Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(37): e2304722120, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37669378

RESUMO

Crimean-Congo hemorrhagic fever (CCHF) caused by CCHF virus (CCHFV) is one of the epidemic-prone diseases prioritized by the World Health Organisation as public health emergency with an urgent need for accelerated research. The trajectory of host response against CCHFV is multifarious and remains unknown. Here, we reported the temporal spectrum of pathogenesis following the CCHFV infection using genome-wide blood transcriptomics analysis followed by advanced systems biology analysis, temporal immune-pathogenic alterations, and context-specific progressive and postinfection genome-scale metabolic models (GSMM) on samples collected during the acute (T0), early convalescent (T1), and convalescent-phase (T2). The interplay between the retinoic acid-inducible gene-I-like/nucleotide-binding oligomerization domain-like receptor and tumor necrosis factor signaling governed the trajectory of antiviral immune responses. The rearrangement of intracellular metabolic fluxes toward the amino acid metabolism and metabolic shift toward oxidative phosphorylation and fatty acid oxidation during acute CCHFV infection determine the pathogenicity. The upregulation of the tricarboxylic acid cycle during CCHFV infection, compared to the noninfected healthy control and between the severity groups, indicated an increased energy demand and cellular stress. The upregulation of glycolysis and pyruvate metabolism potentiated energy generation through alternative pathways associated with the severity of the infection. The downregulation of metabolic processes at the convalescent phase identified by blood cell transcriptomics and single-cell type proteomics of five immune cells (CD4+ and CD8+ T cells, CD14+ monocytes, B cells, and NK cells) potentially leads to metabolic rewiring through the recovery due to hyperactivity during the acute phase leading to post-viral fatigue syndrome.


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Humanos , Linfócitos T CD8-Positivos , Regulação para Cima , Metaboloma
2.
EBioMedicine ; 90: 104523, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36933409

RESUMO

BACKGROUND: The tick-borne bunyavirus, Crimean-Congo Haemorrhagic Fever virus (CCHFV), can cause severe febrile illness in humans and has a wide geographic range that continues to expand due to tick migration. Currently, there are no licensed vaccines against CCHFV for widespread usage. METHODS: In this study, we describe the preclinical assessment of a chimpanzee adenoviral vectored vaccine (ChAdOx2 CCHF) which encodes the glycoprotein precursor (GPC) from CCHFV. FINDINGS: We demonstrate here that vaccination with ChAdOx2 CCHF induces both a humoral and cellular immune response in mice and 100% protection in a lethal CCHF challenge model. Delivery of the adenoviral vaccine in a heterologous vaccine regimen with a Modified Vaccinia Ankara vaccine (MVA CCHF) induces the highest levels of CCHFV-specific cell-mediated and antibody responses in mice. Histopathological examination and viral load analysis of the tissues of ChAdOx2 CCHF immunised mice reveals an absence of both microscopic changes and viral antigen associated with CCHF infection, further demonstrating protection against disease. INTERPRETATION: There is the continued need for an effective vaccine against CCHFV to protect humans from lethal haemorrhagic disease. Our findings support further development of the ChAd platform expressing the CCHFV GPC to seek an effective vaccine against CCHFV. FUNDING: This research was supported by funding from the Biotechnology and Biological Sciences Research Council (UKRI-BBSRC) [BB/R019991/1 and BB/T008784/1].


Assuntos
Vírus da Febre Hemorrágica da Crimeia-Congo , Febre Hemorrágica da Crimeia , Vacinas Virais , Humanos , Animais , Camundongos , Febre Hemorrágica da Crimeia/prevenção & controle , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vacinação , Vetores Genéticos/genética , Vaccinia virus
3.
J Gen Virol ; 103(10)2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36215163

RESUMO

In Africa, several emerging zoonotic viruses have been transmitted from small mammals such as rodents and shrews to humans. Although no clinical cases of small mammal-borne viral diseases have been reported in Central Africa, potential zoonotic viruses have been identified in rodents in the region. Therefore, we hypothesized that there may be unrecognized zoonotic viruses circulating in small mammals in Central Africa. Here, we investigated viruses that have been maintained among wild small mammals in Gabon to understand their potential risks to humans. We identified novel orthonairoviruses in 24.6 % of captured rodents and shrews from their kidney total RNA samples. Phylogenetic analysis revealed that the novel viruses, Lamusara virus (LMSV) and Lamgora virus, were closely related to Erve virus, which was previously identified in shrews of the genus Crocidura and has been suspected to cause neuropathogenic diseases in humans. Moreover, we show that the LMSV ovarian tumour domain protease, one of the virulence determination factors of orthonairoviruses, suppressed interferon signalling in human cells, suggesting the possible human pathogenicity of this virus. Taken together, our study demonstrates the presence of novel orthonairoviruses that may pose unrecognized risks of viral disease transmission in Gabon.


Assuntos
Roedores , Musaranhos , Vírus , Animais , Gabão/epidemiologia , Interferons/genética , Peptídeo Hidrolases , Filogenia , RNA , Roedores/virologia , Musaranhos/virologia , Vírus/genética
4.
Sci Rep ; 10(1): 21431, 2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33293534

RESUMO

In the event of an unpredictable viral outbreak requiring high/maximum biosafety containment facilities (i.e. BSL3 and BSL4), X-ray irradiation has the potential to relieve pressures on conventional diagnostic bottlenecks and expediate work at lower containment. Guided by Monte Carlo modelling and in vitro 1-log10 decimal-reduction value (D-value) predictions, the X-ray photon energies required for the effective inactivation of zoonotic viruses belonging to the medically important families of Flaviviridae, Nairoviridae, Phenuiviridae and Togaviridae are demonstrated. Specifically, it is shown that an optimized irradiation approach is attractive for use in a multitude of downstream detection and functional assays, as it preserves key biochemical and immunological properties. This study provides evidence that X-ray irradiation can support emergency preparedness, outbreak response and front-line diagnostics in a safe, reproducible and scalable manner pertinent to operations that are otherwise restricted to higher containment BSL3 or BSL4 laboratories.


Assuntos
Vírus de RNA/fisiologia , RNA Viral/genética , Inativação de Vírus , Raios X/efeitos adversos , Animais , Chlorocebus aethiops , Defesa Civil , Contenção de Riscos Biológicos , Células Alimentadoras , Humanos , Método de Monte Carlo , Nairovirus/fisiologia , Nairovirus/efeitos da radiação , Vírus de RNA/efeitos da radiação , RNA Viral/efeitos da radiação , Análise de Sequência de RNA , Togaviridae/fisiologia , Togaviridae/efeitos da radiação , Células Vero , Zoonoses Virais/prevenção & controle , Zika virus/fisiologia , Zika virus/efeitos da radiação
5.
Sci Rep ; 10(1): 14289, 2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32868837

RESUMO

Ebola virus (EBOV) is an enveloped, single-stranded RNA virus that can cause Ebola virus disease (EVD). It is thought that EVD survivors are protected against subsequent infection with EBOV and that neutralising antibodies to the viral surface glycoprotein (GP) are potential correlates of protection. Serological studies are vital to assess neutralising antibodies targeted to EBOV GP; however, handling of EBOV is limited to containment level 4 laboratories. Pseudotyped viruses can be used as alternatives to live viruses, which require high levels of bio-containment, in serological and viral entry assays. However, neutralisation capacity can differ among pseudotyped virus platforms. We evaluated the suitability of EBOV GP pseudotyped human immunodeficiency virus type 1 (HIV-1) and vesicular stomatitis virus (VSV) to measure the neutralising ability of plasma from EVD survivors, when compared to results from a live EBOV neutralisation assay. The sensitivity, specificity and correlation with live EBOV neutralisation were greater for the VSV-based pseudotyped virus system, which is particularly important when evaluating EBOV vaccine responses and immuno-therapeutics. Therefore, the EBOV GP pseudotyped VSV neutralisation assay reported here could be used to provide a better understanding of the putative correlates of protection against EBOV.


Assuntos
Anticorpos Neutralizantes/imunologia , Ebolavirus/imunologia , HIV-1/imunologia , Vesiculovirus/imunologia , Proteínas do Envelope Viral/imunologia , Humanos , Testes de Neutralização , Tropismo Viral/imunologia
6.
Vaccines (Basel) ; 8(2)2020 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-32560145

RESUMO

The flavivirus envelope protein domain III (EDIII) was an effective immunogen against dengue virus (DENV) and other related flaviviruses. Whether this can be applied to the Zika virus (ZIKV) vaccinology remains an open question. Here, we tested the efficacy of ZIKV-EDIII against ZIKV infection, using several vaccine platforms that present the antigen in various ways. We provide data demonstrating that mice vaccinated with a ZIKV-EDIII as DNA or protein-based vaccines failed to raise fully neutralizing antibodies and did not control viremia, following a ZIKV challenge, despite eliciting robust antibody responses. Furthermore, we showed that ZIKV-EDIII encoded in replication-deficient Chimpanzee adenovirus (ChAdOx1-EDIII) elicited anti-ZIKV envelope antibodies in vaccinated mice but also provided limited protection against ZIKV in two physiologically different mouse challenge models. Taken together, our data indicate that contrary to what was shown for other flaviviruses like the dengue virus, which has close similarities with ZIKV-EDIII, this antigen might not be a suitable vaccine candidate for the correct induction of protective immune responses against ZIKV.

7.
Vaccines (Basel) ; 8(2)2020 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32455764

RESUMO

In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leaving individuals susceptible to disease after exposure to related virus species. This is a significant hurdle in the field of vaccine development, in which broadly protective vaccines represent an unmet need. This is particularly evident for filoviruses, as there are multiple family members that can cause lethal haemorrhagic fever, including Zaire ebolavirus, Sudan ebolavirus, and Marburg virus. In an attempt to address this need, both pre-clinical and clinical studies previously used mixed or co-administered monovalent vaccines to prevent filovirus mediated disease. However, these multi-vaccine and multi-dose vaccination regimens do not represent a practical immunisation scheme when considering the target endemic areas. We describe here the development of a single multi-pathogen filovirus vaccine candidate based on a replication-deficient simian adenoviral vector. Our vaccine candidate encodes three different filovirus glycoproteins in one vector and induces strong cellular and humoral immunity to all three viral glycoproteins after a single vaccination. Crucially, it was found to be protective in a stringent Zaire ebolavirus challenge in guinea pigs in a one-shot vaccination regimen. This trivalent filovirus vaccine offers a tenable vaccine product that could be rapidly translated to the clinic to prevent filovirus-mediated viral haemorrhagic fever.

8.
Viruses ; 11(11)2019 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-31689981

RESUMO

Recent studies have shown that transcriptomic analysis of blood samples taken from patients with acute Ebola virus disease (EVD) during the 2013-2016 West African outbreak was suggestive that a severe inflammatory response took place in acutely ill patients. The significant knowledge gained from studying the Makona variant, a cause of the largest known EVD outbreak, may be applicable to other species of ebolavirus, and other variants of the Ebola virus (EBOV) species. To investigate the ability of Makona to initiate an inflammatory response in human macrophages and characterise the host response in a similar manner to previously characterised EBOV variants, the human monocytic cell line THP-1 was differentiated into macrophage-like cells and infected with Makona. RNA-Seq and quantitative proteomics were used to identify and quantify host mRNA and protein abundance during infection. Data from infection with Reston virus (RESTV) were used as comparators to investigate changes that may be specific to, or enhanced in, Makona infection in relation to a less pathogenic species of ebolavirus.. This study found demonstrable induction of the inflammatory response, and increase in the activation state of THP-1 macrophages infected with Makona. NFκB and inflammation-associated transcripts displayed significant changes in abundance, reflective of what was observed in human patients during the 2013-2016 EBOV outbreak in West Africa, and demonstrated that transcriptomic changes found in Makona-infected cells were similar to that observed in Reston virus infection and that have been described in previous studies of other variants of EBOV.


Assuntos
Ebolavirus/isolamento & purificação , Ebolavirus/patogenicidade , Citocinas/genética , Citocinas/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Doença pelo Vírus Ebola/virologia , Humanos , Interferons/genética , Interferons/metabolismo , Macrófagos/imunologia , Macrófagos/virologia , Proteômica , Células THP-1
9.
J Biol Chem ; 294(18): 7335-7347, 2019 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-30804209

RESUMO

The Bunyavirales order of segmented negative-sense RNA viruses includes more than 500 isolates that infect insects, animals, and plants and are often associated with severe and fatal disease in humans. To multiply and cause disease, bunyaviruses must translocate their genomes from outside the cell into the cytosol, achieved by transit through the endocytic network. We have previously shown that the model bunyaviruses Bunyamwera virus (BUNV) and Hazara virus (HAZV) exploit the changing potassium concentration ([K+]) of maturing endosomes to release their genomes at the appropriate endosomal location. K+ was identified as a biochemical cue to activate the viral fusion machinery, promoting fusion between viral and cellular membranes, consequently permitting genome release. In this study, we further define the biochemical prerequisites for BUNV and HAZV entry and their K+ dependence. Using drug-mediated cholesterol extraction along with viral entry and K+ uptake assays, we report three major findings: BUNV and HAZV require cellular cholesterol during endosomal escape; cholesterol depletion from host cells impairs K+ accumulation in maturing endosomes, revealing new insights into endosomal K+ homeostasis; and "priming" BUNV and HAZV virions with K+ before infection alleviates their cholesterol requirement. Taken together, our findings suggest a model in which cholesterol abundance influences endosomal K+ levels and, consequently, the efficiency of bunyavirus infection. The ability to inhibit bunyaviruses with existing cholesterol-lowering drugs may offer new options for future antiviral interventions for pathogenic bunyaviruses.


Assuntos
Colesterol/metabolismo , Endossomos/metabolismo , Orthobunyavirus/fisiologia , Potássio/metabolismo , Internalização do Vírus , Linhagem Celular Tumoral , Endocitose , Humanos , Transporte de Íons , Vírion/fisiologia
10.
Mem. Inst. Oswaldo Cruz ; 114: e180586, 2019. graf
Artigo em Inglês | LILACS | ID: biblio-1002682

RESUMO

Here, we report the complete genome sequence of the Aporé virus (Bunyavirales: Arenaviridae), obtained from a wild rodent Oligoryzomys mattogrossae captured in Mato Grosso do Sul state, Brazil. The genome of this virus showed strong similarity to highly pathogenic mammarenavirus from South America.


Assuntos
Humanos , Oligorribonucleotídeos/provisão & distribuição , Arenaviridae , Arenavirus , Brasil/epidemiologia
11.
Cell Rep ; 25(13): 3750-3758.e4, 2018 12 26.
Artigo em Inglês | MEDLINE | ID: mdl-30590046

RESUMO

The Gn subcomponent of the Gn-Gc assembly that envelopes the human and animal pathogen, Rift Valley fever virus (RVFV), is a primary target of the neutralizing antibody response. To better understand the molecular basis for immune recognition, we raised a class of neutralizing monoclonal antibodies (nAbs) against RVFV Gn, which exhibited protective efficacy in a mouse infection model. Structural characterization revealed that these nAbs were directed to the membrane-distal domain of RVFV Gn and likely prevented virus entry into a host cell by blocking fusogenic rearrangements of the Gn-Gc lattice. Genome sequence analysis confirmed that this region of the RVFV Gn-Gc assembly was under selective pressure and constituted a site of vulnerability on the virion surface. These data provide a blueprint for the rational design of immunotherapeutics and vaccines capable of preventing RVFV infection and a model for understanding Ab-mediated neutralization of bunyaviruses more generally.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Antivirais/farmacologia , Vírus da Febre do Vale do Rift/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/farmacologia , Chlorocebus aethiops , Feminino , Glicoproteínas/química , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Imunização , Imunoglobulina G/metabolismo , Camundongos Endogâmicos BALB C , Modelos Biológicos , Testes de Neutralização , Domínios Proteicos , Coelhos , Proteínas Recombinantes/farmacologia , Vírus da Febre do Vale do Rift/efeitos dos fármacos , Células Vero , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
12.
J Biol Chem ; 293(26): 9937-9944, 2018 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-29678879

RESUMO

Many enveloped viruses enter cells through the endocytic network, from which they must subsequently escape through fusion of viral and endosomal membranes. This membrane fusion is mediated by virus-encoded spikes that respond to the dynamic endosomal environment, which triggers conformational changes in the spikes that initiate the fusion process. Several fusion triggers have been identified and include pH, membrane composition, and endosome-resident proteins, and these cues dictate when and where viral fusion occurs. We recently reported that infection with an enveloped bunyavirus requires elevated potassium ion concentrations [K+], controlled by cellular K+ channels, that are encountered during viral transit through maturing endosomes. Here we reveal the molecular basis for the K+ requirement of bunyaviruses through the first direct visualization of a member of the Nairoviridae family, namely Hazara virus (HAZV), using cryo-EM. Using cryo-electron tomography, we observed HAZV spike glycoproteins within infectious HAZV particles exposed to both high and low [K+], which showed that exposure to K+ alone results in dramatic changes to the ultrastructural architecture of the virion surface. In low [K+], the spikes adopted a compact conformation arranged in locally ordered arrays, whereas, following exposure to high [K+], the spikes became extended, and spike-membrane interactions were observed. Viruses exposed to high [K+] also displayed enhanced infectivity, thus identifying K+ as a newly defined trigger that helps promote viral infection. Finally, we confirmed that K+ channel blockers are inhibitory to HAZV infection, highlighting the potential of K+ channels as anti-bunyavirus targets.


Assuntos
Orthobunyavirus/efeitos dos fármacos , Orthobunyavirus/fisiologia , Potássio/farmacologia , Internalização do Vírus/efeitos dos fármacos , Células A549 , Relação Dose-Resposta a Droga , Humanos , Orthobunyavirus/metabolismo , Canais de Potássio/metabolismo , Conformação Proteica/efeitos dos fármacos , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
13.
Cell ; 171(1): 229-241.e15, 2017 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-28938115

RESUMO

Zika virus (ZIKV), a mosquito-borne flavivirus, causes devastating congenital birth defects. We isolated a human monoclonal antibody (mAb), ZKA190, that potently cross-neutralizes multi-lineage ZIKV strains. ZKA190 is highly effective in vivo in preventing morbidity and mortality of ZIKV-infected mice. NMR and cryo-electron microscopy show its binding to an exposed epitope on DIII of the E protein. ZKA190 Fab binds all 180 E protein copies, altering the virus quaternary arrangement and surface curvature. However, ZIKV escape mutants emerged in vitro and in vivo in the presence of ZKA190, as well as of other neutralizing mAbs. To counter this problem, we developed a bispecific antibody (FIT-1) comprising ZKA190 and a second mAb specific for DII of E protein. In addition to retaining high in vitro and in vivo potencies, FIT-1 robustly prevented viral escape, warranting its development as a ZIKV immunotherapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/uso terapêutico , Infecção por Zika virus/terapia , Zika virus/química , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/química , Anticorpos Antivirais/administração & dosagem , Anticorpos Antivirais/química , Microscopia Crioeletrônica , Epitopos , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Modelos Moleculares , Alinhamento de Sequência , Proteínas do Envelope Viral/química , Zika virus/imunologia
14.
J Virol ; 90(20): 9305-16, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27512070

RESUMO

UNLABELLED: The Nairovirus genus of the Bunyaviridae family contains serious human and animal pathogens classified within multiple serogroups and species. Of these serogroups, the Crimean-Congo hemorrhagic fever virus (CCHFV) serogroup comprises sole members CCHFV and Hazara virus (HAZV). CCHFV is an emerging zoonotic virus that causes often-fatal hemorrhagic fever in infected humans for which preventative or therapeutic strategies are not available. In contrast, HAZV is nonpathogenic to humans and thus represents an excellent model to study aspects of CCHFV biology under conditions of more-accessible biological containment. The three RNA segments that form the nairovirus genome are encapsidated by the viral nucleocapsid protein (N) to form ribonucleoprotein (RNP) complexes that are substrates for RNA synthesis and packaging into virus particles. We used quantitative proteomics to identify cellular interaction partners of CCHFV N and identified robust interactions with cellular chaperones. These interactions were validated using immunological methods, and the specific interaction between native CCHFV N and cellular chaperones of the HSP70 family was confirmed during live CCHFV infection. Using infectious HAZV, we showed for the first time that the nairovirus N-HSP70 association was maintained within both infected cells and virus particles, where N is assembled as RNPs. Reduction of active HSP70 levels in cells by the use of small-molecule inhibitors significantly reduced HAZV titers, and a model for chaperone function in the context of high genetic variability is proposed. These results suggest that chaperones of the HSP70 family are required for nairovirus replication and thus represent a genetically stable cellular therapeutic target for preventing nairovirus-mediated disease. IMPORTANCE: Nairoviruses compose a group of human and animal viruses that are transmitted by ticks and associated with serious or fatal disease. One member is Crimean-Congo hemorrhagic fever virus (CCHFV), which is responsible for fatal human disease and is recognized as an emerging threat within Europe in response to climate change. No preventative or therapeutic strategies against nairovirus-mediated disease are currently available. Here we show that the N protein of CCHFV and the related Hazara virus interact with a cellular protein, HSP70, during both the intracellular and extracellular stages of the virus life cycle. The use of inhibitors that block HSP70 function reduces virus titers by up to 1,000-fold, suggesting that this interaction is important within the context of the nairovirus life cycle and may represent a potent target for antinairovirus therapies against which the virus cannot easily develop resistance.


Assuntos
Proteínas de Choque Térmico HSP70/metabolismo , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Vírus da Febre Hemorrágica da Crimeia-Congo/metabolismo , Nairovirus/genética , Nairovirus/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Replicação Viral/genética , Células A549 , Linhagem Celular , Linhagem Celular Tumoral , Mudança Climática , Europa (Continente) , Células HEK293 , Febre Hemorrágica da Crimeia/metabolismo , Febre Hemorrágica da Crimeia/virologia , Humanos , RNA/genética
15.
Science ; 353(6301): 823-6, 2016 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-27417494

RESUMO

Zika virus (ZIKV), a mosquito-borne flavivirus with homology to Dengue virus (DENV), has become a public health emergency. By characterizing memory lymphocytes from ZIKV-infected patients, we dissected ZIKV-specific and DENV-cross-reactive immune responses. Antibodies to nonstructural protein 1 (NS1) were largely ZIKV-specific and were used to develop a serological diagnostic tool. In contrast, antibodies against E protein domain I/II (EDI/II) were cross-reactive and, although poorly neutralizing, potently enhanced ZIKV and DENV infection in vitro and lethally enhanced DENV disease in mice. Memory T cells against NS1 or E proteins were poorly cross-reactive, even in donors preexposed to DENV. The most potent neutralizing antibodies were ZIKV-specific and targeted EDIII or quaternary epitopes on infectious virus. An EDIII-specific antibody protected mice from lethal ZIKV infection, illustrating the potential for antibody-based therapy.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Infecção por Zika virus/imunologia , Zika virus/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/uso terapêutico , Especificidade de Anticorpos , Reações Cruzadas , Vírus da Dengue/imunologia , Modelos Animais de Doenças , Humanos , Epitopos Imunodominantes/imunologia , Memória Imunológica , Estrutura Terciária de Proteína , Linfócitos T/imunologia , Proteínas do Envelope Viral/imunologia , Proteínas não Estruturais Virais/imunologia , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/terapia
16.
J Proteome Res ; 13(11): 5120-35, 2014 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-25158218

RESUMO

Viral pathogenesis in the infected cell is a balance between antiviral responses and subversion of host-cell processes. Many viral proteins specifically interact with host-cell proteins to promote virus biology. Understanding these interactions can lead to knowledge gains about infection and provide potential targets for antiviral therapy. One such virus is Ebola, which has profound consequences for human health and causes viral hemorrhagic fever where case fatality rates can approach 90%. The Ebola virus VP24 protein plays a critical role in the evasion of the host immune response and is likely to interact with multiple cellular proteins. To map these interactions and better understand the potential functions of VP24, label-free quantitative proteomics was used to identify cellular proteins that had a high probability of forming the VP24 cellular interactome. Several known interactions were confirmed, thus placing confidence in the technique, but new interactions were also discovered including one with ATP1A1, which is involved in osmoregulation and cell signaling. Disrupting the activity of ATP1A1 in Ebola-virus-infected cells with a small molecule inhibitor resulted in a decrease in progeny virus, thus illustrating how quantitative proteomics can be used to identify potential therapeutic targets.


Assuntos
Ebolavirus/patogenicidade , Mapeamento de Interação de Proteínas/métodos , ATPase Trocadora de Sódio-Potássio/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/virologia , Ebolavirus/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293/efeitos dos fármacos , Células HEK293/virologia , Interações Hospedeiro-Patógeno , Humanos , Espectrometria de Massas/métodos , Ouabaína/farmacologia , Proteômica/métodos , Reprodutibilidade dos Testes , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Proteínas Virais/genética
17.
Rev. colomb. cienc. pecu ; 22(3): 267-277, Sep. 2009.
Artigo em Inglês | LILACS | ID: lil-559516

RESUMO

This manuscript is an inedited part of my PhD dissertation, based on historical and recent findingson animal models, that was presented as part of the requirements to fulfill the conditions to become aphilosophical doctor on Veterinary Sciences at the University of Wisconsin on October of 2003.The currentmini-review written on a free-version style, underlines some of the cornerstones of immunology as ascience, understood thanks to the use of the Lymphocytic Choriomeningitis virus (LCMV) experimentallyand naturally infected mouse model. It should suffice to say that there have been two Nobel prices ofMedicine for discoveries made through the employment of this animal model, in order to recognize theright importance to it. In addition, several laboratories, Dr. Salvato´s among them, have also employed theLCMV-infected Rhesus monkey model as a tool to unravel the mysteries of arenaviral hemorrhagic fever,and particularly the physiopathology of Lassa disease in humans. Here I show some of the knowledgegenerated through the study of both animal infections.


El siguiente manuscrito, es un capítulo inédito de mi tesis doctoral, basado en hallazgos históricos yrecientes sobre modelos animales, que fue presentado como parte de los requisitos para obtener el títulode Ph.D. en Ciencias veterinarias en la Universidad de Wisconsin, en Octubre de 2003.La actual minirevisiónescrita en estilo de versión libre, subraya algunas de las piedras angulares de la inmunología comociencia, entendidas gracias al uso del modelo murino infectado natural y experimentalmente con el virusde Coriomeningitis Linfocítica (LCMV). Sería suficiente mencionar que han existido dos premios Nóbelde Medicina por descubrimientos realizados a través del empleo de este modelo animal, para reconocer lareal importancia del mismo. Adicionalmente, varios laboratorios, el de la Dra. Salvato entre ellos, tambiénhan empleado el modelo del mono Rhesus como un instrumento para desvelar los misterios de las fiebreshemorrágicas por arenavirus, y particularmente la fisiopatología de la enfermedad de Lassa en humanos.Aquí yo muestro alo del conocimiento generado a través del estudio de ambas infecciones animales.


O seguinte manuscrito é um capítulo inédito da minha tese doutoral, baseado em casos históricose recentes sobre modelos animais, que foram apresentados na defesa da tese de Ph.D. em CiênciasVeterinárias da Universidade de Wisconsin, em outubro de 2003. A atual mini revisão escrita em estilode versão livre, enfatiza em algumas pedras angulares da inmunologìa como ciência. Graças ao usodo modelo murino infectado natural e experimentalmente com o vírus da Coriomeningitis Linfocítica(LCMV). Seria suficiente mencionar que foram outorgados dois prêmios Nobel de Medicina pelosdescobrimentos realizados a través do uso deste modelo, para reconhecer a real importância do mesmo.Adicionalmente, vários laboratórios, entre eles o da Dra. Salvato, tem utilizado o modelo macaco Rhesuscomo um instrumento para desvelar os mistérios das febres hemorrágicas por arenavirus, e particularmentea fisiologia e patologia da doença de Lassa em humanos. Aqui eu indico algo do conhecimento gerado através do estudo das duas infecções animais.


Assuntos
Animais , Primatas
18.
J Virol ; 77(8): 4805-17, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12663787

RESUMO

The assembly and budding of human immunodeficiency virus type 1 (HIV-1) at the plasma membrane are directed by the viral core protein Pr55(gag). We have analyzed whether Pr55(gag) has intrinsic affinity for sphingolipid- and cholesterol-enriched raft microdomains at the plasma membrane. Pr55(gag) has previously been reported to associate with Triton X-100-resistant rafts, since both intracellular membranes and virus-like Pr55(gag) particles (VLPs) yield buoyant Pr55(gag) complexes upon Triton X-100 extraction at cold temperatures, a phenotype that is usually considered to indicate association of a protein with rafts. However, we show here that the buoyant density of Triton X-100-treated Pr55(gag) complexes cannot be taken as a proof for raft association of Pr55(gag), since lipid analyses of Triton X-100-treated VLPs demonstrated that the detergent readily solubilizes the bulk of membrane lipids from Pr55(gag). However, Pr55(gag) might nevertheless be a raft-associated protein, since confocal fluorescence microscopy indicated that coalescence of GM1-positive rafts at the cell surface led to copatching of membrane-bound Pr55(gag). Furthermore, extraction of intracellular membranes or VLPs with Brij98 yielded buoyant Pr55(gag) complexes of low density. Lipid analyses of Brij98-treated VLPs suggested that a large fraction of the envelope cholesterol and phospholipids was resistant to Brij98. Collectively, these results suggest that Pr55(gag) localizes to membrane microdomains that are largely resistant to Brij98 but sensitive to Triton X-100, and these membrane domains provide the platform for assembly and budding of Pr55(gag) VLPs.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Microdomínios da Membrana/metabolismo , Precursores de Proteínas/metabolismo , Montagem de Vírus , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Detergentes/farmacologia , Resistência a Medicamentos , Humanos , Células Jurkat , Microdomínios da Membrana/efeitos dos fármacos , Microscopia Confocal , Octoxinol/farmacologia , Óleos de Plantas/farmacologia , Polietilenoglicóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA