Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Viruses ; 12(6)2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-32560452

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Cápside/ultraestructura , Parvovirus/fisiología , Receptores Virales/metabolismo , Acoplamiento Viral , Adulto , Secuencia de Aminoácidos , Animales , Niño , Microscopía por Crioelectrón , Humanos , Ácido N-Acetilneuramínico/metabolismo , Infecciones por Parvoviridae/patología , Parvovirus/genética , Polisacáridos/metabolismo , Conformación Proteica , Análisis de Secuencia de ADN
2.
Emerg Microbes Infect ; 9(1): 651-663, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32192415

RESUMEN

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.


Asunto(s)
Hepatitis Viral Animal/virología , Enfermedades de los Caballos/virología , Hígado/virología , Infecciones por Parvoviridae/veterinaria , Parvovirus/fisiología , Animales , Dípteros/virología , Heces/virología , Femenino , Hepatitis Viral Animal/patología , Hepatitis Viral Animal/transmisión , Hepatocitos/patología , Hepatocitos/virología , Enfermedades de los Caballos/patología , Enfermedades de los Caballos/transmisión , Caballos , Transmisión Vertical de Enfermedad Infecciosa , Insectos Vectores/virología , Hígado/patología , Linfocitos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/virología , Boca/virología , Necrosis , Infecciones por Parvoviridae/patología , Infecciones por Parvoviridae/transmisión , Infecciones por Parvoviridae/virología , Parvovirus/aislamiento & purificación , Parvovirus/patogenicidad , Tropismo Viral , Viremia , Esparcimiento de Virus
3.
Viruses ; 11(11)2019 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766142

RESUMEN

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.


Asunto(s)
Infecciones por Parvoviridae/virología , Parvovirus/fisiología , Investigación , Animales , Susceptibilidad a Enfermedades , Descubrimiento de Drogas , Humanos , Infecciones por Parvoviridae/tratamiento farmacológico , Relación Estructura-Actividad
4.
J Endocrinol ; 238(1): 61-75, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29743341

RESUMEN

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.


Asunto(s)
Tejido Adiposo/patología , Tejido Adiposo/fisiopatología , Diabetes Mellitus Tipo 1/virología , Inflamación/complicaciones , Paniculitis/complicaciones , Parvovirus/fisiología , Tejido Adiposo/inmunología , Tejido Adiposo/virología , Animales , Células Cultivadas , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Femenino , Inmunidad Innata/fisiología , Inflamación/patología , Inflamación/virología , Macrófagos/fisiología , Masculino , Paniculitis/inmunología , Paniculitis/patología , Paniculitis/virología , Parvovirus/inmunología , Ratas , Transducción de Señal/inmunología
5.
Braz. j. microbiol ; 48(4): 769-773, Oct.-Dec. 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-889183

RESUMEN

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.


Asunto(s)
Animales , Masculino , Femenino , Perros , Coinfección/veterinaria , Enfermedades de los Perros/virología , Gastroenteritis/veterinaria , Infecciones por Parvoviridae/veterinaria , Parvovirus/aislamiento & purificación , Infecciones por Rotavirus/veterinaria , Rotavirus/aislamiento & purificación , Coinfección/virología , Heces/virología , Gastroenteritis/virología , México , Infecciones por Parvoviridae/virología , Parvovirus/genética , Parvovirus/fisiología , Infecciones por Rotavirus/virología , Rotavirus/genética , Rotavirus/fisiología
6.
Viruses ; 9(11)2017 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-29072600

RESUMEN

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.


Asunto(s)
Núcleo Celular/virología , Parvovirus/fisiología , Internalización del Virus , Transporte Activo de Núcleo Celular , Animales , Cápside/metabolismo , Proteínas de la Cápside/genética , ADN Viral/genética , ADN Viral/metabolismo , Genoma Viral , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Viroterapia Oncolítica , Parvovirus/genética , Replicación Viral
7.
Neurotherapeutics ; 14(2): 333-344, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28265902

RESUMEN

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.


