Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Cancers (Basel) ; 16(11)2024 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-38893282

RESUMEN

In the original publication [...].

2.
Virol Sin ; 39(3): 414-421, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677713

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, encodes several accessory proteins that have been shown to play crucial roles in regulating the innate immune response. However, their expressions in infected cells and immunogenicity in infected humans and mice are still not fully understood. This study utilized various techniques such as luciferase immunoprecipitation system (LIPS), immunofluorescence â€‹assay (IFA), and western â€‹blot (WB) to detect accessory protein-specific antibodies in sera of COVID-19 patients. Specific antibodies to proteins 3a, 3b, 7b, 8 and 9c can be detected by LIPS, but only protein 3a antibody was detected by IFA or WB. Antibodies against proteins 3a and 7b were only detected in ICU patients, which may serve as a marker for predicting disease progression. Further, we investigated the expression of accessory proteins in SARS-CoV-2-infected cells and identified the expressions of proteins 3a, 6, 7a, 8, and 9b. We also analyzed their ability to induce antibodies in immunized mice and found that only proteins 3a, 6, 7a, 8, 9b and 9c were able to induce measurable antibody productions, but these antibodies lacked neutralizing activities and did not protect mice from SARS-CoV-2 infection. Our findings validate the expression of SARS-CoV-2 accessory proteins and elucidate their humoral immune response, providing a basis for protein detection assays and their role in pathogenesis.


Asunto(s)
Anticuerpos Antivirales , COVID-19 , Modelos Animales de Enfermedad , Inmunidad Humoral , SARS-CoV-2 , Animales , Humanos , SARS-CoV-2/inmunología , COVID-19/inmunología , COVID-19/virología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Ratones , Femenino , Ratones Endogámicos BALB C , Masculino , Persona de Mediana Edad , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Adulto , Anciano
3.
Mol Genet Genomics ; 298(4): 823-836, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37059908

RESUMEN

Coronavirus 2019 (COVID-19) is a complex disease that affects billions of people worldwide. Currently, effective etiological treatment of COVID-19 is still lacking; COVID-19 also causes damages to various organs that affects therapeutics and mortality of the patients. Surveillance of the treatment responses and organ injury assessment of COVID-19 patients are of high clinical value. In this study, we investigated the characteristic fragmentation patterns and explored the potential in tissue injury assessment of plasma cell-free DNA in COVID-19 patients. Through recruitment of 37 COVID-19 patients, 32 controls and analysis of 208 blood samples upon diagnosis and during treatment, we report gross abnormalities in cfDNA of COVID-19 patients, including elevated GC content, altered molecule size and end motif patterns. More importantly, such cfDNA fragmentation characteristics reflect patient-specific physiological changes during treatment. Further analysis on cfDNA tissue-of-origin tracing reveals frequent tissue injuries in COVID-19 patients, which is supported by clinical diagnoses. Hence, our work demonstrates and extends the translational merit of cfDNA fragmentation pattern as valuable analyte for effective treatment monitoring, as well as tissue injury assessment in COVID-19.


Asunto(s)
COVID-19 , Ácidos Nucleicos Libres de Células , Humanos , COVID-19/diagnóstico , Ácidos Nucleicos Libres de Células/genética
4.
Cancers (Basel) ; 15(6)2023 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-36980779

RESUMEN

Cancer has been the first killer that threatens people's lives and health. Despite recent improvements in cancer treatment, metastasis continues to be the main reason for death from cancer. The functions of microbiome in cancer metastasis have been studied recently, and it is proved that microbiome can influence tumor metastasis, as well as positive or negative responses to therapy. Here, we summarize the mechanisms of microorganisms affecting cancer metastasis, which include epithelial-mesenchymal transition (EMT), immunity, fluid shear stress (FSS), and matrix metalloproteinases (MMPs). This review will not only give a further understanding of relationship between microbiome and cancer metastasis, but also provide a new perspective for the microbiome's application in cancer metastasis prevention, early detection, and treatment.

5.
Proc Natl Acad Sci U S A ; 120(4): e2202820120, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36652473

RESUMEN

Human coronavirus 229E (HCoV-229E) and NL63 (HCoV-NL63) are endemic causes of upper respiratory infections such as the "common cold" but may occasionally cause severe lower respiratory tract disease in the elderly and immunocompromised patients. There are no approved antiviral drugs or vaccines for these common cold coronaviruses (CCCoV). The recent emergence of COVID-19 and the possible cross-reactive antibody and T cell responses between these CCCoV and SARS-CoV-2 emphasize the need to develop experimental animal models for CCCoV. Mice are an ideal experimental animal model for such studies, but are resistant to HCoV-229E and HCoV-NL63 infections. Here, we generated 229E and NL63 mouse models by exogenous delivery of their receptors, human hAPN and hACE2 using replication-deficient adenoviruses (Ad5-hAPN and Ad5-hACE2), respectively. Ad5-hAPN- and Ad5-hACE2-sensitized IFNAR-/- and STAT1-/- mice developed pneumonia characterized by inflammatory cell infiltration with virus clearance occurring 7 d post infection. Ad5-hAPN- and Ad5-hACE2-sensitized mice generated virus-specific T cells and neutralizing antibodies after 229E or NL63 infection, respectively. Remdesivir and a vaccine candidate targeting spike protein of 229E and NL63 accelerated viral clearance of virus in these mice. 229E- and NL63-infected mice were partially protected from SARS-CoV-2 infection, likely mediated by cross-reactive T cell responses. Ad5-hAPN- and Ad5-hACE2-transduced mice are useful for studying pathogenesis and immune responses induced by HCoV-229E and HCoV-NL63 infections and for validation of broadly protective vaccines, antibodies, and therapeutics against human respiratory coronaviruses including SARS-CoV-2.


Asunto(s)
COVID-19 , Resfriado Común , Coronavirus Humano 229E , Coronavirus Humano NL63 , Humanos , Animales , Ratones , Anciano , SARS-CoV-2 , Protección Cruzada
6.
Genome Res ; 32(2): 228-241, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35064006

RESUMEN

The pathogenesis of COVID-19 is still elusive, which impedes disease progression prediction, differential diagnosis, and targeted therapy. Plasma cell-free RNAs (cfRNAs) carry unique information from human tissue and thus could point to resourceful solutions for pathogenesis and host-pathogen interactions. Here, we performed a comparative analysis of cfRNA profiles between COVID-19 patients and healthy donors using serial plasma. Analyses of the cfRNA landscape, potential gene regulatory mechanisms, dynamic changes in tRNA pools upon infection, and microbial communities were performed. A total of 380 cfRNA molecules were up-regulated in all COVID-19 patients, of which seven could serve as potential biomarkers (AUC > 0.85) with great sensitivity and specificity. Antiviral (NFKB1A, IFITM3, and IFI27) and neutrophil activation (S100A8, CD68, and CD63)-related genes exhibited decreased expression levels during treatment in COVID-19 patients, which is in accordance with the dynamically enhanced inflammatory response in COVID-19 patients. Noncoding RNAs, including some microRNAs (let 7 family) and long noncoding RNAs (GJA9-MYCBP) targeting interleukin (IL6/IL6R), were differentially expressed between COVID-19 patients and healthy donors, which accounts for the potential core mechanism of cytokine storm syndromes; the tRNA pools change significantly between the COVID-19 and healthy group, leading to the accumulation of SARS-CoV-2 biased codons, which facilitate SARS-CoV-2 replication. Finally, several pneumonia-related microorganisms were detected in the plasma of COVID-19 patients, raising the possibility of simultaneously monitoring immune response regulation and microbial communities using cfRNA analysis. This study fills the knowledge gap in the plasma cfRNA landscape of COVID-19 patients and offers insight into the potential mechanisms of cfRNAs to explain COVID-19 pathogenesis.


Asunto(s)
COVID-19 , Ácidos Nucleicos Libres de Células , ARN/sangre , COVID-19/sangre , COVID-19/genética , Ácidos Nucleicos Libres de Células/sangre , Síndrome de Liberación de Citoquinas , Humanos , SARS-CoV-2
7.
J Virol ; 96(3): e0184221, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-34817197

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) is a beta coronavirus that emerged in 2012, causing severe pneumonia and renal failure. MERS-CoV encodes five accessory proteins. Some of them have been shown to interfere with host antiviral immune response. However, the roles of protein 8b in innate immunity and viral virulence was rarely studied. Here, we introduced individual MERS-CoV accessory protein genes into the genome of an attenuated murine coronavirus (Mouse hepatitis virus, MHV), respectively, and found accessory protein 8b could enhance viral replication in vivo and in vitro and increase the lethality of infected mice. RNA-seq analysis revealed that protein 8b could significantly inhibit type I interferon production (IFN-I) and innate immune response in mice infected with MHV expressing protein 8b. We also found that MERS-CoV protein 8b could initiate from multiple internal methionine sites and at least three protein variants were identified. Residues 1-23 of protein 8b was demonstrated to be responsible for increased virulence in vivo. In addition, the inhibitory effect on IFN-I of protein 8b might not contribute to its virulence enhancement as aa1-23 deletion did not affect IFN-I production in vitro and in vivo. Next, we also found that protein 8b was localized to the endoplasmic reticulum (ER)/Golgi membrane in infected cells, which was disrupted by C-terminal region aa 88-112 deletion. This study will provide new insight into the pathogenesis of MERS-CoV infection. IMPORTANCE Multiple coronaviruses (CoV) cause severe respiratory infections and become global public health threats such as SARS-CoV, MERS-CoV, and SARS-CoV-2. Each coronavirus contains different numbers of accessory proteins which show high variability among different CoVs. Accessory proteins are demonstrated to play essential roles in pathogenesis of CoVs. MERS-CoV contains 5 accessory proteins (protein 3, 4a, 4b, 5, 8b), and deletion of all four accessory proteins (protein 3, 4a, 4b, 5), significantly affects MERS-CoV replication and pathogenesis. However, whether ORF8b also regulates MERS-CoV infection is unknown. Here, we constructed mouse hepatitis virus (MHV) recombinant virus expressing MERS-CoV protein 8b and demonstrated protein 8b could significantly enhance the virulence of MHV, which is mediated by N-terminal domain of protein 8b. This study will shed light on the understanding of pathogenesis of MERS-CoV infection.


Asunto(s)
Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Virus de la Hepatitis Murina/fisiología , Dominios y Motivos de Interacción de Proteínas , Proteínas Reguladoras y Accesorias Virales/genética , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Ratones , Mortalidad , Proteínas Reguladoras y Accesorias Virales/química , Tropismo Viral , Virulencia/genética , Factores de Virulencia/genética
9.
Signal Transduct Target Ther ; 6(1): 155, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33859163

RESUMEN

Disease progression prediction and therapeutic drug target discovery for Coronavirus disease 2019 (COVID-19) are particularly important, as there is still no effective strategy for severe COVID-19 patient treatment. Herein, we performed multi-platform omics analysis of serial plasma and urine samples collected from patients during the course of COVID-19. Integrative analyses of these omics data revealed several potential therapeutic targets, such as ANXA1 and CLEC3B. Molecular changes in plasma indicated dysregulation of macrophage and suppression of T cell functions in severe patients compared to those in non-severe patients. Further, we chose 25 important molecular signatures as potential biomarkers for the prediction of disease severity. The prediction power was validated using corresponding urine samples and plasma samples from new COVID-19 patient cohort, with AUC reached to 0.904 and 0.988, respectively. In conclusion, our omics data proposed not only potential therapeutic targets, but also biomarkers for understanding the pathogenesis of severe COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/sangre , Descubrimiento de Drogas , Lipidómica , Proteómica , SARS-CoV-2/metabolismo , Biomarcadores/sangre , Femenino , Humanos , Masculino
10.
J Virol ; 95(12)2021 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-33827945

RESUMEN

Immune memory represents the most efficient defense against invasion and transmission of infectious pathogens. In contrast to memory T and B cells, the roles of innate immunity in recall responses remain inconclusive. In this study, we identified a novel mouse spleen NK cell subset expressing NKp46 and NKG2A induced by intranasal influenza virus infection. These memory NK cells specifically recognize N-linked glycosylation sites on influenza hemagglutinin (HA) protein. Different from memory-like NK cells reported previously, these NKp46+ NKG2A+ memory NK cells exhibited HA-specific silence of cytotoxicity but increase of gamma interferon (IFN-γ) response against influenza virus-infected cells, which could be reversed by pifithrin-µ, a p53-heat shock protein 70 (HSP70) signaling inhibitor. During recall responses, splenic NKp46+ NKG2A+ NK cells were recruited to infected lung and modulated viral clearance of virus and CD8+ T cell distribution, resulting in improved clinical outcomes. This long-lived NK memory bridges innate and adaptive immune memory response and promotes the homeostasis of local environment during recall response.IMPORTANCE In this study, we demonstrate a novel hemagglutinin (HA)-specific NKp46+ NKG2A+ NK cell subset induced by influenza A virus infection. These memory NK cells show virus-specific decreased cytotoxicity and increased gamma interferon (IFN-γ) on reencountering the same influenza virus antigen. In addition, they modulate host recall responses and CD8 T cell distribution, thus bridging the innate immune and adaptive immune responses during influenza virus infection.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Memoria Inmunológica , Subtipo H1N1 del Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Infecciones por Orthomyxoviridae/inmunología , Traslado Adoptivo , Animales , Antígenos Ly/análisis , Antígenos Ly/metabolismo , Benzotiazoles/farmacología , Linfocitos T CD8-positivos/inmunología , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Subtipo H9N2 del Virus de la Influenza A/inmunología , Interferón gamma/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Subfamília C de Receptores Similares a Lectina de Células NK/análisis , Receptor 1 Gatillante de la Citotoxidad Natural/análisis , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Bazo/citología , Bazo/inmunología , Tolueno/análogos & derivados , Tolueno/farmacología
11.
J Exp Med ; 218(4)2021 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-33464307

RESUMEN

Virus-specific T cells play essential roles in protection against multiple virus infections, including SARS-CoV and MERS-CoV. While SARS-CoV-2-specific T cells have been identified in COVID-19 patients, their role in the protection of SARS-CoV-2-infected mice is not established. Here, using mice sensitized for infection with SARS-CoV-2 by transduction with an adenovirus expressing the human receptor (Ad5-hACE2), we identified SARS-CoV-2-specific T cell epitopes recognized by CD4+ and CD8+ T cells in BALB/c and C57BL/6 mice. Virus-specific T cells were polyfunctional and were able to lyse target cells in vivo. Further, type I interferon pathway was proved to be critical for generating optimal antiviral T cell responses after SARS-CoV-2 infection. T cell vaccination alone partially protected SARS-CoV-2-infected mice from severe disease. In addition, the results demonstrated cross-reactive T cell responses between SARS-CoV and SARS-CoV-2, but not MERS-CoV, in mice. Understanding the role of the T cell response will guide immunopathogenesis studies of COVID-19 and vaccine design and validation.


Asunto(s)
COVID-19/inmunología , Epítopos de Linfocito T/inmunología , Interacciones Huésped-Patógeno/fisiología , Linfocitos T/inmunología , Linfocitos T/virología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Anticuerpos Neutralizantes/sangre , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Chlorocebus aethiops , Reacciones Cruzadas , Mapeo Epitopo , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Células Vero
12.
Cell ; 182(3): 734-743.e5, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32643603

RESUMEN

COVID-19, caused by SARS-CoV-2, is a virulent pneumonia, with >4,000,000 confirmed cases worldwide and >290,000 deaths as of May 15, 2020. It is critical that vaccines and therapeutics be developed very rapidly. Mice, the ideal animal for assessing such interventions, are resistant to SARS-CoV-2. Here, we overcome this difficulty by exogenous delivery of human ACE2 with a replication-deficient adenovirus (Ad5-hACE2). Ad5-hACE2-sensitized mice developed pneumonia characterized by weight loss, severe pulmonary pathology, and high-titer virus replication in lungs. Type I interferon, T cells, and, most importantly, signal transducer and activator of transcription 1 (STAT1) are critical for virus clearance and disease resolution in these mice. Ad5-hACE2-transduced mice enabled rapid assessments of a vaccine candidate, of human convalescent plasma, and of two antiviral therapies (poly I:C and remdesivir). In summary, we describe a murine model of broad and immediate utility to investigate COVID-19 pathogenesis and to evaluate new therapies and vaccines.


Asunto(s)
Betacoronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/prevención & control , Modelos Animales de Enfermedad , Pandemias/prevención & control , Neumonía Viral/patología , Neumonía Viral/prevención & control , Vacunación , Enzima Convertidora de Angiotensina 2 , Animales , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Evaluación Preclínica de Medicamentos/métodos , Femenino , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/virología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , SARS-CoV-2 , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Organismos Libres de Patógenos Específicos , Transducción Genética , Células Vero , Carga Viral , Replicación Viral
13.
J Clin Invest ; 130(10): 5235-5244, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32634129

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent for coronavirus 2019 (COVID-19) pneumonia. Little is known about the kinetics, tissue distribution, cross-reactivity, and neutralization antibody response in patients with COVID-19. Two groups of patients with RT-PCR-confirmed COVID-19 were enrolled in this study: 12 severely ill patients in intensive care units who needed mechanical ventilation and 11 mildly ill patients in isolation wards. Serial clinical samples were collected for laboratory detection. Results showed that most of the severely ill patients had viral shedding in a variety of tissues for 20-40 days after onset of disease (8/12, 66.7%), while the majority of mildly ill patients had viral shedding restricted to the respiratory tract and had no detectable virus RNA 10 days after onset (9/11, 81.8%). Mildly ill patients showed significantly lower IgM response compared with that of the severe group. IgG responses were detected in most patients in both the severe and mild groups at 9 days after onset, and remained at a high level throughout the study. Antibodies cross-reactive to SARS-CoV and SARS-CoV-2 were detected in patients with COVID-19 but not in patients with MERS. High levels of neutralizing antibodies were induced after about 10 days after onset in both severely and mildly ill patients which were higher in the severe group. SARS-CoV-2 pseudotype neutralization test and focus reduction neutralization test with authentic virus showed consistent results. Sera from patients with COVID-19 inhibited SARS-CoV-2 entry. Sera from convalescent patients with SARS or Middle East respiratory syndrome (MERS) did not. Anti-SARS-CoV-2 S and N IgG levels exhibited a moderate correlation with neutralization titers in patients' plasma. This study improves our understanding of immune response in humans after SARS-CoV-2 infection.


Asunto(s)
Anticuerpos Antivirales/sangre , Betacoronavirus/metabolismo , Infecciones por Coronavirus/sangre , Neumonía Viral/sangre , Carga Viral , Esparcimiento de Virus , Adulto , Anciano , Especificidad de Anticuerpos , COVID-19 , Reacciones Cruzadas , Femenino , Humanos , Cinética , Masculino , Persona de Mediana Edad , Pandemias , SARS-CoV-2 , Índice de Severidad de la Enfermedad
14.
Signal Transduct Target Ther ; 5(1): 74, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32488072

RESUMEN

Influenza epidemics and pandemics are constant threats to global public health. Although strategies including vaccines and antiviral drugs have achieved great advances in controlling influenza virus infection, the efficacy of these strategies is limited by the highly frequent mutations in the viral genome and the emergence of drug-resistant strains. Our previous study indicated that boosting the immunity of human Vγ9Vδ2-T cells with the phosphoantigen pamidronate could be a therapeutic strategy to treat seasonal and avian influenza virus infections. However, one notable drawback of γδ-T cell-based immunotherapy is the rapid exhaustion of proliferation and effector responses due to repeated treatments with phosphoantigens. Here, we found that the expression of CD137 was inducible in Vγ9Vδ2-T cells following antigenic stimulation. CD137+ Vγ9Vδ2-T cells displayed more potent antiviral activity against influenza virus than their CD137- counterparts in vitro and in Rag2-/- γc-/- mice. We further demonstrated that CD137 costimulation was essential for Vγ9Vδ2-T cell activation, proliferation, survival and effector functions. In humanized mice reconstituted with human peripheral blood mononuclear cells, CD137 costimulation with a recombinant human CD137L protein boosted the therapeutic effects of pamidronate against influenza virus. Our study provides a novel strategy of targeting CD137 to improve the efficacy of Vγ9Vδ2-T cell-based immunotherapy.


Asunto(s)
Inmunidad Celular , Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Humanos , Inmunoterapia , Gripe Humana/terapia , Ratones , Ratones Noqueados , Linfocitos T/trasplante
15.
Emerg Microbes Infect ; 9(1): 991-993, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32342724

RESUMEN

SARS-CoV-2 caused a major outbreak of severe pneumonia (COVID-19) in humans. Viral RNA was detected in multiple organs in COVID-19 patients. However, infectious SARS-CoV-2 was only isolated from respiratory specimens. Here, infectious SARS-CoV-2 was successfully isolated from urine of a COVID-19 patient. The virus isolated could infect new susceptible cells and was recognized by its' own patient sera. Appropriate precautions should be taken to avoid transmission from urine.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Infecciones por Coronavirus/orina , Infecciones por Coronavirus/virología , Neumonía Viral/orina , Neumonía Viral/virología , Anciano , Animales , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/transmisión , Genoma Viral/genética , Humanos , Masculino , Pandemias , Neumonía Viral/transmisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , SARS-CoV-2 , Células Vero
16.
Cell Mol Immunol ; 15(9): 827-837, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-28413216

RESUMEN

Natural killer (NK) cells are indispensable components of both the innate and adaptive immune response. However, their precise roles in the cross-talk between innate and adaptive immunity during influenza virus infection remain controversial. By comparing NK cell dynamics and activity under a sub-lethal dose and high dose of influenza virus infection, we showed that influenza virus PR8 directly infected NK cells during natural infection, which was consistent with our previous findings obtained from an in vitro investigation of human NK cells. The impairments in cytotoxicity and IFN-γ production by spleen NK cells following high-dose infection were accompanied by decreased virus-specific killing mediated by cytotoxic T lymphocytes (CTLs). Importantly, the weakened CTL activity could be reversed by adoptive transfer of spleen NK cells harvested from low-dose-infected mice but not healthy donors. Taken together, our data provide direct evidence supporting the contribution of NK cells to antiviral T-cell responses. This study also indicates that a novel NK-targeted immune evasion strategy is used by influenza virus to shrink both innate and adaptive immune responses.


Asunto(s)
Virus de la Influenza A/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos , Infecciones por Orthomyxoviridae/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Femenino , Células Asesinas Naturales/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/patología , Linfocitos T Citotóxicos/patología
17.
Am J Respir Cell Mol Biol ; 58(6): 684-695, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29220578

RESUMEN

Pulmonary fibrosis is a chronic progressive lung disease with few treatments. Human mesenchymal stem cells (MSCs) have been shown to be beneficial in pulmonary fibrosis because they have immunomodulatory capacity. However, there is no reliable model to test the therapeutic effect of human MSCs in vivo. To mimic pulmonary fibrosis in humans, we established a novel bleomycin-induced pulmonary fibrosis model in humanized mice. With this model, the benefit of human MSCs in pulmonary fibrosis and the underlying mechanisms were investigated. In addition, the relevant parameters in patients with pulmonary fibrosis were examined. We demonstrate that human CD8+ T cells were critical for the induction of pulmonary fibrosis in humanized mice. Human MSCs could alleviate pulmonary fibrosis and improve lung function by suppressing bleomycin-induced human T-cell infiltration and proinflammatory cytokine production in the lungs of humanized mice. Importantly, alleviation of pulmonary fibrosis by human MSCs was mediated by the PD-1/programmed death-ligand 1 pathway. Moreover, abnormal PD-1 expression was found in circulating T cells and lung tissues of patients with pulmonary fibrosis. Our study supports the potential benefit of targeting the PD-1/programmed death-ligand 1 pathway in the treatment of pulmonary fibrosis.


Asunto(s)
Antígeno B7-H1/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/fisiología , Receptor de Muerte Celular Programada 1/metabolismo , Fibrosis Pulmonar/terapia , Animales , Bleomicina/toxicidad , Linfocitos T CD4-Positivos/patología , Modelos Animales de Enfermedad , Humanos , Leucocitos Mononucleares/trasplante , Redes y Vías Metabólicas , Ratones Mutantes , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología
18.
Mol Med Rep ; 14(6): 5237-5244, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27779668

RESUMEN

Cancer-associated antigen 215 (CA215) is an immunoglobulin molecule expressed by numerous tumor types. Membrane­bound and soluble CA215 have been detected in the majority of cancer cells and rarely identified in normal tissues. In addition, CA215C is a carbohydrate­associated epitope in the variable region of CA215, which is specifically recognized by the monoclonal antibody, RP215. However, CA215C is not a suitable vaccine candidate as it is a thymus­independent antigen. In the present study, RP215 was used as a target to screen short peptide mimics of CA215C from a phage display peptide library. Following three rounds of screening, 30 positive phage clones that specifically bound to RP215 were identified and sequenced. The result of amino­acid sequence analysis revealed five conserved sequence groups for seventeen of the positive phage clones. The sequences of phage clones 2, 13 and 42 were selected for peptide synthesis and binding analysis. The synthetic peptides R2 and R42 specifically bound RP215. Antisera from mice immunized with R2­BSA or R42­BSA bound purified CA215C and innate CA215C expressed on human hepatic and rectal carcinoma tissues, as demonstrated by immunohistochemistry. Furthermore, R2­BSA and R42­BSA antisera inhibited RP215 binding to cancer tissues. These results revealed that R2­BSA and R42­BSA antisera had similar characteristics to RP215 and that the synthetic peptides R2 and R42 may mimic the CA215C epitope. R2 and R42 peptides may therefore have potential for development into a tumor vaccine.


Asunto(s)
Antígenos de Neoplasias/inmunología , Imitación Molecular/inmunología , Neoplasias/inmunología , Péptidos/inmunología , Secuencia de Aminoácidos , Animales , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/inmunología , Técnicas de Visualización de Superficie Celular , Mapeo Epitopo , Femenino , Humanos , Sueros Inmunes , Ratones , Neoplasias/genética , Neoplasias/terapia , Biblioteca de Péptidos , Unión Proteica/inmunología
19.
J Med Microbiol ; 62(Pt 7): 980-987, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23579397

RESUMEN

Mycobacterium bovis Bacille Calmette-Guérin (BCG) is the current vaccine used against Mycobacterium tuberculosis (MTB) infection. However, exposure to environmental pathogens, such as Mycobacterium avium, interferes with the immune response induced by BCG vaccination. How M. avium affects the efficiency of BCG is unclear. In this study, BCG-vaccinated mice pre-treated with M. avium-derived lipids (MALs) showed a higher mycobacterial load and increased infiltration of inflammatory cells compared to control mice treated with Escherichia coli-derived lipids (ELs). Unexpectedly, there were no changes in cell proliferation or IFN-γ levels in spleen cells stimulated with protein purified derivatives (PPD) or heat-inactivated BCG in MALs-treated mice. However, pre-treatment with MALs decreased the bactericidal effect as well as the production of TNF-α and nitric oxide (NO) in murine macrophages from BCG-vaccinated mice stimulated with IFN-γ. These results suggest that MAL pre-treatment dampens the immune response against MTB and that this dampening is associated with a decreased response to IFN-γ stimulation in murine macrophages. T-lymphocyte responses, however, were unaffected.


Asunto(s)
Vacuna BCG/inmunología , Interferón gamma/farmacología , Lípidos/farmacología , Macrófagos/efectos de los fármacos , Mycobacterium avium/metabolismo , Animales , Femenino , Metabolismo de los Lípidos , Lípidos/química , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico , Nitritos , Reacción en Cadena en Tiempo Real de la Polimerasa , Organismos Libres de Patógenos Específicos
20.
Blood Cells Mol Dis ; 49(3-4): 128-32, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22677106

RESUMEN

Southeast Asian deletion (--(SEA)) α-thalassemia is an inherited monogenic disorder of human hemoglobin, and embryonic globin ζ (hemoglobin ζ, zeta globin chain or Hb zeta chain) has been shown to be a marker that can be used for the identification of carriers of the (--(SEA)) α-thalassemia deletion. In this work, a fluorescence immunochromatographic assay (FL-ICA) was established to detect the zeta globin chain in the hemolysates of carriers of the (--(SEA)) α-thalassemia deletion. This assay can be completed within 10min using a simple UV detector and does not suffer from interference from the red background color of the hemolysate. A total of 314 blood samples were tested by FL-ICA and ELISA. The results of these assays were confirmed by PCR, the standard technique for genetic disease testing. The sensitivity and specificity of this novel FL-ICA were 100% and 98.0%, respectively; the corresponding values for the ELISA performed simultaneously were 100% and 99.2%, respectively. In conclusion, a new FL-ICA-a simple, fast, convenient, low-cost method-was developed that may be useful in both high-throughput screening and individual detection of the (--(SEA)) α-thalassemia deletion in carriers. Additionally, this qualitative FL-ICA may enlighten the development of a new systems for analysis of other target molecules using whole-blood samples.


Asunto(s)
Cromatografía de Afinidad , Talasemia alfa/diagnóstico , Talasemia alfa/genética , Globinas zeta/análisis , Adulto , Anticuerpos Monoclonales , Asia Sudoriental , Secuencia de Bases , Estudios de Casos y Controles , Ensayo de Inmunoadsorción Enzimática , Fluorescencia , Pruebas Genéticas , Heterocigoto , Homocigoto , Humanos , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Sensibilidad y Especificidad , Eliminación de Secuencia , Talasemia alfa/sangre , Globinas zeta/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA