Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.151
Filtrar
1.
Virol J ; 21(1): 220, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39285440

RESUMEN

BACKGROUND: Human adenovirus type 55 (hAd55) infection can lead to acute respiratory diseases that often present with severe symptoms. Despite its persistent prevalence in military camps and communities, there are no commercially available vaccines or vaccine candidates undergoing clinical evaluation; therefore, there is an urgent need to address this. In this study, we evaluated the immunogenicity of inactivated hAd55 isolates and investigated the effects of adjuvants and various immunization intervals. METHODS AND RESULTS: To select a vaccine candidate, four hAd55 strains (6-9, 6-15 (AFMRI 41014), 28-48 (AFMRI 41013), and 12-164 (AFMRI 41012)) were isolated from infected patients in military camps. Sequence analysis revealed no variation in the coding regions of structural proteins, including pentons, hexons, and fibers. Immunization with inactivated hAd55 isolates elicited robust hAd55-specific binding and neutralizing antibody responses in mice, with adjuvants, particularly alum hydroxide (AH), enhancing antibody titers. Co-immunization with AH also induced hAd14-specific neutralizing antibody responses but did not induce hAd11-specific neutralizing antibody responses. Notably, booster immunization administered at a four-week interval resulted in superior immune responses compared with shorter immunization intervals. CONCLUSIONS: Prime-boost immunization with the inactivated hAd55 isolate and an AH adjuvant shows promise as a potential approach for preventing hAd55-induced respiratory disease. Further research is needed to evaluate the efficacy and safety of these vaccine candidates in preventing hAd55-associated respiratory illnesses.


Asunto(s)
Adenovirus Humanos , Adyuvantes Inmunológicos , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Inmunización Secundaria , Vacunas de Productos Inactivados , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Ratones , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Humanos , Adenovirus Humanos/inmunología , Adenovirus Humanos/genética , Adyuvantes Inmunológicos/administración & dosificación , Vacunas de Productos Inactivados/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Femenino , Vacunas contra el Adenovirus/inmunología , Vacunas contra el Adenovirus/administración & dosificación , Ratones Endogámicos BALB C , Adyuvantes de Vacunas/administración & dosificación , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/prevención & control , Infecciones por Adenovirus Humanos/virología
2.
Front Immunol ; 15: 1432226, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39139562

RESUMEN

Introduction: The early transcription unit 3 (E3) of human adenoviruses (HAdVs) encodes several immunoevasins, including the E3/49K protein, which is unique for species D of HAdVs. It is expressed as surface transmembrane protein and shed. E3/49K of HAdV-D64 binds to the protein tyrosine phosphatase surface receptor CD45, thereby modulating activation of T and NK cells. Methods: Considering that E3/49K represents the most polymorphic viral protein among species D HAdVs, we demonstrate here that all tested E3/49K orthologs bind to the immunologically important regulator CD45. Thus, this feature is conserved regardless of the pathological associations of the respective HAdV types. Results: It appeared that modulation of CD45 is a unique property restricted to HAdVs of species D. Moreover, E3/49K treatment inhibited B cell receptor (BCR) signaling and impaired BCR signal phenotypes. The latter were highly comparable to B cells having defects in the expression of CD45, suggesting E3/49K as a potential tool to investigate CD45 specific functions. Conclusion: We identified B cells as new direct target of E3/49K-mediated immune modulation, representing a novel viral immunosubversive mechanism.


Asunto(s)
Proteínas E3 de Adenovirus , Adenovirus Humanos , Linfocitos B , Antígenos Comunes de Leucocito , Receptores de Antígenos de Linfocitos B , Transducción de Señal , Humanos , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos B/inmunología , Transducción de Señal/inmunología , Antígenos Comunes de Leucocito/metabolismo , Antígenos Comunes de Leucocito/inmunología , Adenovirus Humanos/inmunología , Proteínas E3 de Adenovirus/inmunología , Proteínas E3 de Adenovirus/metabolismo , Proteínas E3 de Adenovirus/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/virología , Infecciones por Adenovirus Humanos/metabolismo , Células HEK293
3.
Transpl Immunol ; 86: 102093, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39032616

RESUMEN

Human adenovirus infection (HAdV) may be fatal in patients undergoing allogeneic hematopoietic cell transplantation (HCT). Cidofovir is effective in only a part of the post-HCT HAdV infection. Therefore, posttransplant immune reconstitution is important for HAdV clearance. We describe the detailed immune reconstitution and response of adenovirus-specific T cells in a patient with inborn errors of immunity who had disseminated HAdV infection with hepatitis post-HCT and was treated with cidofovir. Though the patient received cidofovir for only 19 days starting from Day 72 after HCT because of renal dysfunction, we observed T-cell reconstitution, a decrease in HAdV copy number, and amelioration of the symptoms of HAdV infection after Day 90. We initially observed expanded NK and CD8+CD45RO+ memory subsets and later gradual increase of naïve T cells eveloped after cessation of cidofovir treatment. An increase in adenovirus-specific IFN-γ secretion from 2 to 4 months after HCT was confirmed by ELISpot assay. The progression of immune reconstitution and cidofovir treatment are considered to have contributed to survival in this patient. Optimization of transplantation methods, prompt appropriate antiviral medication, and virus-specific T-cell therapy would be necessary as the better strategy for systemic HAdV infection.


Asunto(s)
Infecciones por Adenovirus Humanos , Antivirales , Cidofovir , Citosina , Trasplante de Células Madre Hematopoyéticas , Reconstitución Inmune , Organofosfonatos , Humanos , Cidofovir/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Organofosfonatos/uso terapéutico , Citosina/análogos & derivados , Citosina/uso terapéutico , Infecciones por Adenovirus Humanos/tratamiento farmacológico , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/terapia , Antivirales/uso terapéutico , Trasplante Homólogo , Adenovirus Humanos/inmunología , Masculino , Hepatitis Viral Humana/tratamiento farmacológico , Hepatitis Viral Humana/inmunología
4.
Int J Pharm ; 662: 124480, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39038719

RESUMEN

Adenovirus (Ad) vectors based on human adenovirus serotype 5 (Ad5) have attracted significant attention as vaccine vectors for infectious diseases. However, the effectiveness of Ad5 vectors as vaccines is often inhibited by the anti-Ad5 neutralizing antibodies retained by many adults. To overcome this drawback, we focused on human adenovirus serotype 35 (Ad35) vectors with low seroprevalence in adults. Although Ad35 vectors can circumvent anti-Ad5 neutralizing antibodies, vector yields of Ad35 vectors are often inferior to those of Ad5 vectors. In this study, we developed novel Ad35 vectors containing the Ad5 E4 orf 4, 6, and 6/7 or the Ad5 E4 orf 6 and 6/7 for efficient vector production, and compared their properties. These E4-modified Ad35 vectors efficiently propagated to a similar extent at virus titers comparable to those of Ad5 vectors. An Ad35 vector containing the Ad5 E4 orf 4, 6, and 6/7 mediated more efficient transduction than that containing the Ad5 E4 orf 6 and 6/7 in human cultured cells. Furthermore, insertion of an arginine-glycine-aspartate (RGD) peptide in the fiber region of an Ad35 vector containing the Ad5 E4 orf 4, 6, and 6/7 significantly improved the transgene product-specific antibody production following intramuscular administration in mice. The Ad35 vector containing the RGD peptide mediated efficient vaccine effects even in the mice pre-immunized with an Ad5.


Asunto(s)
Adenovirus Humanos , Vectores Genéticos , Oligopéptidos , Animales , Humanos , Oligopéptidos/química , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Femenino , Ratones , Serogrupo , Ratones Endogámicos BALB C , Vacunas contra el Adenovirus/inmunología , Vacunas contra el Adenovirus/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/sangre , Células HEK293 , Proteínas E4 de Adenovirus/inmunología , Proteínas E4 de Adenovirus/genética , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/sangre
5.
mBio ; 15(8): e0103824, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38940561

RESUMEN

Human adenoviruses (HAdVs) are small DNA viruses that generally cause mild disease. Certain strains, particularly those belonging to species B HAdVs, can cause severe pneumonia and have a relatively high mortality rate. Little is known about the molecular aspects of how these highly pathogenic species affect the infected cell and how they suppress innate immunity. The present study provides molecular insights into how species B adenoviruses suppress the interferon signaling pathway. Our study shows that these viruses, unlike HAdV-C2, are resistant to type I interferon. This resistance likely arises due to the highly efficient suppression of interferon-stimulated gene expression. Unlike in HAdV-C2, HAdV-B7 and B14 sequester STAT2 and RNA polymerase II from interferon-stimulated gene promoters in infected cells. This results in suppressed interferon- stimulated gene activation. In addition, we show that RuvBL1 and RuvBL2, cofactors important for RNA polymerase II recruitment to promoters and interferon-stimulated gene activation, are redirected to the cytoplasm forming high molecular weight complexes that, likely, are unable to associate with chromatin. Proteomic analysis also identified key differences in the way these viruses affect the host cell, providing insights into species B-associated high pathogenicity. Curiously, we observed that at the level of protein expression changes to the infected cell, HAdV-C2 and B7 were more similar than those of the same species, B7 and B14. Collectively, our study represents the first such study of innate immune suppression by the highly pathogenic HAdV-B7 and B14, laying an important foundation for future investigations.IMPORTANCEHuman adenoviruses form a large family of double-stranded DNA viruses known for a variety of usually mild diseases. Certain strains of human adenovirus cause severe pneumonia leading to much higher mortality and morbidity than most other strains. The reasons for this enhanced pathogenicity are unknown. Our study provides a molecular investigation of how these highly pathogenic strains might inactivate the interferon signaling pathway, highlighting the lack of sensitivity of these viruses to type I interferon in general while providing a global picture of how viral changes in cellular proteins drive worse disease outcomes.


Asunto(s)
Adenovirus Humanos , Interferón Tipo I , Humanos , Adenovirus Humanos/genética , Adenovirus Humanos/patogenicidad , Adenovirus Humanos/fisiología , Adenovirus Humanos/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Interferón Tipo I/genética , Factor de Transcripción STAT2/metabolismo , Factor de Transcripción STAT2/genética , Inmunidad Innata , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Transducción de Señal , Infecciones por Adenovirus Humanos/virología , Infecciones por Adenovirus Humanos/inmunología , Virulencia , Interacciones Huésped-Patógeno/inmunología , Animales , Regiones Promotoras Genéticas , Evasión Inmune , Células A549
6.
Vaccine ; 42(22): 126006, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-38806354

RESUMEN

BACKGROUND: Human adenovirus (HAdV) is a prevalent causative agent of acute respiratory disease (ARD) and is frequently responsible for outbreaks, particularly in military environments. Current vaccines do not effectively cover HAdV subtypes commonly found among Korean military personnel, highlighting the need for a new targeted vaccine. This study presents a cost-benefit analysis to evaluate the economic viability of developing and implementing such a vaccine within a military context. METHODS: We adopted a societal perspective for this cost-benefit analysis, which included estimating costs associated with vaccine development, production, and distribution over a projected timeline. We assumed a development period of five years, after which vaccine production and administration were initiated in the sixth year. The cost associated with vaccine development, production, and dispensation was considered. The benefits were calculated based on both direct and indirect cost savings from preventing HAdV infections through vaccination. All financial figures were expressed in 2023 US dollars. A sensitivity analysis was conducted to explore the impact of varying factors such as vaccination rate, incidence of infection, vaccine efficacy, and discount rate. RESULTS: For the base case scenario, we assumed a vaccination rate of 100 %, an incidence rate of 0.02, and a vaccine efficacy of 95 %, applying a 3 % discount rate. Initially, in the sixth year, the benefit-cost ratio stood at 0.71, suggesting a cost disadvantage at the onset of vaccination. However, this ratio improved to 1.32 in the following years, indicating a cost benefit from the seventh year onward. The cumulative benefit-cost ratio over a decade reached 2.72. The outcomes from the sensitivity analysis were consistent with these findings. CONCLUSION: Our cost-benefit analysis demonstrates that the introduction of an HAdV vaccine for the Korean military is economically advantageous, with substantial cost benefits accruing from the seventh year after the commencement of vaccination.


Asunto(s)
Infecciones por Adenovirus Humanos , Vacunas contra el Adenovirus , Adenovirus Humanos , Análisis Costo-Beneficio , Personal Militar , Humanos , República de Corea/epidemiología , Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/prevención & control , Infecciones por Adenovirus Humanos/economía , Infecciones por Adenovirus Humanos/epidemiología , Vacunas contra el Adenovirus/inmunología , Vacunas contra el Adenovirus/economía , Desarrollo de Vacunas , Vacunación/economía , Eficacia de las Vacunas
7.
Int J Mol Sci ; 25(8)2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38673764

RESUMEN

The exacerbation of pneumonia in children with human adenovirus type 3 (HAdV-3E) is secondary to a Staphylococcus aureus (S. aureus) infection. The influence of host-pathogen interactions on disease progression remains unclear. It is important to note that S. aureus infections following an HAdV-3E infection are frequently observed in clinical settings, yet the underlying susceptibility mechanisms are not fully understood. This study utilized an A549 cell model to investigate secondary infection with S. aureus following an HAdV-3E infection. The findings suggest that HAdV-3E exacerbates the S. aureus infection by intensifying lung epithelial cell damage. The results highlight the role of HAdV-3E in enhancing the interferon signaling pathway through RIG-I (DDX58), resulting in the increased expression of interferon-stimulating factors like MX1, RSAD2, and USP18. The increase in interferon-stimulating factors inhibits the NF-κB and MAPK/P38 pro-inflammatory signaling pathways. These findings reveal new mechanisms of action for HAdV-3E and S. aureus in secondary infections, enhancing our comprehension of pathogenesis.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Proteína 58 DEAD Box , Transducción de Señal , Infecciones Estafilocócicas , Staphylococcus aureus , Humanos , Células A549 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/fisiología , Adenovirus Humanos/inmunología , Coinfección/microbiología , Proteína 58 DEAD Box/metabolismo , Interacciones Huésped-Patógeno/inmunología , Inflamación/metabolismo , FN-kappa B/metabolismo , Receptores Inmunológicos/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/patogenicidad , Ubiquitina Tiolesterasa
8.
Front Immunol ; 15: 1294898, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38660301

RESUMEN

Human adenovirus type 7 (HAdV-7) is a significant viral pathogen that causes respiratory infections in children. Currently, there are no specific antiviral drugs or vaccines for children targeting HAdV-7, and the mechanisms of its pathogenesis remain unclear. The NLRP3 inflammasome-driven inflammatory cascade plays a crucial role in the host's antiviral immunity. Our previous study demonstrated that HAdV-7 infection activates the NLRP3 inflammasome. Building upon this finding, our current study has identified the L4 100 kDa protein encoded by HAdV-7 as the primary viral component responsible for NLRP3 inflammasome activation. By utilizing techniques such as co-immunoprecipitation, we have confirmed that the 100 kDa protein interacts with the NLRP3 protein and facilitates the assembly of the NLRP3 inflammasome by binding specifically to the NACHT and LRR domains of NLRP3. These insights offer a deeper understanding of HAdV-7 pathogenesis and contribute to the development of novel antiviral therapies.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas no Estructurales Virales , Humanos , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/metabolismo , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/inmunología , Adenovirus Humanos/fisiología , Células HEK293 , Inflamasomas/metabolismo , Inflamasomas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Unión Proteica , Proteínas Virales/metabolismo , Proteínas Virales/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas no Estructurales Virales/metabolismo
9.
Tumour Virus Res ; 17: 200277, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38428735

RESUMEN

Human Adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous sub-types that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating cellular pathways such as PI3K-Akt-mTOR, Ras, the immune response and further HAdV replication stages than previously anticipated. In this review, we aim to explore the structure, molecular mechanisms, and biological functions of E4orf1, shedding light on its potentially multifaceted roles during HAdV infection, including metabolic diseases and oncogenesis. Furthermore, we discuss the role of functional E4orf1 in biotechnological applications such as Adenovirus (AdV) vaccine vectors and oncolytic AdV. By dissecting the intricate relationships between HAdV types and E4orf1 proteins, this review provides valuable insights into viral pathogenesis and points to promising areas of future research.


Asunto(s)
Proteínas E4 de Adenovirus , Adenovirus Humanos , Carcinogénesis , Replicación Viral , Humanos , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Carcinogénesis/inmunología , Carcinogénesis/genética , Proteínas E4 de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/genética , Infecciones por Adenovirus Humanos/inmunología , Infecciones por Adenovirus Humanos/virología , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/virología , Animales , Vectores Genéticos/inmunología , Vectores Genéticos/genética , Viroterapia Oncolítica/métodos
10.
Intervirology ; 67(1): 19-39, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38452738

RESUMEN

BACKGROUND: Human adenoviruses (HAdVs) are extensively used as vectors for vaccines development and cancer therapy. People who already have antibodies against HAdVs, on the other hand, would have an impact on the preventative or therapeutic effect. This review focuses primarily on the prevalence of pre-existing antibodies against HAdVs in distinct geographical populations. SUMMARY: After screening, 64 studies from 31 countries between 1962 and 2021 were selected, totaling 39,427 samples. The total prevalence of preexisting antibodies to HAdVs varied by country or location, ranging from 2.00 to 95.70%. Southeast Asia had the highest prevalence (54.57%) while Europe had the lowest (18.17%). The prevalence in practically all developing nations was higher than in developed nations. Adults have a greater frequency than children and newborns in most nations. The primary HAdV antibody types varied by country. Adults in China, the USA, the United Kingdom, and Belgium had the lowest prevalence of preexisting antibodies against HAdV55, HAdV37, HAdV8, and HAdV36, respectively. Children in the USA, China, the United Kingdom, and Japan had the lowest rates of HAdV48, HAdV11, HAdV8, and HAdV40. The frequency of antibodies differed significantly between military and civilian groups. KEY MESSAGES: Preexisting antibodies against various types of HAdVs differed greatly throughout worldwide populations. Future development of HAdV-vector vaccines and medicines should focus on preexisting antibodies in target groups rather than a "one-size-fits-all" strategy. It might be advantageous in selecting HAdV vectors for studying the prevalence of preexisting antibodies against HAdVs in different locations and people throughout the world.


Asunto(s)
Infecciones por Adenovirus Humanos , Adenovirus Humanos , Anticuerpos Antivirales , Humanos , Adenovirus Humanos/inmunología , Anticuerpos Antivirales/sangre , Infecciones por Adenovirus Humanos/epidemiología , Infecciones por Adenovirus Humanos/inmunología , Prevalencia , Salud Global , Niño , Adulto , Estudios Seroepidemiológicos
11.
ACS Sens ; 9(4): 1831-1841, 2024 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-38489767

RESUMEN

Detection of pathogenic viruses for point-of-care applications has attracted great attention since the COVID-19 pandemic. Current virus diagnostic tools are laborious and expensive, while requiring medically trained staff. Although user-friendly and cost-effective biosensors are utilized for virus detection, many of them rely on recognition elements that suffer major drawbacks. Herein, computationally designed epitope-imprinted polymers (eIPs) are conjugated with a portable piezoelectric sensing platform to establish a sensitive and robust biosensor for the human pathogenic adenovirus (HAdV). The template epitope is selected from the knob part of the HAdV capsid, ensuring surface accessibility. Computational simulations are performed to evaluate the conformational stability of the selected epitope. Further, molecular dynamics simulations are executed to investigate the interactions between the epitope and the different functional monomers for the smart design of eIPs. The HAdV epitope is imprinted via the solid-phase synthesis method to produce eIPs using in silico-selected ingredients. The synthetic receptors show a remarkable detection sensitivity (LOD: 102 pfu mL-1) and affinity (dissociation constant (Kd): 6.48 × 10-12 M) for HAdV. Moreover, the computational eIPs lead to around twofold improved binding behavior than the eIPs synthesized with a well-established conventional recipe. The proposed computational strategy holds enormous potential for the intelligent design of ultrasensitive imprinted polymer binders.


Asunto(s)
Adenovirus Humanos , Epítopos , Humanos , Adenovirus Humanos/inmunología , Adenovirus Humanos/química , Epítopos/inmunología , Epítopos/química , Técnicas Biosensibles/métodos , Polímeros/química , Simulación de Dinámica Molecular , Polímeros Impresos Molecularmente/química , Impresión Molecular/métodos , Límite de Detección , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/química
12.
J Virol ; 96(6): e0185021, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35080426

RESUMEN

Intramuscular delivery of human adenovirus (HAdV)-based vaccines leads to rapid recruitment of neutrophils, which then release antimicrobial peptides/proteins (AMPs). How these AMPs influence vaccine efficacy over the subsequent 24 h is poorly understood. In this study, we asked if human neutrophil protein 1 (HNP-1), an α-defensin that influences direct and indirect innate immune responses to a range of pathogens, impacts the response of human phagocytes to three HAdV species/types (HAdV-C5, -D26, -B35). We show that HNP-1 binds to the capsids and redirects HAdV-C5, -D26, and -B35 to Toll-like receptor 4 (TLR4), which leads to internalization, an NLRP3-mediated inflammasome response, and interleukin 1 beta (IL-1ß) release. Surprisingly, IL-1ß release was not associated with notable disruption of plasma membrane integrity. These data further our understanding of HAdV vaccine immunogenicity and may provide pathways to extend the efficacy. IMPORTANCE This study examines the interactions between danger-associated molecular patterns and human adenoviruses, and their impact on vaccines. HAdVs and HNP-1 can interact, and these interactions will modify the response of antigen-presenting cells, which will influence vaccine efficacy.


Asunto(s)
Infecciones por Adenoviridae , Vacunas contra el Adenovirus , Adenovirus Humanos , Fagocitos , Receptor Toll-Like 4 , alfa-Defensinas , Infecciones por Adenoviridae/inmunología , Vacunas contra el Adenovirus/inmunología , Adenovirus Humanos/inmunología , Humanos , Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fagocitos/citología , Fagocitos/metabolismo , Receptor Toll-Like 4/metabolismo , alfa-Defensinas/inmunología
13.
J Immunol Res ; 2021: 2958394, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34926703

RESUMEN

Adenovirus (Ad) is a major causal agent of acute respiratory infections. However, they are a powerful delivery system for gene therapy and vaccines. Some Ad serotypes antagonize the immune system leading to meningitis, conjunctivitis, gastroenteritis, and/or acute hemorrhagic cystitis. Studies have shown that the release of small, membrane-derived extracellular vesicles (EVs) may offer a mechanism by which viruses can enter cells via receptor-independent entry and how they influence disease pathogenesis and/or host protection considering their existence in almost all bodily fluids. We proposed that Ad3 could alter EV biogenesis, composition, and trafficking and may stimulate various immune responses in vitro. In the present study, we evaluated the impact of in vitro infection with Ad3 vector on EV biogenesis and composition in the human adenocarcinoma lung epithelial cell line A549. Cells were infected in an exosome-free media at different multiplicity of infections (MOIs) and time points. The cell viability was determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) and fluorometric calcein-AM. EVs were isolated via ultracentrifugation. Isolated EV proteins were quantified and evaluated via nanoparticle tracking, transmission electron microscopy, sodium dodecyl sulfate-polyacrylamide gel electrophoresis, and immunoblotting assays. The cell viability significantly decreased with an increase in MOI and incubation time. A significant increase in particle mean sizes, concentrations, and total EV protein content was detected at higher MOIs when compared to uninfected cells (control group). A549 cell-derived EVs revealed the presence of TSG101, tetraspanins CD9 and CD63, and heat shock proteins 70 and 100 with significantly elevated levels of Rab5, 7, and 35 at higher MOIs (300, 750, and 1500) when compared to the controls. Our findings suggested Ad3 could modulate EV biogenesis, composition, and trafficking which could impact infection pathogenesis and disease progression. This study might suggest EVs could be diagnostic and therapeutic advancement to Ad infections and other related viral infections. However, further investigation is warranted to explore the underlying mechanism(s).


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Vesículas Extracelulares/inmunología , Pulmón/patología , Neumonía Viral/inmunología , Células A549 , Infecciones por Adenovirus Humanos/patología , Infecciones por Adenovirus Humanos/virología , Adenovirus Humanos/genética , Supervivencia Celular/inmunología , Vesículas Extracelulares/metabolismo , Humanos , Pulmón/citología , Pulmón/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , Serogrupo
14.
Front Immunol ; 12: 729837, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34603305

RESUMEN

We have developed a dual-antigen COVID-19 vaccine incorporating genes for a modified SARS-CoV-2 spike protein (S-Fusion) and the viral nucleocapsid (N) protein with an Enhanced T-cell Stimulation Domain (N-ETSD) to increase the potential for MHC class II responses. The vaccine antigens are delivered by a human adenovirus serotype 5 platform, hAd5 [E1-, E2b-, E3-], previously demonstrated to be effective in the presence of Ad immunity. Vaccination of rhesus macaques with the hAd5 S-Fusion + N-ETSD vaccine by subcutaneous prime injection followed by two oral boosts elicited neutralizing anti-S IgG and T helper cell 1-biased T-cell responses to both S and N that protected the upper and lower respiratory tracts from high titer (1 x 106 TCID50) SARS-CoV-2 challenge. Notably, viral replication was inhibited within 24 hours of challenge in both lung and nasal passages, becoming undetectable within 7 days post-challenge.


Asunto(s)
Vacunas contra la COVID-19/inmunología , COVID-19/prevención & control , Proteínas de la Nucleocápside de Coronavirus/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Adenovirus Humanos/metabolismo , Administración Oral , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Vacunas contra la COVID-19/administración & dosificación , Citocinas/sangre , Inmunización Secundaria/métodos , Inmunoglobulina G/sangre , Pulmón/virología , Macaca mulatta , Nariz/virología , Fosfoproteínas/inmunología , Dominios Proteicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Vacunación , Replicación Viral/inmunología
15.
Viruses ; 13(9)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34578281

RESUMEN

Following repeat exposure to many human adenoviruses (HAdVs), most adults harbour long-lived B- and T-cell responses. Combined, this response typically protects us for years from re-infection by the same HAdV type. In spite of these immune responses, some HAdV types are associated with persistent infections that constitute a life-threatening risk when an individual's T-cell response is compromised. By contrast, patients with B-cell deficiencies do not appear to be at a greater risk of HAdV disease. This dichotomy begs the question of the secondary role of anti-HAdV antibodies during host defence. In this study, we explored IgG-complexed (IC)-HAdV5 and primary human plasmacytoid dendritic cell (pDC) interactions. We found that IC-HAdV5 are efficiently internalized in pDCs, stimulate their activation through TLR9 signalling, and cause apoptosis. These data may help reconcile the enigma of robust immune response to HAdVs, while concurrently allowing persistence.


Asunto(s)
Adenovirus Humanos/inmunología , Apoptosis/inmunología , Células Dendríticas/patología , Células Dendríticas/virología , Inmunidad Innata , Inmunoglobulina G/inmunología , Presentación de Antígeno , Células Cultivadas , Células Dendríticas/inmunología , Humanos , Transducción de Señal
16.
J Virol ; 95(22): e0038721, 2021 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-34469243

RESUMEN

Preexisting immune responses toward adenoviral vectors limit the use of a vector based on particular serotypes and its clinical applicability for gene therapy and/or vaccination. Therefore, there is a significant interest in vectorizing novel adenoviral types that have low seroprevalence in the human population. Here, we describe the discovery and vectorization of a chimeric human adenovirus, which we call HAdV-20-42-42. Full-genome sequencing revealed that this virus is closely related to human serotype 42, except for the penton base, which is derived from serotype 20. The HAdV-20-42-42 vector could be propagated stably to high titers on existing E1-complementing packaging cell lines. Receptor-binding studies revealed that the vector utilized both CAR and CD46 as receptors for cell entry. Furthermore, the HAdV-20-42-42 vector was potent in transducing human and murine cardiovascular cells and tissues, irrespective of the presence of blood coagulation factor X. In vivo characterizations demonstrate that when delivered intravenously (i.v.) in mice, HAdV-20-42-42 mainly targeted the lungs, liver, and spleen and triggered robust inflammatory immune responses. Finally, we demonstrate that potent T-cell responses against vector-delivered antigens could be induced upon intramuscular vaccination in mice. In summary, from the data obtained we conclude that HAdV-20-42-42 provides a valuable addition to the portfolio of adenoviral vectors available to develop efficacious products in the fields of gene therapy and vaccination. IMPORTANCE Adenoviral vectors are under investigation for a broad range of therapeutic indications in diverse fields, such as oncology and gene therapy, as well as for vaccination both for human and veterinary use. A wealth of data shows that preexisting immune responses may limit the use of a vector. Particularly in the current climate of global pandemic, there is a need to expand the toolbox with novel adenoviral vectors for vaccine development. Our data demonstrate that we have successfully vectorized a novel adenovirus type candidate with low seroprevalence. The cell transduction data and antigen-specific immune responses induced in vivo demonstrate that this vector is highly promising for the development of gene therapy and vaccine products.


Asunto(s)
Adenovirus Humanos , Terapia Genética/métodos , Vectores Genéticos , Desarrollo de Vacunas/métodos , Células A549 , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Animales , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Células HEK293 , Humanos , Masculino , Ratones , Estudios Seroepidemiológicos
17.
Emerg Microbes Infect ; 10(1): 1947-1959, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34520320

RESUMEN

Human adenovirus types 4 (HAdV4) and 7 (HAdV7) often lead to severe respiratory diseases and occur epidemically in children, adults, immune deficiency patients, and other groups, leading to mild or severe symptoms and even death. However, no licensed adenovirus vaccine has been approved in the market for general use. E3 genes of adenovirus are generally considered nonessential for virulence and replication; however, a few studies have demonstrated that the products of these genes are also functional. In this study, most of the E3 genes were deleted, and two E3-deleted recombinant adenoviruses (ΔE3-rAdVs) were constructed as components of the vaccine. After E3 deletion, the replication efficiencies and cytopathogenicity of ΔE3-rAdVs were reduced, indicating that ΔE3-rAdVs were attenuated after E3 genes deletion. Furthermore, single immunization with live-attenuated bivalent vaccine candidate protects mice against challenge with wild-type human adenovirus types 4 and 7, respectively. Vaccinated mice demonstrated remarkably decreased viral loads in the lungs and less lung pathology compared to the control animals. Taken together, our study confirms the possibility of the two live-attenuated viruses as a vaccine for clinic use and illustrates a novel strategy for the construction of an adenovirus vaccine.


Asunto(s)
Proteínas E3 de Adenovirus/genética , Infecciones por Adenovirus Humanos/prevención & control , Vacunas contra el Adenovirus/inmunología , Adenovirus Humanos/inmunología , Vacunas Atenuadas/inmunología , Células A549 , Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/clasificación , Animales , Línea Celular , Femenino , Eliminación de Gen , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Carga Viral
18.
Sci Immunol ; 6(63): eabe2942, 2021 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-34533978

RESUMEN

Human adenoviruses (HAdVs) are a major cause for disease in children, in particular after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Currently, effective therapies for HAdV infections in immunocompromised hosts are lacking. To decipher immune recognition of HAdV infection and determine new targets for immune-mediated control, we used an HAdV infection 3D organoid system, based on primary human intestinal epithelial cells. HLA-F, the functional ligand for the activating NK cell receptor KIR3DS1, was strongly up-regulated and enabled enhanced killing of HAdV5-infected cells in organoids by KIR3DS1+ NK cells. In contrast, HLA-A and HLA-B were significantly down-regulated in HAdV5-infected organoids in response to adenoviral E3/glycoprotein19K, consistent with evasion from CD8+ T cells. Immunogenetic analyses in a pediatric allo-HSCT cohort showed a reduced risk to develop severe HAdV disease and faster clearance of HAdV viremia in children receiving KIR3DS1+/HLA-Bw4+ donor cells compared with children receiving non­KIR3DS1+/HLA-Bw4+ cells. These findings identify the KIR3DS1/HLA-F axis as a new target for immunotherapeutic strategies against severe HAdV disease.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Células Asesinas Naturales/inmunología , Receptores KIR3DS1/inmunología , Células A549 , Adenovirus Humanos/inmunología , Células HEK293 , Humanos
19.
Viruses ; 13(7)2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-34202573

RESUMEN

Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.


Asunto(s)
Adenovirus Humanos/inmunología , Adenovirus Humanos/fisiología , Cápside/metabolismo , Vectores Genéticos , Vacunas Virales/inmunología , Internalización del Virus , Inmunidad Adaptativa , Adenovirus Humanos/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , COVID-19/inmunología , Cápside/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Ensayos Clínicos como Asunto , Humanos , Inmunidad Innata , Ratones , SARS-CoV-2/inmunología
20.
Front Immunol ; 12: 700438, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322126

RESUMEN

Reactivation of Human Cytomegalovirus (HCMV) and Human Adenovirus (HAdV) in immunocompromised patients following stem cell transplantation (SCT) or solid organ transplantation (SOT) is associated with high morbidity and mortality. The adoptive transfer of virus-specific CD8+ and CD4+ T cells has been shown to re-establish the antiviral T-cell response and improve clinical outcome. The viral load in immunocompromised patients can efficiently be reduced solely by the infusion of virus-specific CD4+ T cells. The identification of CD4+ T-cell epitopes has mainly focused on a limited number of viral proteins that were characterized as immunodominant. Here, we used in silico prediction to determine promiscuous CD4+ T-cell epitopes from the entire proteomes of HCMV and HAdV. Immunogenicity testing with enzyme-linked immuno spot (ELISpot) assays and intracellular cytokine staining (ICS) revealed numerous novel CD4+ T-cell epitopes derived from a broad spectrum of viral antigens. We identified 17 novel HCMV-derived and seven novel HAdV-derived CD4+ T-cell epitopes that were recognized by > 50% of the assessed peripheral blood mononuclear cell (PBMC) samples. The newly identified epitopes were pooled with previously published, retested epitopes to stimulate virus-specific memory T cells in PBMCs from numerous randomly selected blood donors. Our peptide pools induced strong IFNγ secretion in 46 out of 48 (HCMV) and 31 out of 31 (HAdV) PBMC cultures. In conclusion, we applied an efficient method to screen large viral proteomes for promiscuous CD4+ T-cell epitopes to improve the detection and isolation of virus-specific T cells in a clinical setting.


Asunto(s)
Adenovirus Humanos/inmunología , Linfocitos T CD4-Positivos/inmunología , Citomegalovirus/inmunología , Mapeo Epitopo/métodos , Epítopos de Linfocito T/inmunología , Antígenos Virales/inmunología , Humanos , Células T de Memoria/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA