Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 311
Filtrar
1.
Int J Mol Sci ; 25(19)2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39408667

RESUMO

Human exploration of the solar system will expose crew members to galactic cosmic radiation (GCR), with a potential for adverse health effects. GCR particles (protons and ions) move at nearly the speed of light and easily penetrate space station walls, as well as the human body. Previously, we have shown reactivation of latent herpesviruses, including herpes simplex virus, Varicella zoster virus, Epstein-Barr virus, and cytomegalovirus (CMV), during stays at the International Space Station. Given the prevalence of latent CMV and the known propensity of space radiation to cause alterations in many cellular processes, we undertook this study to understand the role of GCR in reactivating latent CMV. Latently infected Kasumi cells with CMV were irradiated with 137Cs gamma rays, 150 MeV protons, 600 MeV/n carbon ions, 600 MeV/n iron ions, proton ions, and simulated GCR. The CMV copy number increased significantly in the cells exposed to radiation as compared with the non-irradiated controls. Viral genome sequencing did not reveal significant nucleotide differences among the compared groups. However, transcriptome analysis showed the upregulation of transcription of the UL49 ORF, implicating it in the switch from latent to lytic replication. These findings support our hypothesis that GCR may be a strong contributor to the reactivation of CMV infection seen in ISS crew members.


Assuntos
Radiação Cósmica , Citomegalovirus , Ativação Viral , Replicação Viral , Radiação Cósmica/efeitos adversos , Citomegalovirus/fisiologia , Citomegalovirus/efeitos da radiação , Humanos , Replicação Viral/efeitos da radiação , Ativação Viral/efeitos da radiação , Latência Viral/efeitos da radiação , Genoma Viral , Raios gama , Infecções por Citomegalovirus/virologia , Linhagem Celular
2.
ACS Nano ; 18(28): 18282-18298, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-38953884

RESUMO

The therapeutic efficacy of oncolytic adenoviruses (OAs) relies on efficient viral transduction and replication. However, the limited expression of coxsackie-adenovirus receptors in many tumors, along with the intracellular antiviral signaling, poses significant obstacles to OA infection and oncolysis. Here, we present sonosensitizer-armed OAs (saOAs) that potentiate the antitumor efficacy of oncolytic virotherapy through sonodynamic therapy-augmented virus replication. The saOAs could not only efficiently infect tumor cells via transferrin receptor-mediated endocytosis but also exhibit enhanced viral replication and tumor oncolysis under ultrasound irradiation. We revealed that the sonosensitizer loaded on the viruses induced the generation of ROS within tumor cells, which triggered JNK-mediated autophagy, ultimately leading to the enhanced viral replication. In mouse models of malignant melanoma, the combination of saOAs and sonodynamic therapy elicited a robust antitumor immune response, resulting in significant inhibition of melanoma growth and improved host survival. This work highlights the potential of sonodynamic therapy in enhancing the effectiveness of OAs and provides a promising platform for fully exploiting the antitumor efficacy of oncolytic virotherapy.


Assuntos
Adenoviridae , Terapia Viral Oncolítica , Vírus Oncolíticos , Replicação Viral , Animais , Terapia Viral Oncolítica/métodos , Adenoviridae/genética , Adenoviridae/fisiologia , Vírus Oncolíticos/fisiologia , Vírus Oncolíticos/genética , Replicação Viral/efeitos da radiação , Camundongos , Humanos , Linhagem Celular Tumoral , Terapia por Ultrassom/métodos , Melanoma/terapia , Melanoma/patologia
3.
Virology ; 595: 110063, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38564935

RESUMO

This experimental study aimed to evaluate the antiviral and synergistic effects of photoenergy irradiation on human herpes simplex virus type I (HSV-1) infection. We assessed viral replication, plaque formation, and relevant viral gene expression to examine the antiviral and synergistic effects of blue light (BL) with acyclovir treatment. Our results showed that daily BL (10 J/cm2) irradiation inhibited plaque-forming ability and decreased viral copy numbers in HSV-1-infected monkey kidney epithelial Vero cells and primary human oral keratinocyte (HOK) cells. Combined treatment with the antiviral agent acyclovir and BL irradiation increased anti-viral activity, reducing viral titers and copy numbers. In particular, accumulated BL irradiation suppressed characteristic viral genes including UL19 and US6, and viral DNA replication-essential genes including UL9, UL30, UL42, and UL52 in HOK cells. Our results suggest that BL irradiation has anti-viral and synergistic properties, making it a promising therapeutic candidate for suppressing viral infections in clinical trials.


Assuntos
Aciclovir , Antivirais , Herpesvirus Humano 1 , Replicação Viral , Antivirais/farmacologia , Animais , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 1/efeitos da radiação , Herpesvirus Humano 1/fisiologia , Herpesvirus Humano 1/genética , Chlorocebus aethiops , Células Vero , Humanos , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação , Aciclovir/farmacologia , Luz , Herpes Simples/virologia , Herpes Simples/tratamento farmacológico , Queratinócitos/virologia , Queratinócitos/efeitos da radiação , Queratinócitos/efeitos dos fármacos , Ensaio de Placa Viral
4.
J Photochem Photobiol B ; 224: 112319, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34598020

RESUMO

The germicidal properties of short wavelength ultraviolet C (UVC) light are well established and used to inactivate many viruses and other microbes. However, much less is known about germicidal effects of terrestrial solar UV light, confined exclusively to wavelengths in the UVA and UVB regions. Here, we have explored the sensitivity of the human coronaviruses HCoV-NL63 and SARS-CoV-2 to solar-simulated full spectrum ultraviolet light (sUV) delivered at environmentally relevant doses. First, HCoV-NL63 coronavirus inactivation by sUV-exposure was confirmed employing (i) viral plaque assays, (ii) RT-qPCR detection of viral genome replication, and (iii) infection-induced stress response gene expression array analysis. Next, a detailed dose-response relationship of SARS-CoV-2 coronavirus inactivation by sUV was elucidated, suggesting a half maximal suppression of viral infectivity at low sUV doses. Likewise, extended sUV exposure of SARS-CoV-2 blocked cellular infection as revealed by plaque assay and stress response gene expression array analysis. Moreover, comparative (HCoV-NL63 versus SARS-CoV-2) single gene expression analysis by RT-qPCR confirmed that sUV exposure blocks coronavirus-induced redox, inflammatory, and proteotoxic stress responses. Based on our findings, we estimate that solar ground level full spectrum UV light impairs coronavirus infectivity at environmentally relevant doses. Given the urgency and global scale of the unfolding SARS-CoV-2 pandemic, these prototype data suggest feasibility of solar UV-induced viral inactivation, an observation deserving further molecular exploration in more relevant exposure models.


Assuntos
Infecções por Coronavirus/prevenção & controle , Coronavirus Humano NL63/efeitos da radiação , Infecções Respiratórias/prevenção & controle , SARS-CoV-2/efeitos da radiação , Luz Solar , Raios Ultravioleta , Animais , Linhagem Celular , Chlorocebus aethiops , Coronavirus Humano NL63/fisiologia , Células Epiteliais/virologia , Genoma Viral/efeitos da radiação , Humanos , SARS-CoV-2/fisiologia , Transcriptoma/efeitos da radiação , Ensaio de Placa Viral , Inativação de Vírus/efeitos da radiação , Replicação Viral/efeitos da radiação
5.
Sci Rep ; 11(1): 20595, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34663881

RESUMO

The delivery of safe, visible wavelengths of light can be an effective, pathogen-agnostic, countermeasure that would expand the current portfolio of SARS-CoV-2 intervention strategies beyond the conventional approaches of vaccine, antibody, and antiviral therapeutics. Employing custom biological light units, that incorporate optically engineered light-emitting diode (LED) arrays, we harnessed monochromatic wavelengths of light for uniform delivery across biological surfaces. We demonstrated that primary 3D human tracheal/bronchial-derived epithelial tissues tolerated high doses of a narrow spectral band of visible light centered at a peak wavelength of 425 nm. We extended these studies to Vero E6 cells to understand how light may influence the viability of a mammalian cell line conventionally used for assaying SARS-CoV-2. The exposure of single-cell monolayers of Vero E6 cells to similar doses of 425 nm blue light resulted in viabilities that were dependent on dose and cell density. Doses of 425 nm blue light that are well-tolerated by Vero E6 cells also inhibited infection and replication of cell-associated SARS-CoV-2 by > 99% 24 h post-infection after a single five-minute light exposure. Moreover, the 425 nm blue light inactivated cell-free betacoronaviruses including SARS-CoV-1, MERS-CoV, and SARS-CoV-2 up to 99.99% in a dose-dependent manner. Importantly, clinically applicable doses of 425 nm blue light dramatically inhibited SARS-CoV-2 infection and replication in primary human 3D tracheal/bronchial tissue. Safe doses of visible light should be considered part of the strategic portfolio for the development of SARS-CoV-2 therapeutic countermeasures to mitigate coronavirus disease 2019 (COVID-19).


Assuntos
Tratamento Farmacológico da COVID-19 , COVID-19/prevenção & controle , Luz , SARS-CoV-2 , Traqueia/efeitos da radiação , Replicação Viral/efeitos da radiação , Adulto , Animais , Antivirais/farmacologia , Brônquios , Calibragem , Sistema Livre de Células , Chlorocebus aethiops , Epitélio/patologia , Feminino , Humanos , Mucosa Respiratória/efeitos da radiação , Traqueia/virologia , Células Vero
6.
Sci Rep ; 11(1): 14295, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34253743

RESUMO

Methylene blue is an FDA (Food and Drug Administration) and EMA (European Medicines Agency) approved drug with an excellent safety profile. It displays broad-spectrum virucidal activity in the presence of UV light and has been shown to be effective in inactivating various viruses in blood products prior to transfusions. In addition, its use has been validated for methemoglobinemia and malaria treatment. In this study, we first evaluated the virucidal activity of methylene blue against influenza virus H1N1 upon different incubation times and in the presence or absence of light activation, and then against SARS-CoV-2. We further assessed the therapeutic activity of methylene blue by administering it to cells previously infected with SARS-CoV-2. Finally, we examined the effect of co-administration of the drug together with immune serum. Our findings reveal that methylene blue displays virucidal preventive or therapeutic activity against influenza virus H1N1 and SARS-CoV-2 at low micromolar concentrations and in the absence of UV-activation. We also confirm that MB antiviral activity is based on several mechanisms of action as the extent of genomic RNA degradation is higher in presence of light and after long exposure. Our work supports the interest of testing methylene blue in clinical studies to confirm a preventive and/or therapeutic efficacy against both influenza virus H1N1 and SARS-CoV-2 infections.


Assuntos
Tratamento Farmacológico da COVID-19 , Influenza Humana/tratamento farmacológico , Azul de Metileno/farmacologia , Inativação de Vírus/efeitos dos fármacos , Animais , COVID-19/genética , COVID-19/virologia , Chlorocebus aethiops , Humanos , Influenza Humana/genética , Influenza Humana/virologia , SARS-CoV-2/efeitos dos fármacos , SARS-CoV-2/patogenicidade , Raios Ultravioleta/efeitos adversos , Células Vero , Inativação de Vírus/efeitos da radiação , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação
7.
Nat Commun ; 12(1): 2187, 2021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33846346

RESUMO

The RNA-sensing pathway contributes to type I interferon (IFN) production induced by DNA damaging agents. However, the potential involvement of RNA sensors in DNA repair is unknown. Here, we found that retinoic acid-inducible gene I (RIG-I), a key cytosolic RNA sensor that recognizes RNA virus and initiates the MAVS-IRF3-type I IFN signaling cascade, is recruited to double-stranded breaks (DSBs) and suppresses non-homologous end joining (NHEJ). Mechanistically, RIG-I interacts with XRCC4, and the RIG-I/XRCC4 interaction impedes the formation of XRCC4/LIG4/XLF complex at DSBs. High expression of RIG-I compromises DNA repair and sensitizes cancer cells to irradiation treatment. In contrast, depletion of RIG-I renders cells resistant to irradiation in vitro and in vivo. In addition, this mechanism suggests a protective role of RIG-I in hindering retrovirus integration into the host genome by suppressing the NHEJ pathway. Reciprocally, XRCC4, while suppressed for its DNA repair function, has a critical role in RIG-I immune signaling through RIG-I interaction. XRCC4 promotes RIG-I signaling by enhancing oligomerization and ubiquitination of RIG-I, thereby suppressing RNA virus replication in host cells. In vivo, silencing XRCC4 in mouse lung promotes influenza virus replication in mice and these mice display faster body weight loss, poorer survival, and a greater degree of lung injury caused by influenza virus infection. This reciprocal regulation of RIG-I and XRCC4 reveals a new function of RIG-I in suppressing DNA repair and virus integration into the host genome, and meanwhile endues XRCC4 with a crucial role in potentiating innate immune response, thereby helping host to prevail in the battle against virus.


Assuntos
Proteína DEAD-box 58/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/metabolismo , Receptores Imunológicos/metabolismo , Transdução de Sinais/imunologia , Células A549 , Animais , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo do DNA por Junção de Extremidades/efeitos da radiação , Reparo do DNA/efeitos da radiação , Relação Dose-Resposta à Radiação , Genoma Humano , Células HEK293 , Humanos , Camundongos , Radiação Ionizante , Retroviridae/metabolismo , Replicação Viral/efeitos da radiação
8.
Sci Rep ; 11(1): 6260, 2021 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-33737536

RESUMO

The potential virucidal effects of UV-C irradiation on SARS-CoV-2 were experimentally evaluated for different illumination doses and virus concentrations (1000, 5, 0.05 MOI). At a virus density comparable to that observed in SARS-CoV-2 infection, an UV-C dose of just 3.7 mJ/cm2 was sufficient to achieve a more than 3-log inactivation without any sign of viral replication. Moreover, a complete inactivation at all viral concentrations was observed with 16.9 mJ/cm2. These results could explain the epidemiological trends of COVID-19 and are important for the development of novel sterilizing methods to contain SARS-CoV-2 infection.


Assuntos
SARS-CoV-2/efeitos da radiação , Raios Ultravioleta , Inativação de Vírus , Replicação Viral/efeitos da radiação
9.
JNCI Cancer Spectr ; 5(1): pkaa103, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33437924

RESUMO

The coronavirus disease-2019 (COVID-19) pandemic caused by SARS-CoV-2 has exacted an enormous toll on healthcare systems worldwide. The cytokine storm that follows pulmonary infection is causally linked to respiratory compromise and mortality in the majority of patients. The sparsity of viable treatment options for this viral infection and the sequelae of pulmonary complications have fueled the quest for new therapeutic considerations. One such option, the long-forgotten idea of using low-dose radiation therapy, has recently found renewed interest in many academic centers. We outline the scientific and logistical rationale for consideration of this option and the mechanistic underpinnings of any potential therapeutic value, particularly as viewed from an immunological perspective. We also discuss the preliminary and/or published results of prospective trials examining low-dose radiation therapy for COVID-19.


Assuntos
COVID-19/radioterapia , Doses de Radiação , SARS-CoV-2/efeitos da radiação , Internalização do Vírus/efeitos da radiação , Replicação Viral/efeitos da radiação , COVID-19/epidemiologia , COVID-19/virologia , Citocinas/metabolismo , Relação Dose-Resposta à Radiação , Interações Hospedeiro-Patógeno/efeitos da radiação , Humanos , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/efeitos da radiação , Leucócitos Mononucleares/virologia , Pandemias , Dosagem Radioterapêutica , SARS-CoV-2/fisiologia
10.
PLoS One ; 15(1): e0226758, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31899921

RESUMO

The pelagophyte Aureococcus anophagefferens has caused recurrent brown tide blooms along the northeast coast of the United States since the mid-1980's, and more recently spread to other regions of the globe. These blooms, due to the high cell densities, are associated with severe light attenuation that destroys the sea grass beds which provide the basis for many fisheries. Data collected by transmission electron microscopy, PCR, and metatranscriptomic studies of the blooms, support the hypothesis that large dsDNA viruses play a role in bloom dynamics. While a large (~140 nm) icosahedral virus, with a 371 kbp genome, was first isolated more than a decade ago, the constraints imposed by environmental parameters on bloom infection dynamics by Aureococcus anophagefferens Virus, (AaV) remain unknown. To investigate the role light plays in infection by this virus, we acclimated A. anophagefferens to light intensities of 30 (low), 60 (medium) or 90 µmol photons m-2 s-1 (high) and infected cultures at these irradiance levels. Moreover, we completed light shift experiments where acclimated cultures were exposed to even lower light intensities (0, 5, and 15 µmol photons m-2 s-1) consistent with irradiance found during the peak of the bloom when cell concentrations are highest. The abundance of viruses produced per lytic event (burst size) was lower in the low irradiance acclimated cultures compared to the medium and high acclimated cultures. Transferring infected cultures to more-limiting light availabilities further decreased burst size and increased the length of time it took for cultures to lyse, regardless of acclimation irradiance level. A hypothetical mechanism for the reduced efficiency of the infection cycle in low light due to ribosome biogenesis was predicted from pre-existing transcriptomes. Overall, these studies provide a framework for understanding light effects on infection dynamics over the course of the summer months when A. anophagefferens blooms occur.


Assuntos
Infecções por Vírus de DNA/virologia , Vírus Gigantes/fisiologia , Interações Hospedeiro-Patógeno , Luz , Microalgas/crescimento & desenvolvimento , Microalgas/virologia , Replicação Viral/efeitos da radiação , Microalgas/efeitos da radiação
11.
Viruses ; 10(11)2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30463209

RESUMO

H9N2 avian influenza viruses are present in poultry worldwide. These viruses are considered to have pandemic potential, because recent isolates can recognize human-type receptor and several sporadic human infections have been reported. In this study, we aimed to identify mutations related to mammalian adaptation of H9N2 influenza virus. We found that mouse-adapted viruses had several mutations in hemagglutinin (HA), PB2, PA, and PB1. Among the detected mutations, PB1-K577E was a novel mutation that had not been previously reported to involve mammalian adaptation. A recombinant H9N2 virus bearing only the PB1-K577E mutation showed enhanced pathogenicity in mice, with increased virus titers in nasal turbinates compared to that in mice infected with the wild-type virus. In addition, the PB1-K577E mutation increased virus polymerase activity in human cell culture at a lower temperature. These data suggest that the PB1-K577E mutation is a novel pathogenicity determinant of H9N2 virus in mice and could be a signature for mammalian adaptation.


Assuntos
Vírus da Influenza A Subtipo H9N2/enzimologia , Vírus da Influenza A Subtipo H9N2/patogenicidade , Mutação de Sentido Incorreto , Infecções por Orthomyxoviridae/virologia , Proteínas Virais/genética , Proteínas Virais/metabolismo , Adaptação Biológica , Animais , Modelos Animais de Doenças , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H9N2/genética , Camundongos , Cavidade Nasal/virologia , Infecções por Orthomyxoviridae/patologia , Temperatura , Carga Viral , Replicação Viral/efeitos da radiação
12.
J Photochem Photobiol B ; 189: 193-200, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30391908

RESUMO

Influenza A viruses (IAVs) pose a serious global threat to humans and their livestock, especially poultry and pigs. This study aimed to investigate how to inactivate IAVs by using different ultraviolet-light-emitting diodes (UV-LEDs). We developed sterilization equipment with light-emitting diodes (LEDs) those peak wavelengths were 365 nm (UVA-LED), 310 nm (UVB-LED), and 280 nm (UVC-LED). These UV-LED irradiations decreased dose fluence-dependent plaque-forming units of IAV H1N1 subtype (A/Puerto Rico/8/1934) infected Madin-Darby canine kidney (MDCK) cells, but the inactivation efficiency of UVA-LED was significantly lower than UVB- and UVC-LED. UV-LED irradiations did not alter hemagglutination titer, but decreased accumulation of intracellular total viral RNA in infected MDCK cells was observed. Additionally, UV-LED irradiations suppressed the accumulation of intracellular mRNA (messenger RNA), vRNA (viral RNA), and cRNA (complementary RNA), as measured by strand-specific RT-PCR. These results suggest that UV-LEDs inhibit host cell replication and transcription of viral RNA. Both UVB- and UVC-LED irradiation decreased focus-forming unit (FFU) of H5N1 subtype (A/Crow/Kyoto/53/2004), a highly pathogenic avian IAV (HPAI), in infected MDCK cells, and the amount of FFU were lower than the H1N1 subtype. From these results, it appears that IAVs may have different sensitivity among the subtypes, and UVB- and UVC-LED may be suitable for HPAI virus inactivation.


Assuntos
Vírus da Influenza A/efeitos da radiação , Raios Ultravioleta , Inativação de Vírus/efeitos da radiação , Animais , Cães , Humanos , Vírus da Influenza A Subtipo H1N1 , Células Madin Darby de Rim Canino/virologia , Infecções por Orthomyxoviridae , RNA Viral/biossíntese , RNA Viral/genética , Transcrição Gênica/efeitos da radiação , Replicação Viral/efeitos da radiação
13.
PLoS One ; 13(10): e0202352, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30365500

RESUMO

More than 70 human adenovirus types were identified divided into 7 different species (A-G). Diseases caused by human adenoviruses are type-dependent and can range from mild to severe respiratory infections, gastrointestinal infections or eye infections such as epidemic keratoconjunctivitis. Unfortunately there is no specific anti-adenovirus therapy available. Here we addressed the question whether treatment with cold atmospheric plasma (CAP) for anti-adenoviral therapy such as virus-mediated ulcerations may be feasible. CAP has already been explored for the treatment of dermatological diseases such as chronic wounds. To investigate whether CAP is an effective antiviral tool, purified human adenovirus types derived from different human adenovirus species (HAdV -4, -5, -20, -35, -37, -50) tagged with luciferase were treated with defined dosages of plasma. The CAP treatment was varied by incrementally increasing the time span of CAP treatment. After CAP treatment, the virus containing solution was added to eukaryotic cells and the viral load was determined by measurement of luciferase expression levels. Through the plasma treatment the adenovirus driven luciferase expression directly correlating with adenovirus transduction efficiencies could be reduced for HAdV-5 and HAdV-37. Plasma treatment had no influence on adenovirus derived luciferase expression levels for HAdV-4 and HAdV-50 and it even had a positive effect on luciferase expression levels for HAdV-20 and HAdV-35. These results suggest that CAP has a type dependent effect on adenoviruses and that infectivity can be even increased for certain adenovirus types. Further studies should address the mechanisms behind this phenomenon. In summary we demonstrate that CAP may represent an interesting option for antiviral treatment in a virus type dependent manner.


Assuntos
Infecções por Adenovirus Humanos/terapia , Adenovírus Humanos/efeitos da radiação , Conjuntivite Viral/terapia , Gases em Plasma/uso terapêutico , Infecções por Adenovirus Humanos/fisiopatologia , Infecções por Adenovirus Humanos/virologia , Adenovírus Humanos/patogenicidade , Conjuntivite Viral/fisiopatologia , Conjuntivite Viral/virologia , DNA Viral , Humanos , Infecções Respiratórias/terapia , Infecções Respiratórias/virologia , Carga Viral/efeitos da radiação , Replicação Viral/efeitos da radiação
14.
Virus Res ; 251: 91-97, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29753891

RESUMO

In the present study we assessed pleiotropic characteristics of the antibody-selected mutations. We investigated pH optimum of fusion, temperatures of HA heat inactivation, in vivo and in vitro replication kinetics, and connectivity with panel of sera of survivors patients in different epidemic seasons of the previously obtained influenza H1 escape mutants. Our results showed that N133D (H3 numbering) mutation significantly lowered the pH of fusion optimum. Several amino acid substitutions, including K163 N, Q192 L, D190E, G228E, and K285 M, reduced the stability of HA as determined by heat inactivation, whereas A198E substitution is associated with significant increase in HA thermostability compared to the wild-type virus. We found that amino acid change D190 N was associated with a significant decrease in viral growth in eggs and mice. Our potential antigenic variants, except readapted variant, which contained A198E mutation, did not reach fixation in infected people. Overall, a co-variation between antigenic specificity and different HA phenotypic properties was demonstrated.


Assuntos
Substituição de Aminoácidos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Evasão da Resposta Imune , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/fisiologia , Mutação de Sentido Incorreto , Animais , Anticorpos Antivirais/imunologia , Embrião de Galinha , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Concentração de Íons de Hidrogênio , Vírus da Influenza A Subtipo H1N1/imunologia , Camundongos , Estabilidade Proteica , Temperatura , Internalização do Vírus/efeitos dos fármacos , Internalização do Vírus/efeitos da radiação , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação
15.
Transfusion ; 58(1): 52-59, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29239484

RESUMO

BACKGROUND: Middle East respiratory syndrome-coronavirus (MERS-CoV) is a novel zoonotic pathogen. Although the potential for MERS-CoV transmission through blood transfusion is not clear, MERS-CoV was recognized as a pathogen of concern for the safety of the blood supply especially after its detection in whole blood, serum, and plasma of infected individuals. Here we investigated the efficacy of amotosalen and ultraviolet A light (UVA) to inactivate MERS-CoV in fresh-frozen plasma (FFP). STUDY DESIGN AND METHODS: Pooled FFP units were spiked with a recent clinical MERS-CoV isolate. Infectious and genomic viral titers were determined in plasma before and after inactivation with amotosalen/UVA treatment by plaque assay and reverse transcription-quantitative polymerase chain reaction, respectively. In addition, residual replicating or live virus after inactivation was examined by passaging in the permissive Vero E6 cells. RESULTS: The mean MERS-CoV infectious titer in pretreatment samples was 4.67 ± 0.25 log plaque-forming units (pfu)/mL, which was reduced to undetectable levels after inactivation with amotosalen/UVA demonstrating a mean log reduction of more than 4.67 ± 0.25 pfu/mL. Furthermore, inoculation of inactivated plasma on Vero E6 cells did not result in any cytopathic effect (CPE) even after 7 days of incubation and three consecutive passages, nor the detection of MERS RNA compared to pretreatment samples which showed complete CPE within 2 to 3 days postinoculation and log viral RNA titer ranging from 9.48 to 10.22 copies/mL in all three passages. CONCLUSION: Our data show that amotosalen/UVA treatment is a potent and effective way to inactivate MERS-CoV infectious particles in FFP to undetectable levels and to minimize the risk of any possible transfusion-related MERS-CoV transmission.


Assuntos
Furocumarinas/farmacologia , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Fármacos Fotossensibilizantes/farmacologia , Plasma/virologia , Raios Ultravioleta , Inativação de Vírus , Animais , Chlorocebus aethiops , Efeito Citopatogênico Viral , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/crescimento & desenvolvimento , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos da radiação , RNA Viral/sangue , Reação em Cadeia da Polimerase em Tempo Real , Células Vero , Carga Viral/efeitos dos fármacos , Carga Viral/efeitos da radiação , Ensaio de Placa Viral , Replicação Viral/efeitos dos fármacos , Replicação Viral/efeitos da radiação
16.
Viruses ; 9(6)2017 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-28574420

RESUMO

Global climate change-induced warming of the Artic seas is predicted to shift the phytoplankton community towards dominance of smaller-sized species due to global warming. Yet, little is known about their viral mortality agents despite the ecological importance of viruses regulating phytoplankton host dynamics and diversity. Here we report the isolation and basic characterization of four prasinoviruses infectious to the common Arctic picophytoplankter Micromonas. We furthermore assessed how temperature influenced viral infectivity and production. Phylogenetic analysis indicated that the putative double-stranded DNA (dsDNA) Micromonas polaris viruses (MpoVs) are prasinoviruses (Phycodnaviridae) of approximately 120 nm in particle size. One MpoV showed intrinsic differences to the other three viruses, i.e., larger genome size (205 ± 2 vs. 191 ± 3 Kb), broader host range, and longer latent period (39 vs. 18 h). Temperature increase shortened the latent periods (up to 50%), increased the burst size (up to 40%), and affected viral infectivity. However, the variability in response to temperature was high for the different viruses and host strains assessed, likely affecting the Arctic picoeukaryote community structure both in the short term (seasonal cycles) and long term (global warming).


Assuntos
Clorófitas/virologia , Phycodnaviridae/fisiologia , Phycodnaviridae/efeitos da radiação , Replicação Viral/efeitos da radiação , Regiões Árticas , DNA Viral/genética , Phycodnaviridae/genética , Temperatura
17.
mBio ; 8(1)2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28174309

RESUMO

Compared with other flaviviruses, Zika virus (ZIKV) is uniquely associated with congenital diseases in pregnant women. One recent study reported that (i) ZIKV has higher thermostability than dengue virus (DENV [a flavivirus closely related to ZIKV]), which might contribute to the disease outcome; (ii) the higher thermostability of ZIKV could arise from an extended loop structure in domain III of the viral envelope (E) protein and an extra hydrogen-bond interaction between E molecules (V. A. Kostyuchenko, E. X. Y. Lim, S. Zhang, G. Fibriansah, T.-S. Ng, J. S. G. Ooi, J. Shi, and S.-M. Lok, Nature 533:425-428, 2016, https://doi.org/10.1038/nature17994). Here we report the functional analysis of the structural information in the context of complete ZIKV and DENV-2 virions. Swapping the prM-E genes between ZIKV and DENV-2 switched the thermostability of the chimeric viruses, identifying the prM-E proteins as the major determinants for virion thermostability. Shortening the extended loop of the E protein by 1 amino acid was lethal for ZIKV assembly/release. Mutations (Q350I and T351V) that abolished the extra hydrogen-bond interaction between the E proteins did not reduce ZIKV thermostability, indicating that the extra interaction does not increase the thermostability. Interestingly, mutant T351V was attenuated in A129 mice defective in type I interferon receptors, even though the virus retained the wild-type thermostability. Furthermore, we found that a chimeric ZIKV with the DENV-2 prM-E and a chimeric DENV-2 with the ZIKV prM-E were highly attenuated in A129 mice; these chimeric viruses were highly immunogenic and protective against DENV-2 and ZIKV challenge, respectively. These results indicate the potential of these chimeric viruses for vaccine development. IMPORTANCE: Analysis of a recently observed high-resolution structure of ZIKV led to a hypothesis that its unusual stability may contribute to the associated, unique disease outcomes. Here we performed a functional analysis to demonstrate that viral prM-E genes are the main determinants for the high stability of ZIKV. The extra hydrogen-bond interaction (observed in the high-resolution structure) between ZIKV E proteins did not enhance virion stability, whereas the extended loop of E protein (CD loop in domain III) was essential for ZIKV assembly. More importantly, we found that a chimeric ZIKV with DENV-2 prM-E genes and a chimeric DENV-2 with ZIKV prM-E genes were highly attenuated in A129 mice. Mice immunized with these chimeric viruses generated robust neutralizing antibody responses and were fully protected from DENV-2 and ZIKV challenge, respectively, indicating that these chimeric viruses could be further developed as vaccine candidates.


Assuntos
Vírus da Dengue/imunologia , Vírus da Dengue/fisiologia , Vacinas Virais/imunologia , Vacinas Virais/isolamento & purificação , Replicação Viral/efeitos da radiação , Zika virus/imunologia , Zika virus/fisiologia , Substituição de Aminoácidos , Animais , Análise Mutacional de DNA , Dengue/prevenção & controle , Vírus da Dengue/genética , Vírus da Dengue/efeitos da radiação , Modelos Animais de Doenças , Camundongos , Recombinação Genética , Deleção de Sequência , Temperatura , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/isolamento & purificação , Proteínas do Envelope Viral/genética , Vacinas Virais/genética , Virulência , Zika virus/genética , Zika virus/efeitos da radiação , Infecção por Zika virus/prevenção & controle
18.
PLoS One ; 11(10): e0165375, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27788196

RESUMO

Cyanophage infecting the marine cyanobacteria Prochlorococcus and Synechococcus require light and host photosystem activity for optimal reproduction. Many cyanophages encode multiple photosynthetic electron transport (PET) proteins, which are presumed to maintain electron flow and produce ATP and NADPH for nucleotide biosynthesis and phage genome replication. However, evidence suggests phage augment NADPH production via the pentose phosphate pathway (PPP), thus calling into question the need for NADPH production by PET. Genes implicated in cyclic PET have since been identified in cyanophage genomes. It remains an open question which mode of PET, cyclic or linear, predominates in infected cyanobacteria, and thus whether the balance is towards producing ATP or NADPH. We sequenced transcriptomes of a cyanophage (P-HM2) and its host (Prochlorococcus MED4) throughout infection in the light or in the dark, and analyzed these data in the context of phage replication and metabolite measurements. Infection was robust in the light, but phage were not produced in the dark. Host gene transcripts encoding high-light inducible proteins and two terminal oxidases (plastoquinol terminal oxidase and cytochrome c oxidase)-implicated in protecting the photosynthetic membrane from light stress-were the most enriched in light but not dark infection. Among the most diminished transcripts in both light and dark infection was ferredoxin-NADP+ reductase (FNR), which uses the electron acceptor NADP+ to generate NADPH in linear photosynthesis. The phage gene for CP12, which putatively inhibits the Calvin cycle enzyme that receives NADPH from FNR, was highly expressed in light infection. Therefore, both PET production of NADPH and its consumption by carbon fixation are putatively repressed during phage infection in light. Transcriptomic evidence is thus consistent with cyclic photophosphorylation using oxygen as the terminal electron acceptor as the dominant mode of PET under infection, with ATP from PET and NADPH from the PPP producing the energy and reducing equivalents for phage nucleotide biosynthesis and replication.


Assuntos
Bacteriófagos/fisiologia , Cianobactérias/virologia , Escuridão , Prochlorococcus/genética , Prochlorococcus/virologia , Transcriptoma/efeitos da radiação , Bacteriófagos/genética , Bacteriófagos/efeitos da radiação , Transporte de Elétrons/efeitos da radiação , Fotossíntese/efeitos da radiação , Prochlorococcus/metabolismo , Replicação Viral/efeitos da radiação
19.
J Virol ; 90(7): 3735-44, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26819307

RESUMO

UNLABELLED: Rift Valley fever (RVF) is endemic to Africa, and the mosquito-borne disease is characterized by "abortion storms" in ruminants and by hemorrhagic fever, encephalitis, and blindness in humans. Rift Valley fever virus (RVFV; family Bunyaviridae, genus Phlebovirus) has a tripartite negative-stranded RNA genome (L, M, and S segments). A live-attenuated vaccine for RVF, the MP-12 vaccine, is conditionally licensed for veterinary use in the United States. MP-12 is fully attenuated by the combination of the partially attenuated L, M, and S segments. Temperature sensitivity (ts) limits viral replication at a restrictive temperature and may be involved with viral attenuation. In this study, we aimed to characterize the ts mutations for MP-12. The MP-12 vaccine showed restricted replication at 38°C and replication shutoff (100-fold or greater reduction in virus titer compared to that at 37°C) at 39°C in Vero and MRC-5 cells. Using rZH501 reassortants with either the MP-12 L, M, or S segment, we found that all three segments encode a temperature-sensitive phenotype. However, the ts phenotype of the S segment was weaker than that of the M or L segment. We identified Gn-Y259H, Gc-R1182G, L-V172A, and L-M1244I as major ts mutations for MP-12. The ts mutations in the L segment decreased viral RNA synthesis, while those in the M segment delayed progeny production from infected cells. We also found that a lack of NSs and/or 78kD/NSm protein expression minimally affected the ts phenotype. Our study revealed that MP-12 is a unique vaccine carrying ts mutations in the L, M, and S segments. IMPORTANCE: Rift Valley fever (RVF) is a mosquito-borne viral disease endemic to Africa, characterized by high rates of abortion in ruminants and severe diseases in humans. Vaccination is important to prevent the spread of disease, and a live-attenuated MP-12 vaccine is currently the only vaccine with a conditional license in the United States. This study determined the temperature sensitivity (ts) of MP-12 vaccine to understand virologic characteristics. Our study revealed that MP-12 vaccine contains ts mutations independently in the L, M, and S segments and that MP-12 displays a restrictive replication at 38°C.


Assuntos
Vírus da Febre do Vale do Rift/fisiologia , Vírus da Febre do Vale do Rift/efeitos da radiação , Vacinas Virais/genética , Replicação Viral/efeitos da radiação , Animais , Linhagem Celular , Análise Mutacional de DNA , Humanos , Mutação de Sentido Incorreto , Vírus da Febre do Vale do Rift/genética , Temperatura , Vacinas Atenuadas/genética
20.
AIDS Res Hum Retroviruses ; 32(2): 120-4, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26765533

RESUMO

The use of highly active antiretroviral therapy against HIV-1 for last two decades has reduced mortality of patients through extension of nonsymptomatic phase of infection. However, HIV-1 can be preserved in long-lived resting CD4(+) T cells, which form a viral reservoir in infected individuals, and potentially in macrophages and astrocytes. Reactivation of viral replication is critical since the host immune response in combination with antiretroviral therapy may eradicate the virus (shock and kill strategy). In this opinion piece, we consider potential application of therapeutic doses of irradiation, the well-known and effective stress signal that induces DNA damage and activates cellular stress response, to resolve two problems: activate HIV-1 replication and virion production in persistent reservoirs under cART and deplete infected cells through selective cell killing using DNA damage responses.


Assuntos
Dano ao DNA/efeitos da radiação , Infecções por HIV/radioterapia , HIV-1/efeitos da radiação , Ativação Viral/efeitos da radiação , Latência Viral/efeitos da radiação , Replicação Viral/efeitos da radiação , Fármacos Anti-HIV/uso terapêutico , Terapia Antirretroviral de Alta Atividade , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/virologia , Reparo do DNA/efeitos da radiação , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/efeitos dos fármacos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA