Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
mSphere ; 5(4)2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32641430

RESUMO

Nipah disease is listed as one of the WHO priority diseases that pose the greatest public health risk due to their epidemic potential. More than 200 experts from around the world convened in Singapore last year to mark the 20th anniversary of the first Nipah virus outbreaks in Malaysia and Singapore. Most of these experts are now involved in responding to the coronavirus disease 2019 (COVID-19) pandemic. Here, members of the Organizing Committee of the 2019 Nipah Virus International Conference review highlights from the Nipah@20 Conference and reflect on key lessons learned from Nipah that could be applied to the understanding of the COVID-19 pandemic and to preparedness against future emerging infectious diseases (EIDs) of pandemic potential.


Assuntos
Infecções por Henipavirus , Vírus Nipah/patogenicidade , Animais , Betacoronavirus/patogenicidade , COVID-19 , Congressos como Assunto , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/prevenção & controle , Infecções por Coronavirus/terapia , Infecções por Henipavirus/diagnóstico , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/terapia , Humanos , Pandemias/prevenção & controle , Pneumonia Viral/diagnóstico , Pneumonia Viral/prevenção & controle , Pneumonia Viral/terapia , SARS-CoV-2 , Zoonoses/epidemiologia
2.
J Infect Dis ; 221(Suppl 4): S471-S479, 2020 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-31686101

RESUMO

BACKGROUND: Nipah virus (NiV) and Hendra virus (HeV) are zoonotic paramyxoviruses that cause severe disease in both animals and humans. There are no approved vaccines or treatments for use in humans; however, therapeutic treatment of both NiV and HeV infection in ferrets and non-human primates with a cross-reactive, neutralizing human monoclonal antibody (mAb), m102.4, targeting the G glycoprotein has been demonstrated. In a previous study, we isolated, characterized, and humanized a cross-reactive, neutralizing anti-F mAb (h5B3.1). The mAb h5B3.1 blocks the required F conformational change needed to facilitate membrane fusion and virus infection, and the epitope recognized by h5B3.1 has been structurally defined; however, the efficacy of h5B3.1 in vivo is unknown. METHODS: The post-infection antiviral activity of h5B3.1 was evaluated in vivo by administration in ferrets after NiV and HeV virus challenge. RESULTS: All subjects that received h5B3.1 from 1 to several days after infection with a high-dose, oral-nasal virus challenge were protected from disease, whereas all controls died. CONCLUSIONS: This is the first successful post-exposure antibody therapy for NiV and HeV using a humanized cross-reactive mAb targeting the F glycoprotein, and the findings suggest that a combination therapy targeting both F and G should be evaluated as a therapy for NiV/HeV infection.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Vírus Hendra , Infecções por Henipavirus/terapia , Vírus Nipah , Proteínas Virais de Fusão/imunologia , Animais , Reações Cruzadas , Furões , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/virologia , Humanos
3.
Infez Med ; 27(2): 117-127, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-31205033

RESUMO

The Nipah virus was discovered twenty years ago, and there is considerable information available regarding the specificities surrounding this virus such as transmission, pathogenesis and genome. Belonging to the Henipavirus genus, this virus can cause fever, encephalitis and respiratory disorders. The first cases were reported in Malaysia and Singapore in 1998, when affected individuals presented with severe febrile encephalitis. Since then, much has been identified about this virus. These single-stranded RNA viruses gain entry into target cells via a process known as macropinocytosis. The viral genome is released into the cell cytoplasm via a cascade of processes that involves conformational changes in G and F proteins which allow for attachment of the viral membrane to the cell membrane. In addition to this, the natural reservoirs of this virus have been identified to be fruit bats from the genus Pteropus. Five of the 14 species of bats in Malaysia have been identified as carriers, and this virus affects horses, cats, dogs, pigs and humans. Various mechanisms of transmission have been proposed such as contamination of date palm saps by bat feces and saliva, nosocomial and human-to-human transmissions. Physical contact was identified as the strongest risk factor for developing an infection in the 2004 Faridpur outbreak. Geographically, the virus seems to favor the Indian sub-continent, Indonesia, Southeast Asia, Pakistan, southern China, northern Australia and the Philippines, as demonstrated by the multiple outbreaks in 2001, 2004, 2007, 2012 in Bangladesh, India and Pakistan as well as the initial outbreaks in Malaysia and Singapore. Multiple routes of the viremic spread in the human body have been identified such as the central nervous system (CNS) and respiratory system, while virus levels in the body remain low, detection in the cerebrospinal fluid is comparatively high. The virus follows an incubation period of 4 days to 2 weeks which is followed by the development of symptoms. The primary clinical signs include fever, headache, vomiting and dizziness, while the characteristic symptoms consist of segmental myoclonus, tachycardia, areflexia, hypotonia, abnormal pupillary reflexes and hypertension. The serum neutralization test (SNT) is the gold standard of diagnosis followed by ELISA if SNT cannot be carried out. On the other hand, treatment is supportive since there a lack of effective pharmacological therapy and only one equine vaccine is currently licensed for use. Prevention of outbreaks seems to be a more viable approach until specific therapeutic strategies are devised.


Assuntos
Doenças Transmissíveis Emergentes/epidemiologia , Epidemias/estatística & dados numéricos , Infecções por Henipavirus/epidemiologia , Vírus Nipah , Animais , Ásia/epidemiologia , Gatos , Quirópteros/virologia , Doenças Transmissíveis Emergentes/terapia , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/veterinária , Reservatórios de Doenças , Cães , Infecções por Henipavirus/terapia , Infecções por Henipavirus/transmissão , Infecções por Henipavirus/veterinária , Cavalos , Humanos , Vírus Nipah/genética , Vírus Nipah/isolamento & purificação , Pinocitose , Suínos , Avaliação de Sintomas , Vacinação/métodos , Vacinação/veterinária , Internalização do Vírus
4.
Microbes Infect ; 21(7): 278-286, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30817995

RESUMO

Nipah virus is an emerging zoonotic paramyxovirus that causes severe and often fatal respiratory and neurological disease in humans. The virus was first discovered after an outbreak of encephalitis in pig farmers in Malaysia and Singapore with subsequent outbreaks in Bangladesh or India occurring almost annually. Due to the highly pathogenic nature of NiV, its pandemic potential, and the lack of licensed vaccines or therapeutics, there is a requirement for research and development into highly sensitive and specific diagnostic tools as well as antivirals and vaccines to help prevent and control future outbreak situations.


Assuntos
Infecções por Henipavirus/diagnóstico , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/imunologia , Vírus Nipah/isolamento & purificação , Animais , Antivirais/uso terapêutico , Técnicas de Laboratório Clínico , Surtos de Doenças/prevenção & controle , Infecções por Henipavirus/epidemiologia , Infecções por Henipavirus/terapia , Humanos , Modelos Biológicos , Vírus Nipah/genética , Vírus Nipah/patogenicidade , Vacinas Virais/imunologia , Zoonoses/epidemiologia , Zoonoses/transmissão
5.
Brain Pathol ; 25(5): 605-13, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26276024

RESUMO

The genus Henipavirus within the family Paramyxoviridae includes the Hendra virus (HeV) and Nipah virus (NiV) which were discovered in the 1990s in Australia and Malaysia, respectively, after emerging to cause severe and often fatal outbreaks in humans and animals. While HeV is confined to Australia, more recent NiV outbreaks have been reported in Bangladesh, India and the Philippines. The clinical manifestations of both henipaviruses in humans appear similar, with a predominance of an acute encephalitic syndrome. Likewise, the pathological features are similar and characterized by disseminated, multi-organ vasculopathy comprising endothelial infection/ulceration, vasculitis, vasculitis-induced thrombosis/occlusion, parenchymal ischemia/microinfarction, and parenchymal cell infection in the central nervous system (CNS), lung, kidney and other major organs. This unique dual pathogenetic mechanism of vasculitis-induced microinfarction and neuronal infection causes severe tissue damage in the CNS. Both viruses can also cause relapsing encephalitis months and years after the acute infection. Many animal models studied to date have largely confirmed the pathology of henipavirus infection, and provided the means to test new therapeutic agents and vaccines. As the bat is the natural host of henipaviruses and has worldwide distribution, spillover events into human populations are expected to occur in the future.


Assuntos
Encefalite Viral/diagnóstico , Infecções por Henipavirus/diagnóstico , Animais , Antivirais/uso terapêutico , Encéfalo/patologia , Encefalite Viral/patologia , Encefalite Viral/terapia , Vírus Hendra/patogenicidade , Infecções por Henipavirus/patologia , Infecções por Henipavirus/terapia , Humanos , Imunização Passiva , Vírus Nipah/patogenicidade
7.
Sci Transl Med ; 6(242): 242ra82, 2014 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-24964990

RESUMO

Nipah virus (NiV) is an emerging zoonotic paramyxovirus that causes severe and often fatal disease in pigs and humans. There are currently no vaccines or treatments approved for human use. Studies in small-animal models of NiV infection suggest that antibody therapy may be a promising treatment. However, most studies have assessed treatment at times shortly after virus exposure before animals show signs of disease. We assessed the efficacy of a fully human monoclonal antibody, m102.4, at several time points after virus exposure including at the onset of clinical illness in a uniformly lethal nonhuman primate model of NiV disease. Sixteen African green monkeys (AGMs) were challenged intratracheally with a lethal dose of NiV, and 12 animals were infused twice with m102.4 (15 mg/kg) beginning at either 1, 3, or 5 days after virus challenge and again about 2 days later. The presence of viral RNA, infectious virus, and/or NiV-specific immune responses demonstrated that all subjects were infected after challenge. All 12 AGMs that received m102.4 survived infection, whereas the untreated control subjects succumbed to disease between days 8 and 10 after infection. AGMs in the day 5 treatment group exhibited clinical signs of disease, but all animals recovered by day 16. These results represent the successful therapeutic in vivo efficacy by an investigational drug against NiV in a nonhuman primate and highlight the potential impact that a monoclonal antibody can have on a highly pathogenic zoonotic human disease.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Chlorocebus aethiops/imunologia , Infecções por Henipavirus/prevenção & controle , Infecções por Henipavirus/terapia , Vírus Nipah/imunologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/virologia , Humanos , Imuno-Histoquímica , Testes de Neutralização , Resultado do Tratamento , Carga Viral
8.
Virus Res ; 177(2): 119-26, 2013 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23954578

RESUMO

Since their first emergence in mid 1990s henipaviruses continued to re emerge in Australia and South East Asia almost every year. In total there has been more than 12 Nipah and 48 Hendra virus outbreaks reported in South East Asia and Australia, respectively. These outbreaks are associated with significant economic and health damages that most high risks countries (particularly in South East Asia) cannot bear the burden of such economical threats. Up until recently, there were no actual therapeutics available to treat or prevent these lethal infections. However, an international collaborative research has resulted in the identification of a potential equine Hendra vaccine capable of providing antibody protection against Hendra virus infections. Consequently, with the current findings and after nearly 2 decades since their first detection, are we there yet? This review recaps the chronicle of the henipavirus emergence and briefly evaluates potential anti-henipavirus vaccines and antivirals.


Assuntos
Doenças Transmissíveis Emergentes/veterinária , Doenças Transmissíveis Emergentes/virologia , Vírus Hendra/fisiologia , Infecções por Henipavirus/veterinária , Infecções por Henipavirus/virologia , Doenças dos Cavalos/virologia , Vírus Nipah/fisiologia , Animais , Doenças Transmissíveis Emergentes/terapia , Vírus Hendra/genética , Infecções por Henipavirus/epidemiologia , Infecções por Henipavirus/terapia , Doenças dos Cavalos/epidemiologia , Doenças dos Cavalos/terapia , Cavalos , Humanos , Vírus Nipah/genética
9.
Viruses ; 4(2): 280-308, 2012 02.
Artigo em Inglês | MEDLINE | ID: mdl-22470837

RESUMO

The Paramyxoviridae genus Henipavirus is presently represented by the type species Hendra and Nipah viruses which are both recently emerged zoonotic viral pathogens responsible for repeated outbreaks associated with high morbidity and mortality in Australia, Southeast Asia, India and Bangladesh. These enveloped viruses bind and enter host target cells through the coordinated activities of their attachment (G) and class I fusion (F) envelope glycoproteins. The henipavirus G glycoprotein interacts with host cellular B class ephrins, triggering conformational alterations in G that lead to the activation of the F glycoprotein, which facilitates the membrane fusion process. Using the recently published structures of HeV-G and NiV-G and other paramyxovirus glycoproteins, we review the features of the henipavirus envelope glycoproteins that appear essential for mediating the viral fusion process, including receptor binding, G-F interaction, F activation, with an emphasis on G and the mutations that disrupt viral infectivity. Finally, recent candidate therapeutics for henipavirus-mediated disease are summarized in light of their ability to inhibit HeV and NiV entry by targeting their G and F glycoproteins.


Assuntos
Antivirais/farmacologia , Henipavirus/efeitos dos fármacos , Henipavirus/patogenicidade , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/metabolismo , Internalização do Vírus/efeitos dos fármacos , Tratamento Farmacológico/métodos , Infecções por Henipavirus/terapia , Infecções por Henipavirus/virologia , Interações Hospedeiro-Patógeno , Humanos , Imunoterapia/métodos , Modelos Moleculares , Conformação Proteica , Proteínas do Envelope Viral/antagonistas & inibidores , Proteínas do Envelope Viral/química , Proteínas Virais de Fusão/antagonistas & inibidores , Proteínas Virais de Fusão/química
10.
Infect Disord Drug Targets ; 11(3): 315-36, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21488828

RESUMO

The Paramyxoviridae family comprises of several genera that contain emerging or re-emerging threats for human and animal health with no real specific effective treatment available. Hendra and Nipah virus are members of a newly identified genus of emerging paramyxoviruses, Henipavirus. Since their discovery in the 1990s, henipaviruses outbreaks have been associated with high economic and public health threat potential. When compared to other paramyxoviruses, henipaviruses appear to have unique characteristics. Henipaviruses are zoonotic paramyxoviruses with a broader tropism than most other paramyxoviruses, and can cause severe acute encephalitis with unique features among viral encephalitides. There are currently no approved effective prophylactic or therapeutic treatments for henipavirus infections. Although ribavirin was empirically used and seemed beneficial during the biggest outbreak caused by one of these viruses, the Nipah virus, its efficacy is disputed in light of its lack of efficacy in several animal models of henipavirus infection. Nevertheless, because of its highly pathogenic nature, much effort has been spent in developing anti-henipavirus therapeutics. In this review we describe the unique features of henipavirus infections and the different strategies and animal models that have been developed so far in order to identify and test potential drugs to prevent or treat henipavirus infections. Some of these components have the potential to be broad-spectrum antivirals as they target effectors of viral pathogenecity common to other viruses. We will focus on small molecules or biologics, rather than vaccine strategies, that have been developed as anti-henipaviral therapeutics.


Assuntos
Encefalite Viral/patologia , Encefalite Viral/terapia , Vírus Hendra , Infecções por Henipavirus/patologia , Infecções por Henipavirus/terapia , Vírus Nipah , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Surtos de Doenças , Encefalite Viral/epidemiologia , Infecções por Henipavirus/epidemiologia , Humanos
11.
PLoS Pathog ; 6(10): e1001168, 2010 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-21060819

RESUMO

In the paramyxovirus cell entry process, receptor binding triggers conformational changes in the fusion protein (F) leading to viral and cellular membrane fusion. Peptides derived from C-terminal heptad repeat (HRC) regions in F have been shown to inhibit fusion by preventing formation of the fusogenic six-helix bundle. We recently showed that the addition of a cholesterol group to HRC peptides active against Nipah virus targets these peptides to the membrane where fusion occurs, dramatically increasing their antiviral effect. In this work, we report that unlike the untagged HRC peptides, which bind to the postulated extended intermediate state bridging the viral and cell membranes, the cholesterol tagged HRC-derived peptides interact with F before the fusion peptide inserts into the target cell membrane, thus capturing an earlier stage in the F-activation process. Furthermore, we show that cholesterol tagging renders these peptides active in vivo: the cholesterol-tagged peptides cross the blood brain barrier, and effectively prevent and treat in an established animal model what would otherwise be fatal Nipah virus encephalitis. The in vivo efficacy of cholesterol-tagged peptides, and in particular their ability to penetrate the CNS, suggests that they are promising candidates for the prevention or therapy of infection by Nipah and other lethal paramyxoviruses.


Assuntos
Colesterol/uso terapêutico , Infecções por Henipavirus/prevenção & controle , Vírus Nipah/fisiologia , Paramyxovirinae/fisiologia , Proteínas Virais de Fusão/antagonistas & inibidores , Internalização do Vírus , Motivos de Aminoácidos/efeitos dos fármacos , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Células Cultivadas , Chlorocebus aethiops , Colesterol/química , Colesterol/farmacologia , Regulação para Baixo , Infecções por Henipavirus/imunologia , Infecções por Henipavirus/terapia , Humanos , Modelos Biológicos , Modelos Moleculares , Dados de Sequência Molecular , Terapia de Alvo Molecular , Vírus Nipah/efeitos dos fármacos , Vírus Nipah/imunologia , Vírus Nipah/patogenicidade , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/uso terapêutico , Células Vero , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo , Proteínas Virais de Fusão/fisiologia
12.
Antiviral Res ; 80(3): 324-31, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18687361

RESUMO

Nipah virus (NiV) and Hendra virus (HeV) are recently emerged zoonotic paramyxoviruses exclusively grouped within a new genus, Henipavirus. These viruses cause fatal disease in a wide range of species, including humans. Both NiV and HeV have continued to re-emerge sporadically in Bangladesh and Australia, respectively. There are currently no therapeutics or vaccines available to treat Henipavirus infection and both are classified as BSL4 pathogens. RNA interference (RNAi) is a process by which double-stranded RNA directs sequence-specific degradation of messenger RNA in animal and plant cells. Small interfering RNAs (siRNAs) mediate RNAi by inhibiting gene expression of homologous mRNA and our preliminary studies suggest RNAi may be a useful approach to developing novel therapies for these highly lethal pathogens. Eight NiV siRNA molecules (four L and four N gene specific), two HeV N gene specific, and two non-specific control siRNA molecules were designed and tested for their ability to inhibit a henipavirus minigenome replication system (which does not require the use of live virus) in addition to live virus infections in vitro. In the minigenome assay three out of the four siRNAs that targeted the L gene of NiV effectively inhibited replication. In contrast, only NiV N gene siRNAs were effective in reducing live NiV replication, suggesting inhibition of early, abundantly expressed gene transcripts may be more effective than later, less abundant transcripts. Additionally, some of the siRNAs effective against NiV infection were only partially effective inhibitors of HeV infection. An inverse correlation between the number of nucleotide mismatches and the efficacy of siRNA inhibition was observed. The demonstration that RNAi effectively inhibits henipavirus replication in vitro, is a novel approach and may provide an effective therapy for these highly lethal, zoonotic pathogens.


Assuntos
Infecções por Henipavirus/terapia , Infecções por Henipavirus/virologia , Henipavirus/genética , Doenças dos Cavalos/virologia , Interferência de RNA , Animais , Sequência de Bases , Linhagem Celular , Chlorocebus aethiops , Cricetinae , Genoma Viral , Henipavirus/fisiologia , Infecções por Henipavirus/genética , Infecções por Henipavirus/veterinária , Cavalos , Humanos , RNA Interferente Pequeno/genética , Alinhamento de Sequência , Células Vero , Replicação Viral
13.
Curr Mol Med ; 5(8): 805-16, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16375714

RESUMO

Within the past decade a number of new zoonotic paramyxoviruses emerged from flying foxes to cause serious disease outbreaks in man and livestock. Hendra virus was the cause of fatal infections of horses and man in Australia in 1994, 1999 and 2004. Nipah virus caused encephalitis in humans both in Malaysia in 1998/99, following silent spread of the virus in the pig population, and in Bangladesh from 2001 to 2004 probably as a result of direct bat to human transmission and spread within the human population. Hendra and Nipah viruses are highly pathogenic in humans with case fatality rates of 40% to 70%. Their genetic constitution, virulence and wide host range make them unique paramyxoviruses and they have been given Biosecurity Level 4 status in a new genus Henipavirus within the family Paramyxoviridae. Recent studies on the virulence, host range and cell tropisms of henipaviruses provide insights into the unique biological properties of these emerging human pathogens and suggest approaches for vaccine development and therapeutic countermeasures.


Assuntos
Vírus Hendra/patogenicidade , Infecções por Henipavirus/terapia , Infecções por Henipavirus/virologia , Vírus Nipah/patogenicidade , Animais , Antivirais/uso terapêutico , Modelos Animais de Doenças , Desenho de Fármacos , Vírus Hendra/classificação , Infecções por Henipavirus/epidemiologia , Humanos , Vírus Nipah/classificação , Vacinas Virais , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...