Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 172
Filtrar
1.
Viruses ; 13(9)2021 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-34578389

RESUMO

Zika virus (ZIKV) infection during pregnancy causes a wide spectrum of congenital abnormalities and postnatal developmental sequelae such as fetal loss, intrauterine growth restriction (IUGR), microcephaly, or motor and neurodevelopmental disorders. Here, we investigated whether a mouse pregnancy model recapitulated a wide range of symptoms after congenital ZIKV infection, and whether the embryonic age of congenital infection changed the fetal or postnatal outcomes. Infection with ZIKV strain PRVABC59 from embryonic day 6.5 (E6.5) to E8.5, corresponding to the mid-first trimester in humans, caused fetal death, fetal resorption, or severe IUGR, whereas infection from E9.5 to E14.5, corresponding to the late-first to second trimester in humans, caused stillbirth, neonatal death, microcephaly, and postnatal growth deficiency. Furthermore, 4-week-old offspring born to dams infected at E12.5 showed abnormalities in neuropsychiatric state, motor behavior, autonomic function, or reflex and sensory function. Thus, our model recapitulated the multiple symptoms seen in human cases, and the embryonic age of congenital infection was one of the determinant factors of offspring outcomes in mice. Furthermore, maternal neutralizing antibodies protected the offspring from neonatal death after congenital infection at E9.5, suggesting that neonatal death in our model could serve as criteria for screening of vaccine candidates.


Assuntos
Feto/virologia , Microcefalia/virologia , Malformações do Sistema Nervoso/virologia , Infecção por Zika virus/congênito , Zika virus/patogenicidade , Animais , Modelos Animais de Doenças , Embrião de Mamíferos/virologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez
2.
PLoS Negl Trop Dis ; 15(5): e0009425, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34048439

RESUMO

Zika virus (ZIKV) infection of neurons leads to neurological complications and congenital malformations of the brain of neonates. To date, ZIKV mechanism of infection and pathogenesis is not entirely understood and different studies on gene regulation of ZIKV-infected cells have identified a dysregulation of inflammatory and stem cell maintenance pathways. MicroRNAs (miRNAs) are post-transcriptional regulators of cellular genes and they contribute to cell development in normal function and disease. Previous reports with integrative analyses of messenger RNAs (mRNAs) and miRNAs during ZIKV infection have not identified neurological pathway defects. We hypothesized that dysregulation of pathways involved in neurological functions will be identified by RNA profiling of ZIKV-infected fetal neurons. We therefore used microarrays to analyze gene expression levels following ZIKV infection of fetal murine neurons. We observed that the expression levels of transcription factors such as neural PAS domain protein 4 (Npas4) and of three members of the orphan nuclear receptor 4 (Nr4a) were severely decreased after viral infection. We confirmed that their downregulation was at both the mRNA level and at the protein level. The dysregulation of these transcription factors has been previously linked to aberrant neural functions and development. We next examined the miRNA expression profile in infected primary murine neurons by microarray and found that various miRNAs were dysregulated upon ZIKV infection. An integrative analysis of the differentially expressed miRNAs and mRNAs indicated that miR-7013-5p targets Nr4a3 gene. Using miRmimics, we corroborated that miR-7013-5p downregulates Nr4a3 mRNA and protein levels. Our data identify a profound dysregulation of neural transcription factors with an overexpression of miR-7013-5p that results in decreased Nr4a3 expression, likely a main contributor to ZIKV-induced neuronal dysfunction.


Assuntos
Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Infecção por Zika virus/patologia , Zika virus/patogenicidade , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , Regulação para Baixo , Embrião de Mamíferos/virologia , Perfilação da Expressão Gênica , Camundongos , MicroRNAs/genética , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , RNA Mensageiro/genética , Fatores de Transcrição/genética
3.
J Assist Reprod Genet ; 38(7): 1709-1720, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33913101

RESUMO

PURPOSE: To predict if developing human embryos are permissive to multiple coronaviruses. METHOD: We analyzed publicly available single-cell RNA-seq datasets of human embryos for the known canonical and non-canonical receptors and spike protein cleavage enzymes for multiple coronaviruses like SARS-CoV, SARS-CoV-2, MERS-CoV, hCoV-229E, and hCoV-NL63. We also analyzed the expression of host genes involved in viral replication, host proteins involved in viral endosomal sorting complexes required for transport (ESCRT), genes of host proteins that physically interact with proteins of SARS-CoV-2, and the host genes essential for coronavirus infectivity. RESULTS: Of the known receptors of SARS viruses, ACE2, BSG, GOLGA7, and ZDHHC5 were expressed in different proportions in the zygote, 4-cell, 8-cell, morula, and blastocysts including the trophectoderm. The MERS-CoV receptor, DPP4, and hCoV-229E receptor, ANPEP, were expressed mainly from the compact morula to the blastocyst stages. Transcripts of the MERS-CoV alternate receptor LGALS1 were detected in most cells at all stages of development. TMPRSS2 transcripts were detected in the epiblast, primitive endoderm, and trophectoderm, while transcripts of the endosomal proteases CTSL, CTSB, and FURIN were expressed in most cells at all stages of development. ACE2 and TMPRSS2 were co-expressed in a proportion of epiblast and trophectoderm cells. The embryonic cells expressed genes involved in ESCRT, viral replication, SARS-CoV-2 interactions, and coronavirus infectivity. The ACE2 and TMPRSS2 co-expressing cells were enriched in genes associated with lipid metabolism, lysosome, peroxisome, and oxidative phosphorylation pathways. CONCLUSION: Preimplantation and implantation stage human embryos could be permissive to multiple hCoVs.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Blastocisto/metabolismo , Infecções por Coronavirus/metabolismo , Embrião de Mamíferos/metabolismo , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Replicação Viral , Enzima de Conversão de Angiotensina 2/genética , Blastocisto/patologia , Blastocisto/virologia , Coronavirus/fisiologia , Infecções por Coronavirus/patologia , Infecções por Coronavirus/virologia , Implantação do Embrião , Embrião de Mamíferos/patologia , Embrião de Mamíferos/virologia , Complexos Endossomais de Distribuição Requeridos para Transporte , Humanos , Serina Endopeptidases/genética , Glicoproteína da Espícula de Coronavírus/genética
4.
Reprod Toxicol ; 100: 1-6, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33338580

RESUMO

Human sperm nucleus contains diverse RNA populations. This study aimed to screen and identify host microRNAs (miRs) that regulate gene expression of hepatitis B virus (HBV) during transmission from patients' sperm to sperm-derived embryos. Using microarrays, 336 miRs were found to be differentially expressed. After validation using real-time quantitative RT-PCR (RT-qPCR), four miRs were selected as targets. Using RT-qPCR and enzyme-linked immunosorbent assays, when patients' sperm were treated with mimics (or inhibitors) specific for hsa-miR-19a-3p and hsa-miR-29c-3p, the S gene transcription in sperm and translation in sperm-derived embryos was downregulated (or upregulated). There were significant differences in transcriptional and translational levels of the S gene between the test and control groups. These findings suggest that hsa-miR-19a-3p and hsa-miR-29c-3p significantly suppressed expression of the S gene, offering potential therapeutic targets for treating patients with HBV infection, and further reducing the negative impact of HBV infection on sperm fertilizing capacity.


Assuntos
Embrião de Mamíferos/virologia , Regulação Viral da Expressão Gênica/genética , Vírus da Hepatite B/genética , Hepatite B/transmissão , MicroRNAs/genética , Espermatozoides/virologia , Adulto , Humanos , Transmissão Vertical de Doenças Infecciosas , Masculino , MicroRNAs/fisiologia , Reprodutibilidade dos Testes
5.
Mol Biol Rep ; 47(12): 9959-9965, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33226564

RESUMO

In-vitro fertilization is a routine livestock-breeding technique widely used around the world. Several studies have reported the interaction of bovine viral-diarrhea virus (BVDV) with gametes and in-vitro-produced (IVP) bovine embryos. Since, gene expression in BVDV-infected IVP bovine embryos is scarcely addressed. The aim of this work was to evaluate the differential expression of genes involved in immune and inflammatory response. Groups of 20-25 embryos on Day 6 (morula stage) were exposed (infected) or not (control) to an NCP-BVDV strain in SOF medium. After 24 h, embryos that reached expanded blastocyst stage were washed. Total RNA of each embryo group was extracted to determine the transcription levels of 9 specific transcripts related with antiviral and inflammatory response by SYBR Green real time quantitative (RT-qPCR). Culture media and an aliquot of the last embryos wash on Day 7 were analyzed by titration and virus isolation, respectively. A conventional PCR confirmed BVDV presence in IVP embryos. A significantly higher expression of interferon-α was observed in blastocysts exposed to NCP-BVDV compared to the controls (p < 0.05). In this study, the upregulation of INFα and TLR7 genes involved in inflammatory and immune response in BVDV-infected IVP bovine embryos is a new finding in this field. This differential expression suggest that embryonic cells could function in a manner like immune cells by recognizing and responding early to interaction with viral pathogens. These results provide new insights into the action of BVDV on the complex molecular pathways controlling bovine early embryonic development.


Assuntos
Doença das Mucosas por Vírus da Diarreia Viral Bovina , Bovinos , Vírus da Diarreia Viral Bovina/imunologia , Desenvolvimento Embrionário/imunologia , Expressão Gênica/imunologia , Interferon-alfa , Animais , Doença das Mucosas por Vírus da Diarreia Viral Bovina/embriologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/imunologia , Doença das Mucosas por Vírus da Diarreia Viral Bovina/virologia , Bovinos/embriologia , Bovinos/imunologia , Vírus da Diarreia Viral Bovina/isolamento & purificação , Embrião de Mamíferos/imunologia , Embrião de Mamíferos/virologia , Feminino , Fertilização in vitro , Interferon-alfa/imunologia , Receptor 7 Toll-Like/imunologia
6.
J Assist Reprod Genet ; 37(10): 2393-2398, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32748306

RESUMO

Examine good tissue practices as relates to in vitro fertilization, biopsying, and vitrificationto compare current knowledge of ova, sperm, and embryos as vectors for disease transmission as it relates to our current knowledge regarding the SARS-CoV-2 virus.Unknown risks relating to the SARS-CoV-2 virus and sperm, ova, and embryos necessitate a reexamining of how human IVF is performed. Over the last decade, improvements in cryosurvival and live birth outcomes have been associated with zona pellucida breaching procedures (e.g., blastocyst collapsing and biopsying). In turn, today embryos are generally no longer protected by an intact zona pellucida when vitrified and in cryostorage. Additionally, high security storage containers have proven to be resilient to potential cross-contamination and reliable for routine human sperm freezing and embryo vitrification.Several options to current IVF practices are presented that can effectively mitigate the risks of cross-contamination and infection due to the current Covid-19 pandemic or other viral exposures. The question remains; is heightened security and change warranted where the risks of disease transmission likely remain negligible?


Assuntos
Infecções por Coronavirus/virologia , Fertilização in vitro , Oócitos/crescimento & desenvolvimento , Pandemias , Pneumonia Viral/virologia , Betacoronavirus/patogenicidade , Blastocisto/virologia , COVID-19 , Infecções por Coronavirus/complicações , Infecções por Coronavirus/epidemiologia , Criopreservação , Técnicas de Cultura Embrionária/métodos , Transferência Embrionária/métodos , Embrião de Mamíferos/virologia , Feminino , Humanos , Nascido Vivo/epidemiologia , Oócitos/virologia , Pneumonia Viral/complicações , Pneumonia Viral/epidemiologia , Gravidez , Complicações Infecciosas na Gravidez/virologia , Taxa de Gravidez , SARS-CoV-2 , Vitrificação , Zona Pelúcida
7.
Reprod Biomed Online ; 41(2): 157-159, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32466995

RESUMO

The COVID-19 pandemic is an extraordinary global situation, and all countries have adopted their own strategies to diminish and eliminate the spread of the virus. All measures are in line with the recommendations provided by the World Health Organization. Scientific societies, such as the European Society for Human Reproduction and Embryology and American Society for Reproductive Medicine, have provided recommendations and guidance to overcome and flatten the growing curve of infection in patients who undergo IVF treatments. Although there is as yet no evidence that the virus causing COVID-19 might have negative effects on IVF outcomes, fertility treatments have been postponed in order to support healthcare systems by avoiding placing them under additional stress. The possibility of the virus affecting sperm function and egg performance cannot be excluded. In addition, an indirect effect of the virus on gametes and embryos during their manipulation cannot be ruled out. This commentary aims to provide some ideas on the possible effect of the virus on gametes and embryos, as well as how it could affect the normal functioning of the embryology laboratory.


Assuntos
Betacoronavirus , Infecções por Coronavirus/prevenção & controle , Fertilidade , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Técnicas de Reprodução Assistida/estatística & dados numéricos , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/epidemiologia , Embrião de Mamíferos/virologia , Feminino , Clínicas de Fertilização/estatística & dados numéricos , Fertilização in vitro/métodos , Fertilização in vitro/estatística & dados numéricos , Humanos , Masculino , Oócitos/virologia , Pneumonia Viral/epidemiologia , Gravidez , SARS-CoV-2 , Espermatozoides/virologia , Resultado do Tratamento
8.
J Virol ; 93(14)2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31043529

RESUMO

Chandipura virus (CHPV), a cytoplasmic RNA virus, has been implicated in several outbreaks of acute encephalitis in India. Despite the relevance of CHPV to human health, how the virus interacts with the host signaling machinery remains obscure. In response to viral infections, mammalian cells activate RelA/NF-κB heterodimers, which induce genes encoding interferon beta (IFN-ß) and other immune mediators. Therefore, RelA is generally considered to be an antiviral transcription factor. However, RelA activates a wide spectrum of genes in physiological settings, and there is a paucity of direct genetic evidence substantiating antiviral RelA functions. Using mouse embryonic fibroblasts, we genetically dissected the role of RelA in CHPV pathogenesis. We found that CHPV indeed activated RelA and that RelA deficiency abrogated the expression of IFN-ß in response to virus infections. Unexpectedly, infection of Rela-/- fibroblasts led to a decreased CHPV yield. Our investigation clarified that RelA-dependent synthesis of prosurvival factors restrained infection-inflicted cell death and that exacerbated cell death processes prevented multiplication of CHPV in RelA-deficient cells. Chikungunya virus, a cytopathic RNA virus associated also with epidemics, required RelA, and Japanese encephalitis virus, which produced relatively minor cytopathic effects in fibroblasts, circumvented the need of RelA for their propagation. In sum, we documented a proviral function of the pleiotropic factor RelA linked to its prosurvival properties. RelA promoted the growth of cytopathic RNA viruses by extending the life span of infected cells, which serve as the replicative niche of intracellular pathogens. We argue that our finding bears significance for understanding host-virus interactions and may have implications for antiviral therapeutic regimes.IMPORTANCE RelA/NF-κB participates in a wide spectrum of physiological processes, including shaping immune responses against invading pathogens. In virus-infected cells, RelA typically induces the expression of IFN-ß, which restrains viral propagation in neighboring cells involving paracrine mechanisms. Our study suggested that RelA might also play a proviral role. A cell-autonomous RelA activity amplified the yield of Chandipura virus, a cytopathic RNA virus associated with human epidemics, by extending the life span of infected cells. Our finding necessitates a substantial revision of our understanding of host-virus interactions and indicates a dual role of NF-κB signaling during the course of RNA virus infections. Our study also bears significance for therapeutic regimes which alter NF-κB activities while alleviating the viral load.


Assuntos
Embrião de Mamíferos/metabolismo , Fibroblastos/metabolismo , Interações Hospedeiro-Patógeno , Infecções por Rhabdoviridae/metabolismo , Fator de Transcrição RelA/metabolismo , Vesiculovirus/fisiologia , Células 3T3 , Animais , Linhagem Celular , Chlorocebus aethiops , Embrião de Mamíferos/patologia , Embrião de Mamíferos/virologia , Fibroblastos/patologia , Fibroblastos/virologia , Camundongos , Infecções por Rhabdoviridae/patologia , Células Vero
9.
Transbound Emerg Dis ; 66(4): 1539-1547, 2019 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30892826

RESUMO

Lumpy skin disease (LSD) is an important transboundary animal disease of cattle with significant economic impact because of the implications for international trade in live animals and animal products. LSD is caused by a Capripoxvirus, LSD virus (LSDV), and results in extensive hide and udder damage, fever and pneumonia. LSDV can be shed in semen of infected bulls for prolonged periods and transmitted venereally to cows at high doses. This study examined the effects of LSDV in frozen-thawed semen on in vitro embryo production parameters, including viral status of media and resulting embryos. Bovine oocytes were harvested from abattoir-collected ovaries and split into three experimental groups. After maturation, the oocytes were fertilized in vitro with frozen-thawed semen spiked with a high (HD) or a lower (LD) dose of LSDV, or with LSDV-free semen (control). Following day 7 and day 8 blastocyst evaluation, PCR and virus isolation were performed on all embryonic structures. After completing sufficient replicates to reach 1,000 inseminated oocytes, further in vitro fertilization (IVF) runs were performed to provide material for electron microscopy (EM) and embryo washing procedures. Overall, in vitro embryo yield was significantly reduced by the presence of LSDV in frozen-thawed semen, irrespective of viral dose. When semen with a lower viral dose was used, significantly lower oocyte cleavage rates were observed. LSDV could be detected in fertilization media and all embryo structures, when higher doses of LSDV were present in the frozen-thawed semen used for IVF. Electron microscopy demonstrated LSDV virions inside blastocysts. Following the International Embryo Transfer Society washing procedure resulted in embryos free of viral DNA; however, this may be attributable to a sampling dilution effect and should be interpreted with caution. Further research is required to better quantify the risk of LSDV transmission via assisted reproductive procedures.


Assuntos
Embrião de Mamíferos/virologia , Doença Nodular Cutânea/virologia , Vírus da Doença Nodular Cutânea/isolamento & purificação , Sêmen/virologia , Animais , Blastocisto/virologia , Bovinos , Criopreservação/veterinária , Meios de Cultura , Feminino , Fertilização in vitro/veterinária , Masculino , Carga Viral/veterinária
10.
Methods Mol Biol ; 1857: 93-99, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30136233

RESUMO

In multicellular organisms, regulated cell death plays a vital role in development, adult tissue homeostasis, and clearance of damaged or infected cells. Necroptosis is one such form of regulated cell death, characterized by its reliance on receptor-interacting protein kinase 3 (RIPK3). Once activated, RIPK3 nucleates a protein complex, termed the "necrosome," which includes the adaptors RIPK1 and FADD, and the effector protein MLKL. From the necrosome, RIPK3 phosphorylates MLKL to drive necroptosis, and can also induce RIPK1/FADD-mediated apoptosis, via caspase-8. Assembly of the necrosome thus serves as a useful readout of RIPK3 activation. In this chapter, we describe molecular methods for examining necrosome activation in response to the cytokines TNF-α, IFN-ß, and IFN-γ, and upon infection with influenza A virus (IAV).


Assuntos
Citocinas/farmacologia , Embrião de Mamíferos/patologia , Fibroblastos/patologia , Necrose , Infecções por Orthomyxoviridae/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/virologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Vírus da Influenza A/efeitos dos fármacos , Camundongos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Fosforilação , Transdução de Sinais
11.
Sci Rep ; 8(1): 12774, 2018 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-30143723

RESUMO

Zika virus (ZIKV) is associated with brain development abnormalities such as primary microcephaly, a severe reduction in brain growth. Here we demonstrated in vivo the impact of congenital ZIKV infection in blood vessel development, a crucial step in organogenesis. ZIKV was injected intravenously in the pregnant type 2 interferon (IFN)-deficient mouse at embryonic day (E) 12.5. The embryos were collected at E15.5 and postnatal day (P)2. Immunohistochemistry for cortical progenitors and neuronal markers at E15.5 showed the reduction of both populations as a result of ZIKV infection. Using confocal 3D imaging, we found that ZIKV infected brain sections displayed a reduction in the vasculature density and vessel branching compared to mocks at E15.5; altogether, cortical vessels presented a comparatively immature pattern in the infected tissue. These impaired vascular patterns were also apparent in the placenta and retina. Moreover, proteomic analysis has shown that angiogenesis proteins are deregulated in the infected brains compared to controls. At P2, the cortical size and brain weight were reduced in comparison to mock-infected animals. In sum, our results indicate that ZIKV impairs angiogenesis in addition to neurogenesis during development. The vasculature defects represent a limitation for general brain growth but also could regulate neurogenesis directly.


Assuntos
Neovascularização Fisiológica , Infecção por Zika virus/congênito , Zika virus/fisiologia , Animais , Vasos Sanguíneos/patologia , Encéfalo/irrigação sanguínea , Encéfalo/patologia , Encéfalo/virologia , Modelos Animais de Doenças , Embrião de Mamíferos/patologia , Embrião de Mamíferos/virologia , Células Endoteliais/patologia , Células Endoteliais/virologia , Feminino , Camundongos Endogâmicos C57BL , Neurogênese , Tamanho do Órgão , Infecção por Zika virus/patologia , Infecção por Zika virus/virologia
12.
Cold Spring Harb Protoc ; 2018(3)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29496815

RESUMO

This protocol describes the injection of lentivirus into the perivitelline space of fertilized oocytes. Embryos usually survive lentiviral injection well because only the zona is pierced.


Assuntos
Embrião de Mamíferos/virologia , Lentivirus/fisiologia , Microinjeções/métodos , Animais , Camundongos
13.
Cold Spring Harb Protoc ; 2018(3)2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29496816

RESUMO

Lentiviral vectors make it possible to produce transgenic animals without the need to purchase expensive microinjection equipment because integration of the transgene is possible by coculture of embryos with virus. This protocol describes the infection of fertilized oocytes with lentivirus by coculture.


Assuntos
Técnicas de Cocultura/métodos , Embrião de Mamíferos/virologia , Lentivirus/fisiologia , Animais , Camundongos , Microinjeções
14.
Biomed Res Int ; 2017: 1840417, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28890894

RESUMO

The introduction and widespread application of vitrification are one of the most important achievements in human assisted reproduction techniques (ART) of the past decade despite controversy and unclarified issues, mostly related to concerns about disease transmission. Guidance documents published by US Food and Drug Administration, which focused on the safety of tissue/organ donations during Zika virus spread in 2016, as well as some reports of virus, bacteria, and fungi survival to cryogenic temperatures, highlighted the need for a review of the way how potentially infectious material is handled and stored in ART-related procedures. It was experimentally demonstrated that cross-contamination between liquid nitrogen (LN2) and embryos may occur when infectious agents are present in LN2 and oocytes/embryos are not protected by a hermetically sealed device. Thus, this review summarizes pertinent data and opinions regarding the potential hazard of infectious transmission through cryopreserved and banked reproductive cells and tissues in LN2. Special attention is given to the survival of pathogens in LN2, the risk of cross-contamination, vitrification methods, sterility of LN2, and the risks associated with the use of straws, cryovials, and storage dewars.


Assuntos
Criopreservação , Embrião de Mamíferos/virologia , Células Germinativas/virologia , Infecção por Zika virus/virologia , Células Germinativas/crescimento & desenvolvimento , Humanos , Oócitos/virologia , Técnicas de Reprodução Assistida , Obtenção de Tecidos e Órgãos , Estados Unidos , United States Food and Drug Administration , Vitrificação , Zika virus/patogenicidade , Infecção por Zika virus/transmissão
15.
Cell ; 170(2): 273-283.e12, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28708997

RESUMO

The emergence of Zika virus (ZIKV) and its association with congenital malformations has prompted the rapid development of vaccines. Although efficacy with multiple viral vaccine platforms has been established in animals, no study has addressed protection during pregnancy. We tested in mice two vaccine platforms, a lipid nanoparticle-encapsulated modified mRNA vaccine encoding ZIKV prM and E genes and a live-attenuated ZIKV strain encoding an NS1 protein without glycosylation, for their ability to protect against transmission to the fetus. Vaccinated dams challenged with a heterologous ZIKV strain at embryo day 6 (E6) and evaluated at E13 showed markedly diminished levels of viral RNA in maternal, placental, and fetal tissues, which resulted in protection against placental damage and fetal demise. As modified mRNA and live-attenuated vaccine platforms can restrict in utero transmission of ZIKV in mice, their further development in humans to prevent congenital ZIKV syndrome is warranted.


Assuntos
Vacinas Virais/administração & dosagem , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/fisiologia , Aedes/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Sanguíneas/virologia , Embrião de Mamíferos/virologia , Feminino , Feto/virologia , Humanos , Lipídeos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Organismos Livres de Patógenos Específicos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/imunologia , Infecção por Zika virus/virologia
16.
Sci Signal ; 10(476)2017 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-28442624

RESUMO

Interferon (IFN)-stimulated genes (ISGs) are antiviral effectors that are induced by IFNs through the formation of a tripartite transcription factor ISGF3, which is composed of IRF9 and phosphorylated forms of STAT1 and STAT2. However, we found that IFN-independent ISG expression was detectable in immortalized cell lines, primary intestinal and liver organoids, and liver tissues. The constitutive expression of ISGs was mediated by the unphosphorylated ISGF3 (U-ISGF3) complex, consisting of IRF9 together with unphosphorylated STAT1 and STAT2. Under homeostatic conditions, STAT1, STAT2, and IRF9 were found in the nucleus. Analysis of a chromatin immunoprecipitation sequencing data set revealed that STAT1 specifically bound to the promoters of ISGs even in the absence of IFNs. Knockdown of STAT1, STAT2, or IRF9 by RNA interference led to the decreased expression of various ISGs in Huh7.5 human liver cells, which was confirmed in mouse embryonic fibroblasts (MEFs) from STAT1-/-, STAT2-/-, or IRF9-/- mice. Furthermore, decreased ISG expression was accompanied by increased replication of hepatitis C virus and hepatitis E virus. Conversely, simultaneous overexpression of all ISGF3 components, but not any single factor, induced the expression of ISGs and inhibited viral replication; however, no phosphorylated STAT1 and STAT2 were detected. A phosphorylation-deficient STAT1 mutant was comparable to the wild-type protein in mediating the IFN-independent expression of ISGs and antiviral activity, suggesting that ISGF3 works in a phosphorylation-independent manner. These data suggest that the U-ISGF3 complex is both necessary and sufficient for constitutive ISG expression and antiviral immunity under homeostatic conditions.


Assuntos
Hepatite C/prevenção & controle , Hepatite E/prevenção & controle , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Fator Gênico 3 Estimulado por Interferon/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT2/metabolismo , Animais , Antivirais/farmacologia , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/efeitos dos fármacos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/virologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação da Expressão Gênica/efeitos dos fármacos , Hepacivirus/efeitos dos fármacos , Hepatite C/metabolismo , Hepatite C/virologia , Hepatite E/metabolismo , Hepatite E/virologia , Vírus da Hepatite E/efeitos dos fármacos , Humanos , Fator Gênico 3 Estimulado por Interferon/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Interferon-alfa/farmacologia , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/efeitos dos fármacos , Intestinos/virologia , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/virologia , Camundongos , Camundongos Knockout , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
18.
J Biol Chem ; 292(1): 292-304, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27879319

RESUMO

The host protein Stimulator of Interferon Genes (STING) has been shown to be essential for recognition of both viral and intracellular bacterial pathogens, but its regulation remains unclear. Previously, we reported that mitochondrial membrane potential regulates STING-dependent IFN-ß induction independently of ATP synthesis. Because mitochondrial membrane potential controls calcium homeostasis, and AMP-activated protein kinase (AMPK) is regulated, in part, by intracellular calcium, we postulated that AMPK participates in STING activation; however, its role has yet to be been defined. Addition of an intracellular calcium chelator or an AMPK inhibitor to either mouse macrophages or mouse embryonic fibroblasts (MEFs) suppressed IFN-ß and TNF-α induction following stimulation with the STING-dependent ligand 5,6-dimethyl xanthnone-4-acetic acid (DMXAA). These pharmacological findings were corroborated by showing that MEFs lacking AMPK activity also failed to up-regulate IFN-ß and TNF-α after treatment with DMXAA or the natural STING ligand cyclic GMP-AMP (cGAMP). As a result, AMPK-deficient MEFs exhibit impaired control of vesicular stomatitis virus (VSV), a virus sensed by STING that can cause an influenza-like illness in humans. This impairment could be overcome by pretreatment of AMPK-deficient MEFs with type I IFN, illustrating that de novo production of IFN-ß in response to VSV plays a key role in antiviral defense during infection. Loss of AMPK also led to dephosphorylation at Ser-555 of the known STING regulator, UNC-51-like kinase 1 (ULK1). However, ULK1-deficient cells responded normally to DMXAA, indicating that AMPK promotes STING-dependent signaling independent of ULK1 in mouse cells.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Antivirais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/fisiologia , Imunidade Inata/imunologia , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/fisiologia , Vírus da Estomatite Vesicular Indiana/imunologia , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Embrião de Mamíferos/imunologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/virologia , Fibroblastos/citologia , Fibroblastos/imunologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Macrófagos Peritoneais , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosforilação , Estomatite Vesicular/imunologia , Estomatite Vesicular/metabolismo , Estomatite Vesicular/virologia
19.
Theriogenology ; 86(7): 1631-6, 2016 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-27423729

RESUMO

In the modern biological area, the applications of pig as a laboratory model have extensive prospects, such as gene transfer, IVF, SCNT, and xenotransplantation. However, the risk of pathogen transmission by porcine embryos is always a topic to be investigated, especially the viruses related to reproductive failure, for instance, pseudorabies virus, porcine reproductive and respiratory syndrome virus, porcine parvovirus, and porcine circovirus type 2. It should be mentioned that the zona pellucida (ZP) of porcine embryos can be a barrier against the viruses, but certain pathogens may stick to or even pass through the ZP. With intact, free, and damaged ZP, porcine preimplantation embryos are susceptible to these viruses in varying degrees, which may be associated with the virus-specific receptor on embryonic cell membrane. These topics are discussed in the present review.


Assuntos
Blastocisto/virologia , Embrião de Mamíferos/virologia , Suínos/embriologia , Vírus/classificação , Animais , Zona Pelúcida/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...