , Parvovirus/fisiologia , Struthioniformes/virologia , Animais , Sequência de Bases , Testes Genéticos , Genômica/métodos , Infecções por Parvoviridae/diagnóstico , Filogenia , Reação em Cadeia da Polimerase
5.
Emerg Microbes Infect ; 9(1): 651-663, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32192415

RESUMO

Equine parvovirus-hepatitis (EqPV-H) has recently been associated with cases of Theiler's disease, a form of fulminant hepatic necrosis in horses. To assess whether EqPV-H is the cause of Theiler's disease, we first demonstrated hepatotropism by PCR on tissues from acutely infected horses. We then experimentally inoculated horses with EqPV-H and 8 of 10 horses developed hepatitis. One horse showed clinical signs of liver failure. The onset of hepatitis was temporally associated with seroconversion and a decline in viremia. Liver histology and in situ hybridization showed lymphocytic infiltrates and necrotic EqPV-H-infected hepatocytes. We next investigated potential modes of transmission. Iatrogenic transmission via allogeneic stem cell therapy for orthopedic injuries was previously suggested in a case series of Theiler's disease, and was demonstrated here for the first time. Vertical transmission and mechanical vectoring by horse fly bites could not be demonstrated in this study, potentially due to limited sample size. We found EqPV-H shedding in oral and nasal secretions, and in feces. Importantly, we could demonstrate EqPV-H transmission via oral inoculation with viremic serum. Together, our findings provide additional information that EqPV-H is the likely cause of Theiler's disease and that transmission of EqPV-H occurs via both iatrogenic and natural routes.


Assuntos
Hepatite Viral Animal/virologia , Doenças dos Cavalos/virologia , Fígado/virologia , Infecções por Parvoviridae/veterinária , Parvovirus/fisiologia , Animais , Dípteros/virologia , Fezes/virologia , Feminino , Hepatite Viral Animal/patologia , Hepatite Viral Animal/transmissão , Hepatócitos/patologia , Hepatócitos/virologia , Doenças dos Cavalos/patologia , Doenças dos Cavalos/transmissão , Cavalos , Transmissão Vertical de Doenças Infecciosas , Insetos Vetores/virologia , Fígado/patologia , Linfócitos , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/virologia , Boca/virologia , Necrose , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/transmissão , Infecções por Parvoviridae/virologia , Parvovirus/isolamento & purificação , Parvovirus/patogenicidade , Tropismo Viral , Viremia , Eliminação de Partículas Virais
6.
Viruses ; 11(11)2019 11 13.
Artigo em Inglês | MEDLINE | ID: mdl-31766142

RESUMO

The family Parvoviridae includes an ample and most diverse collection of viruses. Exploring the biological diversity and the inherent complexity in these apparently simple viruses has been a continuous commitment for the scientific community since their first discovery more than fifty years ago. The Special Issue of 'Viruses' dedicated to the 'New Insights into Parvovirus Research' aimed at presenting a 'state of the art' in many aspects of research in the field, at collecting the newest contributions on unresolved issues, and at presenting new approaches exploiting systemic (-omic) methodologies.


Assuntos
Infecções por Parvoviridae/virologia , Parvovirus/fisiologia , Pesquisa , Animais , Suscetibilidade a Doenças , Descoberta de Drogas , Humanos , Infecções por Parvoviridae/tratamento farmacológico , Relação Estrutura-Atividade
7.
J Virol ; 93(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30996096

RESUMO

Carnivore parvoviruses infect wild and domestic carnivores, and cross-species transmission is believed to occur. However, viral dynamics are not well understood, nor are the consequences for wild carnivore populations of the introduction of new strains into wild ecosystems. To clarify the ecology of these viruses in a multihost system such as the Serengeti ecosystem and identify potential threats for wildlife conservation, we analyzed, through real-time PCR, 152 samples belonging to 14 wild carnivore species and 62 samples from healthy domestic dogs. We detected parvovirus DNA in several wildlife tissues. Of the wild carnivore and domestic dog samples tested, 13% and 43%, respectively, were positive for carnivore parvovirus infection, but little evidence of transmission between the wild and domestic carnivores was detected. Instead, we describe two different epidemiological scenarios with separate routes of transmission: first, an endemic feline parvovirus (FPV) route of transmission maintained by wild carnivores inside the Serengeti National Park (SNP) and, second, a canine parvovirus (CPV) route of transmission among domestic dogs living around the periphery of the SNP. Twelve FPV sequences were characterized; new host-virus associations involving wild dogs, jackals, and hyenas were discovered; and our results suggest that mutations in the fragment of the vp2 gene were not required for infection of different carnivore species. In domestic dogs, 6 sequences belonged to the CPV-2a strain, while 11 belonged to the CPV-2 vaccine-derived strain. This is the first description of a vaccine-derived parvovirus strain being transmitted naturally.IMPORTANCE Carnivore parvoviruses are widespread among wild and domestic carnivores, which are vulnerable to severe disease under certain circumstances. This study furthers the understanding of carnivore parvovirus epidemiology, suggesting that feline parvoviruses are endemic in wild carnivores in the Serengeti National Park (SNP), with new host species identified, and that canine parvoviruses are present in the dog population living around the SNP. Little evidence of transmission of canine parvoviruses into wild carnivore species was found; however, the detection of vaccine-derived virus (described here for the first time to be circulating naturally in domestic dogs) highlights the importance of performing epidemiological research in the region.


Assuntos
Ecologia , Ecossistema , Especificidade de Hospedeiro , Infecções por Parvoviridae/virologia , Parvovirus/fisiologia , Vacinas , Animais , Animais Selvagens , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Gatos , Cães , Vírus da Panleucopenia Felina/genética , Vírus da Panleucopenia Felina/fisiologia , Epidemiologia Molecular , Mutação , Parvovirus/genética , Parvovirus/imunologia , Parvovirus Canino/genética , Parvovirus Canino/fisiologia , Filogenia , Análise de Sequência , Tanzânia
9.
J Endocrinol ; 238(1): 61-75, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29743341

RESUMO

The etiopathogenesis of type 1 diabetes (T1D) remains poorly understood. We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to better understand the role of the innate immune system in the mechanism of virus-induced disease. We observed that infection with KRV results in cell influx into visceral adipose tissue soon following infection prior to insulitis and hyperglycemia. In sharp contrast, subcutaneous adipose tissue is free of cellular infiltration, whereas ß cell inflammation and diabetes are observed beginning on day 14 post infection. Immunofluorescence studies further demonstrate that KRV triggers CD68+ macrophage recruitment and the expression of KRV transcripts and proinflammatory cytokines and chemokines in visceral adipose tissue. Adipocytes from naive rats cultured in the presence of KRV express virus transcripts and upregulate cytokine and chemokine gene expression. KRV induces apoptosis in visceral adipose tissue in vivo, which is reflected by positive TUNEL staining and the expression of cleaved caspase-3. Moreover, KRV leads to an oxidative stress response and downregulates the expression of adipokines and genes associated with mediating insulin signaling. Activation of innate immunity with Poly I:C in the absence of KRV leads to CD68+ macrophage recruitment to visceral adipose tissue and a decrease in adipokine expression detected 5 days following Poly (I:C) treatment. Finally, proof-of-principle studies show that brief anti-inflammatory steroid therapy suppresses visceral adipose tissue inflammation and protects from virus-induced disease. Our studies provide evidence raising the hypothesis that visceral adipose tissue inflammation and dysfunction may be involved in early mechanisms triggering ß cell autoimmunity.


Assuntos
Tecido Adiposo/patologia , Tecido Adiposo/fisiopatologia , Diabetes Mellitus Tipo 1/virologia , Inflamação/complicações , Paniculite/complicações , Parvovirus/fisiologia , Tecido Adiposo/imunologia , Tecido Adiposo/virologia , Animais , Células Cultivadas , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/fisiopatologia , Feminino , Imunidade Inata/fisiologia , Inflamação/patologia , Inflamação/virologia , Macrófagos/fisiologia , Masculino , Paniculite/imunologia , Paniculite/patologia , Paniculite/virologia , Parvovirus/imunologia , Ratos , Transdução de Sinais/imunologia
10.
Braz. j. microbiol ; 48(4): 769-773, Oct.-Dec. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-889183

RESUMO

ABSTRACT This is the first report on circulating canine rotavirus in Mexico. Fifty samples from dogs with gastroenteritis were analyzed used polymerase chain reaction and reverse transcription polymerase chain reaction in order to identify parvovirus and rotavirus, respectively; 7% of dogs were infected with rotavirus exclusively, while 14% were co-infected with both rotavirus and parvovirus; clinical signs in co-infected dogs were more severe.


Assuntos
Animais , Masculino , Feminino , Cães , Coinfecção/veterinária , Doenças do Cão/virologia , Gastroenterite/veterinária , Infecções por Parvoviridae/veterinária , Parvovirus/isolamento & purificação , Infecções por Rotavirus/veterinária , Rotavirus/isolamento & purificação , Coinfecção/virologia , Fezes/virologia , Gastroenterite/virologia , México , Infecções por Parvoviridae/virologia , Parvovirus/genética , Parvovirus/fisiologia , Infecções por Rotavirus/virologia , Rotavirus/genética , Rotavirus/fisiologia
11.
Vet Microbiol ; 210: 17-23, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29103688

RESUMO

Goose parvovirus (GPV) usually affects goslings and Muscovy ducks but not Pekin ducks. Earlier works showed that a variant GPV can cause short beak and dwarfism syndrome (SBDS) in Pekin ducks. Here, we investigated the pathogenicity of a variant GPV of Pekin duck-origin (JS1) and a classical GPV of goose-origin (H) in Pekin ducklings. Following intramuscular infection at two days of age, both JS1 and H strains influenced weight gain and development of beaks and bones of wings and legs, and caused microscopic lesions of internal organs of ducks. However, the clinical signs typical of SBDS could only be replicated with the JS1 isolate. The findings suggest that both variant and classical GPVs are pathogenic for Pekin ducklings, while the former is more virulent than the latter. Using a quantitative real-time PCR assay, high levels of viral load were detected from bloods, internal organs, leg muscles, and ileac contents in JS1- and H-infected ducks from 6h to 35days postinfection (DPI). Using a GPV VP3-based ELISA, antibodies in sera of JS1- and H-infected ducks were detectable at 1 DPI and then persistently rose during the subsequent five weeks. These results suggest that both variant and classical GPVs can infect Pekin ducklings. The present work contributes to the understanding of pathogenicity of GPV to Pekin ducks and may provide clues to pathogenesis of GPV-related SBDS.


Assuntos
Patos/virologia , Gansos/virologia , Infecções por Parvoviridae/veterinária , Parvovirus/patogenicidade , Doenças das Aves Domésticas/virologia , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Bico/patologia , Bico/virologia , Ensaio de Imunoadsorção Enzimática/veterinária , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/virologia , Parvovirus/genética , Parvovirus/fisiologia , Doenças das Aves Domésticas/patologia , Reação em Cadeia da Polimerase em Tempo Real/veterinária , Língua/patologia , Língua/virologia , Carga Viral/veterinária , Replicação Viral , Aumento de Peso , Asas de Animais/patologia , Asas de Animais/virologia
12.
Viruses ; 9(11)2017 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-29088070

RESUMO

Protoparvoviruses are simple single-stranded DNA viruses that infect many animal species. The protoparvovirus minute virus of mice (MVM) infects murine and transformed human cells provoking a sustained DNA damage response (DDR). This DDR is dependent on signaling by the ATM kinase and leads to a prolonged pre-mitotic cell cycle block that features the inactivation of ATR-kinase mediated signaling, proteasome-targeted degradation of p21, and inhibition of cyclin B1 expression. This review explores how protoparvoviruses, and specifically MVM, co-opt the common mechanisms regulating the DDR and cell cycle progression in order to prepare the host nuclear environment for productive infection.


Assuntos
Núcleo Celular/genética , Núcleo Celular/fisiologia , Dano ao DNA , Interações Hospedeiro-Patógeno , Vírus Miúdo do Camundongo/fisiologia , Parvovirus/fisiologia , Animais , Ciclo Celular , Linhagem Celular , Ciclina B1/metabolismo , Humanos , Camundongos , Mitose , Transdução de Sinais , Replicação Viral
13.
Viruses ; 9(11)2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-29072600

RESUMO

The Protoparvovirus (PtPV) genus of the Parvoviridae family of viruses includes important animal pathogens and reference molecular models for the entire family. Some virus members of the PtPV genus have arisen as promising tools to treat tumoral processes, as they exhibit marked oncotropism and oncolytic activities while being nonpathogenic for humans. The PtPVs invade and replicate within the nucleus making extensive use of the transport, transcription and replication machineries of the host cells. In order to reach the nucleus, PtPVs need to cross over several intracellular barriers and traffic through different cell compartments, which limit their infection efficiency. In this review we summarize molecular interactions, capsid structural transitions and hijacking of cellular processes, by which the PtPVs enter and deliver their single-stranded DNA genome into the host cell nucleus. Understanding mechanisms that govern the complex PtPV entry will be instrumental in developing approaches to boost their anticancer therapeutic potential and improving their safety profile.


Assuntos
Núcleo Celular/virologia , Parvovirus/fisiologia , Internalização do Vírus , Transporte Ativo do Núcleo Celular , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , DNA Viral/genética , DNA Viral/metabolismo , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Modelos Moleculares , Terapia Viral Oncolítica , Parvovirus/genética , Replicação Viral
14.
Viruses ; 9(10)2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28974036

RESUMO

Protoparvoviruses target the nucleus due to their dependence on the cellular reproduction machinery during the replication and expression of their single-stranded DNA genome. In recent years, our understanding of the multistep process of the capsid nuclear import has improved, and led to the discovery of unique viral nuclear entry strategies. Preceded by endosomal transport, endosomal escape and microtubule-mediated movement to the vicinity of the nuclear envelope, the protoparvoviruses interact with the nuclear pore complexes. The capsids are transported actively across the nuclear pore complexes using nuclear import receptors. The nuclear import is sometimes accompanied by structural changes in the nuclear envelope, and is completed by intranuclear disassembly of capsids and chromatinization of the viral genome. This review discusses the nuclear import strategies of protoparvoviruses and describes its dynamics comprising active and passive movement, and directed and diffusive motion of capsids in the molecularly crowded environment of the cell.


Assuntos
Transporte Ativo do Núcleo Celular , Núcleo Celular/virologia , Parvovirus/fisiologia , Internalização do Vírus , Animais , Capsídeo/metabolismo , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Genoma Viral , Humanos , Carioferinas/genética , Carioferinas/metabolismo , Camundongos , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Membrana Nuclear/virologia , Poro Nuclear/metabolismo , Parvovirus/genética , Parvovirus/metabolismo , Replicação Viral
15.
Phys Rev E ; 95(6-1): 062402, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28709270

RESUMO

We present a Landau theory for large-l orientational phase transitions and apply it to the assembly of icosahedral viral capsids. The theory predicts two distinct types of ordering transitions. Transitions dominated by the l=6,10,12, and 18 icosahedral spherical harmonics resemble robust first-order phase transitions that are not significantly affected by chirality. The remaining transitions depend essentially on including mixed l states denoted as l=15+16 corresponding to a mixture of l=15 and l=16 spherical harmonics. The l=15+16 transition is either continuous or weakly first-order and it is strongly influenced by chirality, which suppresses spontaneous chiral symmetry breaking. The icosahedral state is in close competition with states that have tetrahedral, D_{5}, and octahedral symmetries. We present a group-theoretic method to analyze the competition between the different symmetries. The theory is applied to a variety of viral shells.


Assuntos
Capsídeo , Modelos Biológicos , Montagem de Vírus , Bacteriófagos/fisiologia , Bromovirus/fisiologia , Capsídeo/metabolismo , Vírus da Dengue/fisiologia , Modelos Moleculares , Parvovirus/fisiologia , Picornaviridae/fisiologia , Montagem de Vírus/fisiologia
16.
Braz J Microbiol ; 48(4): 769-773, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28716388

RESUMO

This is the first report on circulating canine rotavirus in Mexico. Fifty samples from dogs with gastroenteritis were analyzed used polymerase chain reaction and reverse transcription polymerase chain reaction in order to identify parvovirus and rotavirus, respectively; 7% of dogs were infected with rotavirus exclusively, while 14% were co-infected with both rotavirus and parvovirus; clinical signs in co-infected dogs were more severe.


Assuntos
Coinfecção/veterinária , Doenças do Cão/virologia , Gastroenterite/veterinária , Infecções por Parvoviridae/veterinária , Parvovirus/isolamento & purificação , Infecções por Rotavirus/veterinária , Rotavirus/isolamento & purificação , Animais , Coinfecção/virologia , Cães , Fezes/virologia , Feminino , Gastroenterite/virologia , Masculino , México , Infecções por Parvoviridae/virologia , Parvovirus/genética , Parvovirus/fisiologia , Rotavirus/genética , Rotavirus/fisiologia , Infecções por Rotavirus/virologia
17.
Neurotherapeutics ; 14(2): 333-344, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28265902

RESUMO

Malignant glioma is the most common primary brain tumor and carries a grim prognosis, with a median survival of just over 14 months. Given the poor outcomes with standard-of-care treatments, novel treatment strategies are needed. The concept of virotherapy for the treatment of malignant tumors dates back more than a century and can be divided into replication-competent oncolytic viruses and replication-deficient viral vectors. Oncolytic viruses are designed to selectively target, infect, and replicate in tumor cells, while sparing surrounding normal brain. A host of oncolytic viruses has been evaluated in early phase human trials with promising safety results, but none has progressed to phase III trials. Despite the 25 years that has passed since the initial publication of genetically engineered oncolytic viruses for the treatment of glioma, much remains to be learned about the use of this therapy, including its mechanism of action, optimal treatment paradigm, appropriate targets, and integration with adjuvant agents. Oncolytic viral therapy for glioma remains promising and will undoubtedly impact the future of patient care.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Terapia Viral Oncolítica , Adenoviridae/fisiologia , Animais , Neoplasias Encefálicas/virologia , Ensaios Clínicos como Assunto , Vetores Genéticos , Glioma/virologia , Humanos , Vírus Oncolíticos/fisiologia , Paramyxoviridae/fisiologia , Parvovirus/fisiologia , Poliovirus/fisiologia , Reoviridae/fisiologia , Resultado do Tratamento , Replicação Viral
18.
J Virol ; 91(2)2017 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-27847360

RESUMO

Parvovirus capsids are small but complex molecular machines responsible for undertaking many of the steps of cell infection, genome packing, and cell-to-cell as well as host-to-host transfer. The details of parvovirus infection of cells are still not fully understood, but the processes must involve small changes in the capsid structure that allow the endocytosed virus to escape from the endosome, pass through the cell cytoplasm, and deliver the single-stranded DNA (ssDNA) genome to the nucleus, where viral replication occurs. Here, we examine capsid substitutions that eliminate canine parvovirus (CPV) infectivity and identify how those mutations changed the capsid structure or altered interactions with the infectious pathway. Amino acid substitutions on the exterior surface of the capsid (Gly299Lys/Ala300Lys) altered the binding of the capsid to transferrin receptor type 1 (TfR), particularly during virus dissociation from the receptor, but still allowed efficient entry into both feline and canine cells without successful infection. These substitutions likely control specific capsid structural changes resulting from TfR binding required for infection. A second set of changes on the interior surface of the capsid reduced viral infectivity by >100-fold and included two cysteine residues and neighboring residues. One of these substitutions, Cys270Ser, modulates a VP2 cleavage event found in ∼10% of the capsid proteins that also was shown to alter capsid stability. A neighboring substitution, Pro272Lys, significantly reduced capsid assembly, while a Cys273Ser change appeared to alter capsid transport from the nucleus. These mutants reveal additional structural details that explain cell infection processes of parvovirus capsids. IMPORTANCE: Parvoviruses are commonly found in both vertebrate and invertebrate animals and cause widespread disease. They are also being developed as oncolytic therapeutics and as gene therapy vectors. Most functions involved in infection or transduction are mediated by the viral capsid, but the structure-function correlates of the capsids and their constituent proteins are still incompletely understood, especially in relation to identifying capsid processes responsible for infection and release from the cell. Here, we characterize the functional effects of capsid protein mutations that result in the loss of virus infectivity, giving a better understanding of the portions of the capsid that mediate essential steps in successful infection pathways and how they contribute to viral infectivity.


Assuntos
Proteínas do Capsídeo/química , Proteínas do Capsídeo/metabolismo , Infecções por Parvoviridae/virologia , Parvovirus/fisiologia , Conformação Proteica , Sequência de Aminoácidos , Proteínas do Capsídeo/genética , Endopeptidases/metabolismo , Interações Hospedeiro-Patógeno , Modelos Moleculares , Mutação , Transporte Proteico , Proteólise , Receptores Virais/metabolismo , Relação Estrutura-Atividade , Ligação Viral
19.
Diabetes ; 66(1): 145-157, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27999109

RESUMO

The pathogenesis of human type 1 diabetes, characterized by immune-mediated damage of insulin-producing ß-cells of pancreatic islets, may involve viral infection. Essential components of the innate immune antiviral response, including type I interferon (IFN) and IFN receptor-mediated signaling pathways, are candidates for determining susceptibility to human type 1 diabetes. Numerous aspects of human type 1 diabetes pathogenesis are recapitulated in the LEW.1WR1 rat model. Diabetes can be induced in LEW.1WR1 weanling rats challenged with virus or with the viral mimetic polyinosinic:polycytidylic acid (poly I:C). We hypothesized that disrupting the cognate type I IFN receptor (type I IFN α/ß receptor [IFNAR]) to interrupt IFN signaling would prevent or delay the development of virus-induced diabetes. We generated IFNAR1 subunit-deficient LEW.1WR1 rats using CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) genome editing and confirmed functional disruption of the Ifnar1 gene. IFNAR1 deficiency significantly delayed the onset and frequency of diabetes and greatly reduced the intensity of insulitis after poly I:C treatment. The occurrence of Kilham rat virus-induced diabetes was also diminished in IFNAR1-deficient animals. These findings firmly establish that alterations in innate immunity influence the course of autoimmune diabetes and support the use of targeted strategies to limit or prevent the development of type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Animais , Sistemas CRISPR-Cas/genética , Sistemas CRISPR-Cas/fisiologia , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/virologia , Feminino , Imunidade Inata/genética , Imunidade Inata/fisiologia , Interferon Tipo I/metabolismo , Masculino , Parvovirus/genética , Parvovirus/fisiologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Interferon alfa e beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Virus Res ; 222: 24-28, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27212684

RESUMO

Virulent mink enteritis parvovirus (MEV) strain MEV-LHV replicated to higher titers in feline F81 cells than attenuated strain MEV-L. Phylogenetic and sequence analyses of the VP2 gene of MEV-LHV, MEV-L and other strains in GenBank revealed two evolutionary branches separating virulent and attenuated strains. Three residues, 101, 232 and 411, differed between virulent and attenuated strains but were conserved within the two branches. Site-directed mutagenesis of the VP2 gene of infectious plasmids of attenuated strain MEV-L respectively replacing residues 101 Ile and 411 Ala with Thr and Glu of virulent strains (MEV-L I101T and MEV-L A411E) increased replication efficiency but still to lower levels than MEV-LHV. However, viruses with mutation of residue 232 (MEV-L I232V and MEV-L I101T/I232V/A411E) decreased viral transcription and replication levels. The three VP2 residues 101, 232 and 411, located on or near the capsid surface, played different roles in the infection processes of MEV.


Assuntos
Aminoácidos/genética , Proteínas do Capsídeo/genética , Parvovirus/fisiologia , Replicação Viral , Substituição de Aminoácidos , Animais , Proteínas do Capsídeo/química , Códon , Vison , Mutação , Infecções por Parvoviridae , Parvovirus/classificação , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...