Asunto(s)
Neoplasias Encefálicas/terapia , Glioma/terapia , Viroterapia Oncolítica , Adenoviridae/fisiología , Animales , Neoplasias Encefálicas/virología , Ensayos Clínicos como Asunto , Vectores Genéticos , Glioma/virología , Humanos , Virus Oncolíticos/fisiología , Paramyxoviridae/fisiología , Parvovirus/fisiología , Poliovirus/fisiología , Reoviridae/fisiología , Resultado del Tratamiento , Replicación Viral
8.
J Virol ; 91(2)2017 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-27847360

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Infecciones por Parvoviridae/virología , Parvovirus/fisiología , Conformación Proteica , Secuencia de Aminoácidos , Proteínas de la Cápside/genética , Endopeptidasas/metabolismo , Interacciones Huésped-Patógeno , Modelos Moleculares , Mutación , Transporte de Proteínas , Proteolisis , Receptores Virales/metabolismo , Relación Estructura-Actividad , Acoplamiento Viral
9.
Curr Opin Virol ; 12: 59-65, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25846849

RESUMEN

The nuclear import of viral genomes is an important step of the infectious cycle for viruses that replicate in the nucleus of their host cells. Although most viruses use the cellular nuclear import machinery or some components of this machinery, others have developed sophisticated ways to reach the nucleus. Some of these have been known for some time; however, recent studies have changed our understanding of how some non-enveloped DNA viruses access the nucleus. For example, parvoviruses enter the nucleus through small disruptions of the nuclear membranes and nuclear lamina, and adenovirus tugs at the nuclear pore complex, using kinesin-1, to disassemble their capsids and deliver viral proteins and genomes into the nucleus. Here we review recent findings of the nuclear import strategies of three small non-enveloped DNA viruses, including adenovirus, parvovirus, and the polyomavirus simian virus 40.


Asunto(s)
Núcleo Celular/metabolismo , Núcleo Celular/virología , Virus ADN/fisiología , ADN Viral/metabolismo , Poro Nuclear/virología , Transporte Activo de Núcleo Celular , Adenoviridae/fisiología , Virus ADN/genética , Genoma Viral , Humanos , Parvovirus/fisiología , Virus 40 de los Simios/fisiología , Internalización del Virus , Replicación Viral
10.
Virol J ; 12: 6, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25630937

RESUMEN

Accumulated evidence gathered over recent decades demonstrated that some members of the Parvoviridae family, in particular the rodent protoparvoviruses H-1PV, the minute virus of mice and LuIII have natural anticancer activity while being nonpathogenic to humans. These studies have laid the foundations for the launch of a first phase I/IIa clinical trial, in which the rat H-1 parvovirus is presently undergoing evaluation for its safety and first signs of efficacy in patients with glioblastoma multiforme. After a brief overview of the biology of parvoviruses, this review focuses on the studies which unraveled the antineoplastic properties of these agents and supported their clinical use as anticancer therapeutics. Furthermore, the development of novel parvovirus-based anticancer strategies with enhanced specificity and efficacy is discussed, in particular the development of second and third generation vectors and the combination of parvoviruses with other anticancer agents. Lastly, we address the key challenges that remain towards a more rational and efficient use of oncolytic parvoviruses in clinical settings, and discuss how a better understanding of the virus life-cycle and of the cellular factors involved in virus infection, replication and cytotoxicity may promote the further development of parvovirus-based anticancer therapies, open new prospects for treatment and hopefully improve clinical outcome.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Parvovirus/fisiología , Animales , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos
11.
Adv Virus Res ; 89: 85-139, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24751195

RESUMEN

As shrimp aquaculture has evolved from a subsistent farming activity to an economically important global industry, viral diseases have also become a serious threat to the sustainable growth and productivity of this industry. Parvoviruses represent an economically important group of viruses that has greatly affected shrimp aquaculture. In the early 1980s, an outbreak of a shrimp parvovirus, infectious hypodermal and hematopoietic necrosis virus (IHHNV), led to the collapse of penaeid shrimp farming in the Americas. Since then, considerable progress has been made in characterizing the parvoviruses of shrimp and developing diagnostic methods aimed to preventing the spread of diseases caused by these viruses. To date, four parvoviruses are known that infect shrimp; these include IHHNV, hepatopancreatic parvovirus (HPV), spawner-isolated mortality virus (SMV), and lymphoid organ parvo-like virus. Due to the economic repercussions that IHHNV and HPV outbreaks have caused to shrimp farming over the years, studies have been focused mostly on these two pathogens, while information on SMV and LPV remains limited. IHHNV was the first shrimp virus to be sequenced and the first for which highly sensitive diagnostic methods were developed. IHHNV-resistant lines of shrimp were also developed to mitigate the losses caused by this virus. While the losses due to IHHNV have been largely contained in recent years, reports of HPV-induced mortalities in larval stages in hatchery and losses due to reduced growth have increased. This review presents a comprehensive account of the history and current knowledge on the biology, diagnostics methods, genomic features, mechanisms of evolution, and management strategies of shrimp parvoviruses. We also highlighted areas where research efforts should be focused in order to gain further insight on the mechanisms of parvoviral pathogenicity in shrimp that will help to prevent future losses caused by these viruses.


Asunto(s)
Organismos Acuáticos/virología , Orden Génico , Genoma Viral , Parvovirus/genética , Parvovirus/fisiología , Penaeidae/virología , Animales , Acuicultura , Evolución Molecular
12.
Virology ; 447(1-2): 221-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24210118

RESUMEN

Anti-angiogenic therapy has been recognized as a powerful potential strategy for impeding the growth of various tumors. However no major therapeutic effects have been observed to date, mainly because of the emergence of several resistance mechanisms. Among novel strategies to target tumor vasculature, some oncolytic viruses open up new prospects. In this context, we addressed the question whether the rodent parvovirus H-1PV can target endothelial cells. We show that cultures of human normal (HUVEC) and immortalized (KS-IMM) endothelial cells sustain an abortive viral cycle upon infection with H-1PV and are sensitive to H-1PV cytotoxicity. H-1PV significantly inhibits infected KS-IMM tumor growth. This effect may be traced back by the virus ability to both kill proliferating endothelial cells and inhibit VEGF production Recombinant H-1PV vectors can also transduce tumor cells with chemokines endowed with anti-angiogenesis properties, and warrant further validation for the treatment of highly vascularized tumors.


Asunto(s)
Quimiocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Neovascularización Patológica/prevención & control , Parvovirus/fisiología , Animales , Terapia Biológica/métodos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Ratones , Neoplasias/terapia , Análisis de Supervivencia , Resultado del Tratamiento
13.
EMBO Mol Med ; 5(10): 1537-55, 2013 10.
Artículo en Inglés | MEDLINE | ID: mdl-24092664

RESUMEN

The rat parvovirus H-1PV has oncolytic and tumour-suppressive properties potentially exploitable in cancer therapy. This possibility is being explored and results are encouraging, but it is necessary to improve the oncotoxicity of the virus. Here we show that this can be achieved by co-treating cancer cells with H-1PV and histone deacetylase inhibitors (HDACIs) such as valproic acid (VPA). We demonstrate that these agents act synergistically to kill a range of human cervical carcinoma and pancreatic carcinoma cell lines by inducing oxidative stress, DNA damage and apoptosis. Strikingly, in rat and mouse xenograft models, H-1PV/VPA co-treatment strongly inhibits tumour growth promoting complete tumour remission in all co-treated animals. At the molecular level, we found acetylation of the parvovirus nonstructural protein NS1 at residues K85 and K257 to modulate NS1-mediated transcription and cytotoxicity, both of which are enhanced by VPA treatment. These results warrant clinical evaluation of H-1PV/VPA co-treatment against cervical and pancreatic ductal carcinomas.


Asunto(s)
Carcinoma/terapia , Virus Oncolíticos/fisiología , Parvovirus/fisiología , Ácido Valproico/farmacología , Animales , Apoptosis/efectos de los fármacos , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Células HeLa , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Estrés Oxidativo/efectos de los fármacos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Parvovirus/metabolismo , Ratas , Ratas Desnudas , Neoplasias del Cuello Uterino/tratamiento farmacológico , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Ácido Valproico/uso terapéutico
14.
J Virol ; 87(24): 13816-24, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24109231

RESUMEN

Parvoviruses are rapidly evolving viruses that infect a wide range of hosts, including vertebrates and invertebrates. Extensive methylation of the parvovirus genome has been recently demonstrated. A global pattern of methylation of CpG dinucleotides is seen in vertebrate genomes, compared to "fractional" methylation patterns in invertebrate genomes. It remains unknown if the loss of CpG dinucleotides occurs in all viruses of a given DNA virus family that infect host species spanning across vertebrates and invertebrates. We investigated the link between the extent of CpG dinucleotide depletion among autonomous parvoviruses and the evolutionary lineage of the infected host. We demonstrate major differences in the relative abundance of CpG dinucleotides among autonomous parvoviruses which share similar genome organization and common ancestry, depending on the infected host species. Parvoviruses infecting vertebrate hosts had significantly lower relative abundance of CpG dinucleotides than parvoviruses infecting invertebrate hosts. The strong correlation of CpG dinucleotide depletion with the gain in TpG/CpA dinucleotides and the loss of TpA dinucleotides among parvoviruses suggests a major role for CpG methylation in the evolution of parvoviruses. Our data present evidence that links the relative abundance of CpG dinucleotides in parvoviruses to the methylation capabilities of the infected host. In sum, our findings support a novel perspective of host-driven evolution among autonomous parvoviruses.


Asunto(s)
Fosfatos de Dinucleósidos/genética , Evolución Molecular , Interacciones Huésped-Patógeno , Invertebrados/virología , Infecciones por Parvoviridae/veterinaria , Infecciones por Parvoviridae/virología , Parvovirus/genética , Vertebrados/virología , Animales , Metilación de ADN , Fosfatos de Dinucleósidos/metabolismo , Genoma Viral , Humanos , Invertebrados/genética , Invertebrados/metabolismo , Metilación , Datos de Secuencia Molecular , Infecciones por Parvoviridae/genética , Infecciones por Parvoviridae/metabolismo , Parvovirus/clasificación , Parvovirus/fisiología , Filogenia , Vertebrados/genética , Vertebrados/metabolismo
15.
PLoS Pathog ; 9(9): e1003605, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068925

RESUMEN

Progeny particles of non-enveloped lytic parvoviruses were previously shown to be actively transported to the cell periphery through vesicles in a gelsolin-dependent manner. This process involves rearrangement and destruction of actin filaments, while microtubules become protected throughout the infection. Here the focus is on the intracellular egress pathway, as well as its impact on the properties and release of progeny virions. By colocalization with cellular marker proteins and specific modulation of the pathways through over-expression of variant effector genes transduced by recombinant adeno-associated virus vectors, we show that progeny PV particles become engulfed into COPII-vesicles in the endoplasmic reticulum (ER) and are transported through the Golgi to the plasma membrane. Besides known factors like sar1, sec24, rab1, the ERM family proteins, radixin and moesin play (an) essential role(s) in the formation/loading and targeting of virus-containing COPII-vesicles. These proteins also contribute to the transport through ER and Golgi of the well described analogue of cellular proteins, the secreted Gaussia luciferase in absence of virus infection. It is therefore likely that radixin and moesin also serve for a more general function in cellular exocytosis. Finally, parvovirus egress via ER and Golgi appears to be necessary for virions to gain full infectivity through post-assembly modifications (e.g. phosphorylation). While not being absolutely required for cytolysis and progeny virus release, vesicular transport of parvoviruses through ER and Golgi significantly accelerates these processes pointing to a regulatory role of this transport pathway.


Asunto(s)
Linfocitos B/virología , Retículo Endoplásmico/virología , Aparato de Golgi/virología , Interacciones Huésped-Patógeno , Parvovirus/fisiología , Virión/fisiología , Ensamble de Virus , Sustitución de Aminoácidos , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/ultraestructura , Transporte Biológico , Proteínas de la Cápside/metabolismo , Efecto Citopatogénico Viral , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Hibridomas , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Mutación , Infecciones por Parvoviridae/inmunología , Infecciones por Parvoviridae/metabolismo , Infecciones por Parvoviridae/patología , Infecciones por Parvoviridae/virología , Parvovirus/inmunología , Parvovirus/ultraestructura , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo , Virión/inmunología , Virión/ultraestructura , Liberación del Virus
16.
Subcell Biochem ; 68: 307-28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23737056

RESUMEN

Icosahedral viruses exhibit elegant pathways of capsid assembly and maturation regulated by symmetry principles. Assembly is a dynamic process driven by consecutive and genetically programmed morphogenetic interactions between protein subunits. The non-symmetric capsid subunits are gathered by hydrophobic contacts and non-covalent interactions in assembly intermediates, which serve as blocks to build a symmetric capsid. In some cases, non-symmetric interactions among intermediates are involved in assembly, highlighting the remarkable capacity of capsid proteins to fold into demanding conformations compatible with a closed protein shell. In this chapter, the morphogenesis of structurally simple icosahedral viruses, including representative members of the parvoviruses, picornaviruses or polyomaviruses as paradigms, is described in some detail. Icosahedral virus assembly may occur in different subcellular compartments and involve a panoplia of cellular and viral factors, chaperones, and protein modifications that, in general, are still poorly characterized. Mechanisms of viral genome encapsidation may imply direct interactions between the genome and the assembly intermediates, or active packaging into a preformed empty capsid. High stability of intermediates and proteolytic cleavages during viral maturation usually contribute to the overall irreversible character of the assembly process. These and other simple icosahedral viruses were pioneer models to understand basic principles of virus assembly, continue to be leading subjects of morphogenetic analyses, and have inspired ongoing studies on the assembly of larger viruses and cellular and synthetic macromolecular complexes.


Asunto(s)
Parvovirus/fisiología , Picornaviridae/fisiología , Poliomavirus/fisiología , Ensamble de Virus , Animales , Humanos
17.
Vestn Ross Akad Med Nauk ; (12): 4-8, 2013.
Artículo en Ruso | MEDLINE | ID: mdl-24741936

RESUMEN

UNLABELLED: Currently one of the most promising approaches in development of cancer virotherapy is based on the ability of oncolytic viruses to selective infection and lysis of tumor cells. AIM: The goal of the study was to identify and evaluate perspective oncolytic viruses capable of selectively destroying human glioma cells. PATIENTS AND METHODS: Original GB2m, GA14m and GB22m glioma cell cultures derived from patients were used for evaluating in vitro oncolytic activity of some typical orthopoxviruses, adenoviruses and parvoviruses. RESULTS: The oncolytic activity in the human glioma cell models was confirmed for LIVP and WR strains of vaccinia virus, Adel2 and Ad2del strains with deletions within E1B/55K gene and derived from human adenoviruses type 2 and 5, respectively. CONCLUSIONS: We consider these oncolytic viruses as promising agents for the treatment of human malignant glioma.


Asunto(s)
Glioma , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/fisiología , Técnicas de Cultivo de Célula , Glioma/terapia , Glioma/virología , Humanos , Orthopoxvirus/fisiología , Parvovirus/fisiología , Células Tumorales Cultivadas/virología , Fenómenos Fisiológicos de los Virus
18.
J Virol ; 86(13): 7280-91, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22553327

RESUMEN

Because productive infection by parvoviruses requires cell division and is enhanced by oncogenic transformation, some parvoviruses may have potential utility in killing cancer cells. To identify the parvovirus(es) with the optimal oncolytic effect against human glioblastomas, we screened 12 parvoviruses at a high multiplicity of infection (MOI). MVMi, MVMc, MVM-G17, tumor virus X (TVX), canine parvovirus (CPV), porcine parvovirus (PPV), rat parvovirus 1A (RPV1A), and H-3 were relatively ineffective. The four viruses with the greatest oncolytic activity, LuIII, H-1, MVMp, and MVM-G52, were tested for the ability, at a low MOI, to progressively infect the culture over time, causing cell death at a rate higher than that of cell proliferation. LuIII alone was effective in all five human glioblastomas tested. H-1 progressively infected only two of five; MVMp and MVM-G52 were ineffective in all five. To investigate the underlying mechanism of LuIII's phenotype, we used recombinant parvoviruses with the LuIII capsid replacing the MVMp capsid or with molecular alteration of the P4 promoter. The LuIII capsid enhanced efficient replication and oncolysis in MO59J gliomas cells; other gliomas tested required the entire LuIII genome to exhibit enhanced infection. LuIII selectively infected glioma cells over normal glial cells in vitro. In mouse models, human glioblastoma xenografts were selectively infected by LuIII when administered intratumorally; LuIII reduced tumor growth by 75%. LuIII also had the capacity to selectively infect subcutaneous or intracranial gliomas after intravenous inoculation. Intravenous or intracranial LuIII caused no adverse effects. Intracranial LuIII caused no infection of mature mouse neurons or glia in vivo but showed a modest infection of developing neurons.


Asunto(s)
Muerte Celular , Glioblastoma/virología , Especificidad del Huésped , Neuroglía/virología , Virus Oncolíticos/fisiología , Parvovirus/fisiología , Replicación Viral , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Ratones , Viroterapia Oncolítica/efectos adversos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/crecimiento & desarrollo , Virus Oncolíticos/patogenicidad , Parvovirus/crecimiento & desarrollo , Parvovirus/patogenicidad , Resultado del Tratamiento
19.
Clin Cancer Res ; 18(13): 3516-23, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22566376

RESUMEN

Rodent parvoviruses (PV) are recognized for their intrinsic oncotropism and oncolytic activity, which contribute to their natural oncosuppressive effects. Although PV uptake occurs in most host cells, some of the subsequent steps leading to expression and amplification of the viral genome and production of progeny particles are upregulated in malignantly transformed cells. By usurping cellular processes such as DNA replication, DNA damage response, and gene expression, and/or by interfering with cellular signaling cascades involved in cytoskeleton dynamics, vesicular integrity, cell survival, and death, PVs can induce cytostasis and cytotoxicity. Although productive PV infections normally culminate in cytolysis, virus spread to neighboring cells and secondary rounds of infection, even abortive infection or the sole expression of the PV nonstructural protein NS1, is sufficient to cause significant tumor cell death, either directly or indirectly (through activation of host immune responses). This review highlights the molecular pathways involved in tumor cell targeting by PVs and in PV-induced cell death. It concludes with a discussion of the relevance of these pathways to the application of PVs in cancer therapy, linking basic knowledge of PV-host cell interactions to preclinical assessment of PV oncosuppression.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Parvovirus/fisiología , Animales , Ensayos Clínicos como Asunto , Interacciones Huésped-Patógeno , Humanos , Tropismo Viral
20.
J Virol ; 86(7): 3452-65, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22258256

RESUMEN

The rat parvovirus H-1PV is a promising anticancer agent given its oncosuppressive properties and the absence of known side effects in humans. H-1PV replicates preferentially in transformed cells, but the virus can enter both normal and cancer cells. Uptake by normal cells sequesters a significant portion of the administered viral dose away from the tumor target. Hence, targeting H-1PV entry specifically to tumor cells is important to increase the efficacy of parvovirus-based treatments. In this study, we first found that sialic acid plays a key role in H-1PV entry. We then genetically engineered the H-1PV capsid to improve its affinity for human tumor cells. By analogy with the resolved crystal structure of the closely related parvovirus minute virus of mice, we developed an in silico three-dimensional (3D) model of the H-1PV wild-type capsid. Based on this model, we identified putative amino acids involved in cell membrane recognition and virus entry at the level of the 2-fold axis of symmetry of the capsid, within the so-called dimple region. In situ mutagenesis of these residues significantly reduced the binding and entry of H-1PV into permissive cells. We then engineered an entry-deficient viral capsid and inserted a cyclic RGD-4C peptide at the level of its 3-fold axis spike. This peptide binds α(v)ß(3) and α(v)ß(5) integrins, which are overexpressed in cancer cells and growing blood vessels. The insertion of the peptide rescued viral infectivity toward cells overexpressing α(v)ß(5) integrins, resulting in the efficient killing of these cells by the reengineered virus. This work demonstrates that H-1PV can be genetically retargeted through the modification of its capsid, showing great promise for a more efficient use of this virus in cancer therapy.


Asunto(s)
Proteínas de la Cápside/genética , Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/genética , Parvovirus/genética , Animales , Células CHO , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Cricetinae , Ingeniería Genética , Humanos , Modelos Moleculares , Neoplasias/virología , Virus Oncolíticos/química , Virus Oncolíticos/fisiología , Infecciones por Parvoviridae/virología , Parvovirus/química , Parvovirus/fisiología , Ratas , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA