Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.314
Filtrar
1.
J Ethnopharmacol ; 336: 118720, 2025 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-39197802

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Jinye Baidu granules (JYBD) have been used to treat acute respiratory tract infections and demonstrated clinical efficacy for the treatment of emerging or epidemic respiratory viruses such as SARS-CoV-2 and influenza virus. AIM OF THE STUDY: This study is to investigate the antiviral effect of JYBD against influenza A viruses (IAV) in vitro and in vivo and elucidate its underlying mechanism. MATERIALS AND METHODS: Ultra-high-performance liquid chromatography connected with Orbitrap mass spectrometer (UHPLC-Orbitrap MS) was employed to describe the chemical profile of JYBD. The potential pathways and targets involved in JYBD against IAV infection were predicted by network pharmacology. The efficacy and mechanism of JYBD were validated through both in vivo and in vitro experiments. Moreover, combination therapy with JYBD and the classic anti-influenza drugs was also investigated. RESULTS: A total of 126 compounds were identified by UHPLC-Orbitrap MS, of which 9 compounds were unambiguously confirmed with reference standards. JYBD could significantly inhibit the replication of multiple strains of IAV, especially oseltamivir-resistant strains. The results of qRT-PCR and WB demonstrated that JYBD could inhibit the excessive induction of pro-inflammatory cytokines induced by IAV infection and regulate inflammatory response through inhibiting JAK/STAT, NF-κB and MAPK pathways. Moreover, both JYBD monotherapy or in combination with oseltamivir could alleviate IAV-induced severe lung injury in mice. CONCLUSIONS: JYBD could inhibit IAV replication and mitigate virus-induced excessive inflammatory response. Combinations of JYBD and neuraminidase inhibitors conferred synergistic suppression of IAV both in vitro and in vivo. It might provide a scientific basis for clinical applications of JYBD against influenza virus infected diseases.


Assuntos
Antivirais , Medicamentos de Ervas Chinesas , Vírus da Influenza A , Farmacologia em Rede , Infecções por Orthomyxoviridae , Antivirais/farmacologia , Animais , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/química , Vírus da Influenza A/efeitos dos fármacos , Cães , Camundongos , Humanos , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Células Madin Darby de Rim Canino , Replicação Viral/efeitos dos fármacos , Células A549 , Camundongos Endogâmicos BALB C , Masculino , Feminino , Cromatografia Líquida de Alta Pressão
2.
Biomaterials ; 312: 122721, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39106817

RESUMO

Silver nanoparticles (AgNPs) are a potential antiviral agent due to their ability to disrupt the viral particle or alter the virus metabolism inside the host cell. In vitro, AgNPs exhibit antiviral activity against the most common human respiratory viruses. However, their capacity to modulate immune responses during respiratory viral infections has yet to be explored. This study demonstrates that administering AgNPs directly into the lungs prior to infection can reduce viral loads and therefore virus-induced cytokines in mice infected with influenza virus or murine pneumonia virus. The prophylactic effect was diminished in mice with depleted lymphoid cells. We showed that AgNPs-treatment resulted in the recruitment and activation of lymphocytes in the lungs, particularly natural killer (NK) cells. Mechanistically, AgNPs enhanced the ability of alveolar macrophages to promote both NK cell migration and IFN-γ production. By contrast, following infection, in mice treated with AgNPs, NK cells exhibited decreased activation, indicating that these nanoparticles can regulate the potentially deleterious activation of these cells. Overall, the data suggest that AgNPs may possess prophylactic antiviral properties by recruiting and controlling the activation of lymphoid cells through interaction with alveolar macrophages.


Assuntos
Células Matadoras Naturais , Pulmão , Nanopartículas Metálicas , Infecções por Orthomyxoviridae , Prata , Animais , Prata/química , Prata/farmacologia , Nanopartículas Metálicas/química , Pulmão/virologia , Pulmão/patologia , Pulmão/efeitos dos fármacos , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Camundongos , Células Matadoras Naturais/efeitos dos fármacos , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virologia , Camundongos Endogâmicos C57BL , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Antivirais/farmacologia , Antivirais/uso terapêutico , Feminino , Ativação Linfocitária/efeitos dos fármacos
3.
Biomaterials ; 312: 122736, 2025 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39121728

RESUMO

The resurgence of influenza viruses as a significant global threat emphasizes the urgent need for innovative antiviral strategies beyond existing treatments. Here, we present the development and evaluation of a novel super-multivalent sialyllactosylated filamentous phage, termed t-6SLPhage, as a potent entry blocker for influenza A viruses. Structural variations in sialyllactosyl ligands, including linkage type, valency, net charge, and spacer length, were systematically explored to identify optimal binding characteristics against target hemagglutinins and influenza viruses. The selected SLPhage equipped with optimal ligands, exhibited exceptional inhibitory potency in in vitro infection inhibition assays. Furthermore, in vivo studies demonstrated its efficacy as both a preventive and therapeutic intervention, even when administered post-exposure at 2 days post-infection, under 4 lethal dose 50% conditions. Remarkably, co-administration with oseltamivir revealed a synergistic effect, suggesting potential combination therapies to enhance efficacy and mitigate resistance. Our findings highlight the efficacy and safety of sialylated filamentous bacteriophages as promising influenza inhibitors. Moreover, the versatility of M13 phages for surface modifications offers avenues for further engineering to enhance therapeutic and preventive performance.


Assuntos
Antivirais , Animais , Antivirais/farmacologia , Antivirais/química , Humanos , Cães , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Células Madin Darby de Rim Canino , Inovirus/efeitos dos fármacos , Oseltamivir/farmacologia , Oseltamivir/química , Camundongos , Influenza Humana/virologia , Influenza Humana/tratamento farmacológico , Camundongos Endogâmicos BALB C , Ácido N-Acetilneuramínico/química , Ácido N-Acetilneuramínico/metabolismo , Feminino
4.
Virol J ; 21(1): 240, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354538

RESUMO

BACKGROUND: Infection of mice with mouse-adapted strains of influenza virus has been widely used to establish mouse pneumonia models. Intranasal inoculation is the traditional route for constructing an influenza virus-induced pneumonia mouse model, while intratracheal inoculation has been gradually applied in recent years. In this article, the pathogenicity of influenza virus-induced pneumonia mouse models following intranasal and aerosolized intratracheal inoculation were compared. METHODS: By comparing the two ways of influenza inoculation, intranasal and intratracheal, a variety of indices such as survival rate, body weight change, viral titer and load, pathological change, lung wet/dry ratio, and inflammatory factors were investigated. Meanwhile, the transcriptome was applied for the initial exploration of the mechanism underlying the variations in the results between the two inoculation methods. RESULTS: The findings suggest that aerosolized intratracheal infection leads to more severe lung injury and higher viral loads in the lungs compared to intranasal infection, which may be influenced by the initial site of infection, sialic acid receptor distribution, and host innate immunity. CONCLUSION: Intratracheal inoculation is a better method for modelling severe pneumonia in mice than intranasal infection.


Assuntos
Administração Intranasal , Modelos Animais de Doenças , Pulmão , Infecções por Orthomyxoviridae , Carga Viral , Animais , Camundongos , Pulmão/virologia , Pulmão/patologia , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/patologia , Feminino , Aerossóis , Camundongos Endogâmicos BALB C , Pneumonia Viral/virologia , Pneumonia Viral/patologia , Pneumonia Viral/imunologia , Orthomyxoviridae/patogenicidade , Perfilação da Expressão Gênica
5.
J Clin Invest ; 134(19)2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39352386

RESUMO

Ineffective recovery from pneumonia can lead to interstitial lung disease characterized by aberrant epithelial cells in fibrotic regions. In this issue of the JCI, Lin et al. define molecular pathways leading to the development and persistence of keratin 5+ (Krt5+) epithelial cells in the alveolar parenchyma when mice struggle to recover from influenza infection. The receptor for IFN-γ on lung epithelium was essential for the formation of aberrant Krt5+ cells and fibrotic lung disease. The transcription factor Yes-associated protein 1 (YAP) was necessary for persistence of these Krt5+ cells, and IFN-γ activated YAP in lung epithelial cells via JAK, focal adhesion kinase (FAK), and Src kinases. These findings establish a targetable pathway underlying some of the pulmonary postacute sequelae of pneumonia.


Assuntos
Interferon gama , Proteínas de Sinalização YAP , Animais , Camundongos , Proteínas de Sinalização YAP/metabolismo , Interferon gama/metabolismo , Humanos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia
6.
Commun Biol ; 7(1): 1230, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39354058

RESUMO

Frequent interspecies transmission of human influenza A viruses (FLUAV) to pigs contrasts with the limited subset that establishes in swine. While hemagglutinin mutations are recognized for their role in cross-species transmission, the contribution of neuraminidase remains understudied. Here, the NA's role in FLUAV adaptation was investigated using a swine-adapted H3N2 reassortant virus with human-derived HA and NA segments. Adaptation in pigs resulted in mutations in both HA (A138S) and NA (D113A). The D113A mutation abolished calcium (Ca2+) binding in the low-affinity Ca2+-binding pocket of NA, enhancing enzymatic activity and thermostability under Ca2+-depleted conditions, mirroring swine-origin FLUAV NA behavior. Structural analysis predicts that swine-adapted H3N2 viruses lack Ca2+ binding in this pocket. Further, residue 93 in NA (G93 in human, N93 in swine) also influences Ca2+ binding and impacts NA activity and thermostability, even when D113 is present. These findings demonstrate that mutations in influenza A virus surface proteins alter evolutionary trajectories following interspecies transmission and reveal distinct mechanisms modulating NA activity during FLUAV adaptation, highlighting the importance of Ca2+ binding in the low-affinity calcium-binding pocket.


Assuntos
Cálcio , Neuraminidase , Neuraminidase/metabolismo , Neuraminidase/genética , Neuraminidase/química , Humanos , Animais , Cálcio/metabolismo , Suínos , Sítios de Ligação , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/transmissão , Influenza Humana/virologia , Influenza Humana/transmissão , Adaptação Fisiológica/genética , Proteínas Virais/metabolismo , Proteínas Virais/genética , Proteínas Virais/química , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/metabolismo , Mutação , Ligação Proteica , Doenças dos Suínos/virologia
8.
J Med Virol ; 96(9): e29906, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39262090

RESUMO

Influenza virus-induced viral pneumonia is a major threat to human health, and specific therapeutic agents for viral pneumonia are still lacking. MoringaA (MA) is an anti-influenza virus active compound isolated from Moringa seeds, which can inhibit influenza virus by activating the TFEB-autophagic lysosomal pathway in host cells. In this study, we obtained exosomes from M2-type macrophages and encapsulated and delivered MA (MA-Exos), and we investigated the efficacy of MA-Exos in antiviral and viral pneumonia in vivo and in vitro, respectively. In addition, we provided insights into the mechanism by which MA-Exos regulates TFEB-lysosomal autophagy by RNA sequencing. The MA-Exos showed broad-spectrum inhibition of IAV, and significant promotion of the autophagic lysosomal pathway. Meanwhile, we found that GCN5 gene and protein were significantly down-regulated in IAV-infected cells after MA-Exos intervention, indicating its blocking the acetylation of TFEB by GCN5. In addition, MA-Exos also significantly promoted autophagy in lung tissue cells of mice with viral pneumonia. MA-Exos can inhibit and clear influenza virus by mediating the TFEB-autophagy lysosomal pathway by a mechanism related to the down-regulation of histone acetyltransferase GCN5. Our study provides a strategy for targeting MA-Exos for the treatment of viral pneumonia from both antiviral and virus-induced inflammation inhibition pathways.


Assuntos
Antivirais , Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Exossomos , Vírus da Influenza A , Lisossomos , Animais , Camundongos , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Lisossomos/metabolismo , Lisossomos/efeitos dos fármacos , Lisossomos/virologia , Exossomos/metabolismo , Antivirais/farmacologia , Autofagia/efeitos dos fármacos , Humanos , Vírus da Influenza A/efeitos dos fármacos , Vírus da Influenza A/fisiologia , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Macrófagos/virologia , Macrófagos/efeitos dos fármacos , Pulmão/virologia
9.
PLoS One ; 19(9): e0308648, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39312544

RESUMO

BACKGROUND: The alveolar epithelium is protected by a heparan sulfate-rich, glycosaminoglycan layer called the epithelial glycocalyx. It is cleaved in patients with acute respiratory distress syndrome (ARDS) and in murine models of influenza A (IAV) infection, shedding fragments into the airspace from the cell surface. Glycocalyx shedding results in increased permeability of the alveolar-capillary barrier, amplifying acute lung injury. The mechanisms underlying alveolar epithelial glycocalyx shedding in IAV infection are unknown. We hypothesized that induction of host sheddases such as matrix metalloproteinases (MMPs) during IAV infection results in glycocalyx shedding and increased lung injury. MATERIALS AND METHODS: We measured glycocalyx shedding and lung injury during IAV infection with and without treatment with the pan-MMP inhibitor Ilomastat (ILO) and in an MMP-7 knock out (MMP-7KO) mouse. C57BL/6 or MMP-7KO male and female mice were given IAV A/PR/8/34 (H1N1) at 30,000 PFU/mouse or PBS intratracheally. For some experiments, C56BL/6 mice were infected in the presence of ILO (100mg/kg) or vehicle given daily by IP injection. Bronchoalveolar lavage (BAL) and lung tissue were collected on day 1, 3, and 7 for analysis of glycocalyx shedding (BAL Syndecan-1) and lung injury (histology, BAL protein, BAL cytokines, BAL immune cell infiltrates, BAL RAGE). Expression and localization of the sheddase MMP-7 and its inhibitor TIMP-1 was examined by RNAScope. For in vitro experiments, MLE-12 mouse lung epithelial cells were cultured and treated with active or heat-inactivated heparinase (2.5 U/mL) prior to infection with IAV (MOI 1) and viral load and MMP-7 and TIMP-1 expression analyzed. RESULTS: IAV infection caused shedding of the epithelial glycocalyx into the BAL. Inhibition of MMPs with ILO reduced glycocalyx shedding by 36% (p = 0.0051) and reduced lung epithelial injury by 40% (p = 0.0404). ILO also reduced viral load by 68% (p = 0.027), despite having no significant effect on lung cytokine production. Both MMP-7 and its inhibitor TIMP-1 were upregulated in IAV infected mice: MMP-7 colocalized with IAV, while TIMP-1 was limited to cells adjacent to infection. However, MMP-7KO mice had similar glycocalyx shedding, epithelial injury, and viral load compared to WT littermates, suggesting redundancy in MMP sheddase function in the lung. In vitro, heparinase treatment before infection led to a 52% increase in viral load (p = 0.0038) without altering MMP-7 or TIMP-1 protein levels. CONCLUSIONS: Glycocalyx shedding and MMPs play key roles in IAV-induced epithelial injury, with significant impact on IAV viral load. Further studies are needed to understand which specific MMPs regulate lung epithelial glycocalyx shedding.


Assuntos
Glicocálix , Metaloproteinase 7 da Matriz , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae , Animais , Glicocálix/metabolismo , Camundongos , Feminino , Masculino , Metaloproteinase 7 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/genética , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/patologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Camundongos Knockout , Alvéolos Pulmonares/virologia , Alvéolos Pulmonares/metabolismo , Alvéolos Pulmonares/patologia , Ácidos Hidroxâmicos/farmacologia , Inibidores de Metaloproteinases de Matriz/farmacologia , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Metaloproteinases da Matriz/metabolismo , Indóis
10.
Sci Rep ; 14(1): 21324, 2024 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-39266641

RESUMO

The lymphatic system consists of a vessel network lined by specialized lymphatic endothelial cells (LECs) that are responsible for tissue fluid homeostasis and immune cell trafficking. The mechanisms for organ-specific LEC responses to environmental cues are not well understood. We found robust lymphangiogenesis during influenza A virus infection in the adult mouse lung. We show that the number of LECs increases twofold at 7 days post-influenza infection (dpi) and threefold at 21 dpi, and that lymphangiogenesis is preceded by lymphatic dilation. We also show that the expanded lymphatic network enhances fluid drainage to mediastinal lymph nodes. Using EdU labeling, we found that a significantly higher number of pulmonary LECs are proliferating at 7 dpi compared to LECs in homeostatic conditions. Lineage tracing during influenza indicates that new pulmonary LECs are derived from preexisting LECs rather than non-LEC progenitors. Lastly, using a conditional LEC-specific YAP/TAZ knockout model, we established that lymphangiogenesis, fluid transport and the immune response to influenza are independent of YAP/TAZ activity in LECs. These findings were unexpected, as they indicate that YAP/TAZ signaling is not crucial for these processes.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Células Endoteliais , Pulmão , Linfangiogênese , Infecções por Orthomyxoviridae , Proteínas de Sinalização YAP , Animais , Proteínas de Sinalização YAP/metabolismo , Células Endoteliais/metabolismo , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Pulmão/metabolismo , Pulmão/patologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/patologia , Vírus da Influenza A/fisiologia , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patologia , Camundongos Knockout , Transdução de Sinais , Proliferação de Células , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Camundongos Endogâmicos C57BL , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
11.
Open Vet J ; 14(8): 1896-1904, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39308731

RESUMO

Background: Dogs in close contact with humans can serve as a source of potentially dangerous reassortant influenza viruses (IVs) with zoonotic potential. The dog's body can serve as a vessel for the emergence of new IVs. These new viruses can become a source of infection for other animals and humans. The potential for zoonotic transmission of IVs from dogs to humans poses a public health risk. Aim: Study of the circulation of IVs in the dog population in Almaty, Kazakhstan. Methods: Biosamples (oropharyngeal swabs and blood serum) from dogs were collected from veterinary clinics in Almaty in 2023-2024. Samples were screened using RT-PCR, HI assay, and ELISA. Results: RT-PCR analysis of 355 nasopharyngeal swabs showed the presence of influenza A virus (IAV) in 32 samples (9.01% of the total number of samples analyzed). When subtyping IAV H1N1 RNA was detected in 19 swabs (5.35%). IAV subtype could not be determined in 13 PCR-positive samples (3.66%). The genetic material of IAV H3N2, H5, H7, and H9, as well as coronavirus, bocavirus, and adenovirus has not been identified. In a serological analysis of 180 blood sera using ELISA, antibodies to IAV were detected in 5.56% (n = 10). The results of the HI assay showed the presence of antihemagglutinins to A/H1N1pdm in 6.11% (11 samples), to A/H3N2 in 9.44% (17 samples), and no antibodies to IAV H5, H7, and type B were detected. Conclusion: There is no information about human infection with any canine influenza virus. However, many cases of infection in dogs with human IAVs H1N1, H1N1pdm09, and H3N2 have been described. When dogs are co-infected with different IAVs, new recombinant IAVs may emerge that can infect humans and other animals. Therefore, ongoing global surveillance of animal populations is necessary to monitor the evolution and circulation of viruses dangerous to public health. This is also important for timely preparation for the emergence of a new zoonotic influenza virus that has pandemic potential for humans.


Assuntos
Doenças do Cão , Infecções por Orthomyxoviridae , Animais , Cães , Doenças do Cão/epidemiologia , Doenças do Cão/virologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Cazaquistão/epidemiologia , Vírus da Influenza A/isolamento & purificação , Humanos , Ensaio de Imunoadsorção Enzimática/veterinária
12.
Emerg Microbes Infect ; 13(1): 2396864, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39331815

RESUMO

Single B cells-based antibody platforms offer an effective approach for the discovery of useful antibodies for therapeutic or research purposes. Here we present a method for screening equine immunoglobins F(ab)2, which offers the potential advantage of reacting with multiple epitopes on the virus. Using equine influenza virus (EIV) as model, a hemagglutinin (HA) trimer was constructed to bait B cells in vaccinated horses. We screened 370 HA-specific B cells from 1 × 106 PBMCs and identified a diverse set of equine variable region gene sequences of heavy and light chains and then recombined with humanized Ig Fc. Recombinant equine Ig was then self-assembled in co-transfected 293 T cells, and subsequently optimized to obtain HA binding B-cell receptor (s). The recombinant antibodies exhibited a high binding affinity to the HA protein. Antibody H81 exhibited the highest cross neutralizing activities against EIV strains in vitro. Furthermore, it effectively protected EIV-challenged mice, resulting in significantly improved survival, reduced pulmonary inflammation and decreased viral titers. In silico predication identified a functional region of H81 comprising 27 key amino acids cross the main circulating EIV strains. The 12 amino acid residues in this region with the highest binding affinities were screened. Notably, the predicted epitopes of H81 encompassed the documented equine HA receptor binding site, validating its cross-neutralization. In summary, a rapid platform was successfully established to investigate the profiling of equine antigen-recognizing receptors (BCRs) following infection. This platform has the potential to optimize the screening of virus-neutralizing antibodies and aid in vaccine design.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Linfócitos B , Infecções por Orthomyxoviridae , Animais , Cavalos , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Camundongos , Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Anticorpos Neutralizantes/imunologia , Humanos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H3N8/imunologia , Vírus da Influenza A Subtipo H3N8/genética , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/genética , Reações Cruzadas , Células HEK293 , Vacinas contra Influenza/imunologia , Memória Imunológica , Feminino , Epitopos/imunologia
13.
Emerg Microbes Infect ; 13(1): 2400546, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39221898

RESUMO

The vast majority of data obtained from sequence analysis of influenza A viruses (IAVs) have revealed that nonstructural 1 (NS1) proteins from H1N1 swine, H3N8 equine, H3N2 avian and the correspondent subtypes from dogs have a conserved four C-terminal amino acid motif when independent cross-species transmission occurs between these species. To test the influence of the C-terminal amino acid motifs of NS1 protein on the replication and virulence of IAVs, we systematically generated 7 recombinants, which carried naturally truncated NS1 proteins, and their last four C-terminal residues were replaced with PEQK and SEQK (for H1N1), EPEV and KPEI (for H3N8) and ESEV and ESEI (for H3N2) IAVs. Another recombinant was generated by removing the C-terminal residues by reverse genetics. Remarkably, the ESEI and KPEI motifs circulating in canines largely contributed efficient replication in cultured cells and these had enhanced virulence. In contrast, the avian ESEV motif was only responsible for high pathogenicity in mice. We examined the effects of these motifs upon interferon (IFN) induction. The 7 mutant viruses replicated in vitro in an IFN-independent manner, and the canine SEQK motif was able to induced higher levels of IFN-ß in human cell lines. These findings shed further new light on the role of the four C-terminal residues in replication and virulence of IAVs and suggest that these motifs can modulate viral replication in a species-specific manner.


Assuntos
Motivos de Aminoácidos , Vírus da Influenza A Subtipo H1N1 , Infecções por Orthomyxoviridae , Proteínas não Estruturais Virais , Replicação Viral , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Proteínas não Estruturais Virais/química , Animais , Cães , Virulência , Camundongos , Vírus da Influenza A Subtipo H1N1/genética , Vírus da Influenza A Subtipo H1N1/patogenicidade , Vírus da Influenza A Subtipo H1N1/fisiologia , Infecções por Orthomyxoviridae/virologia , Humanos , Células Madin Darby de Rim Canino , Camundongos Endogâmicos BALB C , Doenças do Cão/virologia , Vírus da Influenza A Subtipo H3N2/genética , Vírus da Influenza A Subtipo H3N2/patogenicidade , Vírus da Influenza A Subtipo H3N2/fisiologia , Vírus da Influenza A Subtipo H3N8/genética , Vírus da Influenza A Subtipo H3N8/patogenicidade , Feminino
14.
Proc Natl Acad Sci U S A ; 121(41): e2408469121, 2024 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-39348541

RESUMO

Despite much research, considerable data suggest that influenza virus remains a serious health problem because i) the effectiveness of current vaccines ranges only from 19% to 60%, ii) available therapies remain ineffective in advanced stages of disease, iii) death rates vary between 25,000 and 72,000/year in the United States, and iv) avian influenza strains are now being transmitted to dairy cattle that in turn are infecting humans. To address these concerns, we have developed zanDR, a bispecific small molecule that binds and inhibits viral neuraminidase expressed on both free virus and virus-infected cells and recruits naturally occurring anti-rhamnose and anti-dinitrophenyl (DNP) antibodies with rhamnose and DNP haptens. Because the neuraminidase inhibition replicates the chemotherapeutic mechanism of zanamivir and oseltamivir, while rhamnose and DNP recruit endogenous antibodies much like an anti-influenza vaccine, zanDR reproduces most of the functions of current methods of protection against influenza virus infections. Importantly, studies on cells in culture demonstrate that both of the above protective mechanisms remain highly functional in the zanDR conjugate, while studies in lethally infected mice with advanced-stage disease establish that a single intranasal dose of zanDR not only yields 100% protection but also reduces lung viral loads faster and ~1,000× more thoroughly than current antiviral therapies. Since zanDR also lowers secretion of proinflammatory cytokines and protects against virus-induced damage to the lungs better than current therapies, we suggest that combining an immunotherapy with a chemotherapy in single pharmacological agent constitutes a promising approach for treating the more challenging forms of influenza.


Assuntos
Infecções por Orthomyxoviridae , Animais , Camundongos , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/tratamento farmacológico , Infecções por Orthomyxoviridae/virologia , Humanos , Antivirais/farmacologia , Influenza Humana/imunologia , Influenza Humana/tratamento farmacológico , Influenza Humana/prevenção & controle , Neuraminidase/imunologia , Neuraminidase/antagonistas & inibidores , Cães , Feminino , Camundongos Endogâmicos BALB C , Anticorpos Antivirais/imunologia
16.
Emerg Microbes Infect ; 13(1): 2400530, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39221652

RESUMO

Multiple genetic variants of H1 and H3 influenza A viruses (IAVs) circulate concurrently in US swine farms. Understanding the spatial transmission patterns of IAVs among these farms is crucial for developing effective control strategies and mitigating the emergence of novel IAVs. In this study, we analysed 1909 IAV genomic sequences from 785 US swine farms, representing 33 farming systems across 12 states, primarily in the Midwest from 2004 to 2023. Bayesian phylogeographic analyses were performed to identify the dispersal patterns of both H1 and H3 virus genetic lineages and to elucidate their spatial migration patterns within and between different systems. Our results showed that both intra-system and inter-system migrations occurred between the swine farms, with intra-system migrations being more frequent. However, migration rates for H1 and H3 IAVs were similar between intra-system and inter-system migration events. Spatial migration patterns aligned with expected pig movement across different compartments of swine farming systems. Sow-Farms were identified as key sources of viruses, with bi-directional migration observed between these farms and other parts of the system, including Wean-to-Finish and Gilt-Development-Units. High intra-system migration was detected across farms in the same region, while spread to geographically distant intra- and inter-system farms was less frequent. These findings suggest that prioritizing resources towards systems frequently confronting influenza problems and targeting pivotal source farms, such as sow farms, could be an effective strategy for controlling influenza in US commercial swine operations.


Assuntos
Fazendas , Vírus da Influenza A , Infecções por Orthomyxoviridae , Doenças dos Suínos , Animais , Suínos , Estados Unidos/epidemiologia , Doenças dos Suínos/virologia , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/transmissão , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/transmissão , Infecções por Orthomyxoviridae/epidemiologia , Vírus da Influenza A/genética , Vírus da Influenza A/classificação , Vírus da Influenza A/isolamento & purificação , Filogeografia , Filogenia , Teorema de Bayes
18.
Emerg Infect Dis ; 30(10): 2033-2041, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39240548

RESUMO

The prevalence of highly pathogenic avian influenza (HPAI) A(H5N1) viruses has increased in wild birds and poultry worldwide, and concomitant outbreaks in mammals have occurred. During 2023, outbreaks of HPAI H5N1 virus infections were reported in cats in South Korea. The H5N1 clade 2.3.4.4b viruses isolated from 2 cats harbored mutations in the polymerase basic protein 2 gene encoding single amino acid substitutions E627K or D701N, which are associated with virus adaptation in mammals. Hence, we analyzed the pathogenicity and transmission of the cat-derived H5N1 viruses in other mammals. Both isolates caused fatal infections in mice and ferrets. We observed contact infections between ferrets, confirming the viruses had high pathogenicity and transmission in mammals. Most HPAI H5N1 virus infections in humans have occurred through direct contact with poultry or a contaminated environment. Therefore, One Health surveillance of mammals, wild birds, and poultry is needed to prevent potential zoonotic threats.


Assuntos
Furões , Virus da Influenza A Subtipo H5N1 , Infecções por Orthomyxoviridae , Animais , Furões/virologia , República da Coreia/epidemiologia , Camundongos , Gatos , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/epidemiologia , Virus da Influenza A Subtipo H5N1/patogenicidade , Virus da Influenza A Subtipo H5N1/genética , Filogenia , Doenças do Gato/virologia , Doenças do Gato/epidemiologia , Virulência , Surtos de Doenças , Humanos , Feminino
19.
Sci Adv ; 10(39): eado7087, 2024 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-39331716

RESUMO

Pregnancy is associated with profound changes in immunity. However, pregnancy-related respiratory immune adaptations in response to influenza infection and their impact on disease severity remain unclear. Here, we describe, in a preclinical model of mid-gestation pregnancy, a mechanism of enhanced host defense against influenza A virus (IAV) localized to the nasal cavity that limits viral replication and reduces the magnitude of intrapulmonary immune responses. Consequently, the pregnant mice show reduced pulmonary pathology and preserved airway function after IAV infection. The early restriction of viral replication is independent of type I interferon (IFN) but dependent on increased antimicrobial peptides (AMPs) driven by interleukin-17+ (IL-17+) γδ+ T cells within the nasal passages. This pathway of host defense against IAV infection in the upper airways during pregnancy restricts early viral infection and prevents virus dissemination into the lung supporting maternal fitness.


Assuntos
Vírus da Influenza A , Interferon Tipo I , Interleucina-17 , Mucosa Nasal , Infecções por Orthomyxoviridae , Animais , Feminino , Gravidez , Interleucina-17/metabolismo , Interleucina-17/imunologia , Camundongos , Interferon Tipo I/metabolismo , Interferon Tipo I/imunologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Mucosa Nasal/metabolismo , Vírus da Influenza A/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Replicação Viral , Pulmão/imunologia , Pulmão/virologia
20.
Viruses ; 16(9)2024 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-39339851

RESUMO

The emergence and spread of highly pathogenic avian influenza virus A subtype H5N1 (HP H5N1-IAV), particularly clade H5N1 2.3.4.4b, pose a severe global health threat, affecting various species, including mammals. Historically, cattle have been considered less susceptible to IAV, but recent outbreaks of H5N1-IAV 2.3.4.4b in dairy farms suggest a shift in host tropism, underscoring the urgency of expanded surveillance and the need for adaptable diagnostic tools in outbreak management. This study investigated the presence of anti-nucleoprotein (NP) antibodies in serum and milk and viral RNA in milk on dairy farms affected by outbreaks in Texas, Kansas, and Michigan using a multi-species IAV ELISA and RT-qPCR. The analysis of ELISA results from a Michigan dairy farm outbreak demonstrated a positive correlation between paired serum and milk sample results, confirming the reliability of both specimen types. Our findings also revealed high diagnostic performance during the convalescent phase (up to 96%), further improving sensitivity through serial sampling. Additionally, the evaluation of diagnostic specificity using serum and milk samples from IAV-free farms showed an excellent performance (99.6%). This study underscores the efficacy of the IAV NP-blocking ELISA for detecting and monitoring H5N1-IAV 2.3.4.4b circulation in dairy farms, whose recent emergence raises significant animal welfare and zoonotic concerns, necessitating expanded surveillance efforts.


Assuntos
Doenças dos Bovinos , Surtos de Doenças , Leite , Infecções por Orthomyxoviridae , Animais , Bovinos , Infecções por Orthomyxoviridae/veterinária , Infecções por Orthomyxoviridae/epidemiologia , Infecções por Orthomyxoviridae/virologia , Infecções por Orthomyxoviridae/diagnóstico , Surtos de Doenças/veterinária , Leite/virologia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Doenças dos Bovinos/diagnóstico , Anticorpos Antivirais/sangue , Vírus da Influenza A/isolamento & purificação , Vírus da Influenza A/genética , Vírus da Influenza A/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Ensaio de Imunoadsorção Enzimática/veterinária , Estados Unidos/epidemiologia , RNA Viral/genética , Indústria de Laticínios , Feminino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA