RESUMO
Colorectal cancer (CRC) is a prevalent malignant tumor of the gastrointestinal system, with the third and second highest incidence and mortality rates globally in 2020, respectively. Immunotherapy has developed rapidly in recent years. Natural killer (NK) cells have received increasing attention in the field of tumor immunotherapy due to their recognition and killing tumor cells without the limitations of major histocompatibility complexes. However, constraints within the tumor microenvironment that impede the infiltration and proliferation of NK cells result in poor efficacy of NK cell therapy for solid tumors. Oncolytic viral therapy is an immunogenic treatment with the potential to enhance anti-tumour immune responses and promote immune cell infiltration. In this study, we synergistically combine NK cells with an oncolytic adenovirus carrying Decorin (rAd.DCN) for the treatment of colorectal cancer (CRC) in a xenograft mouse model. By using Flow cytometry, real-time quantitative PCR and Calcein-AM release assay, we found that rAd.DCN could effectively promote proliferation, activation and degranulation of NK cells, up-regulate expression and secretion of NK cell killing activity-related factors, and enhance their killing activity. The efficacy is better than that of the blank control oncolytic virus rAd.Null. Combined treatment significantly inhibited tumor growth, increased the number of NK cells in peripheral blood, promoted the killing function of NK cells, and increased the expression levels of perforin and IFN-γ. At the same time, more NK cells were recruited to infiltrate tumor tissue. Our study established the feasibility of combination NK cells and oncolytic adenovirus application, thus expanding the scope of potentially curative treatments for NK cells in CRC.
Assuntos
Adenoviridae , Neoplasias Colorretais , Decorina , Células Matadoras Naturais , Terapia Viral Oncolítica , Vírus Oncolíticos , Células Matadoras Naturais/imunologia , Animais , Neoplasias Colorretais/terapia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Decorina/genética , Decorina/metabolismo , Adenoviridae/genética , Humanos , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/genética , Camundongos , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Microambiente Tumoral/imunologia , Ativação Linfocitária , Proliferação de Células , FemininoRESUMO
AIM: The human adenovirus (HAdV) is beginning to spread rapidly in children through human, surface and animal vectors. Around 12,000 cases were recognised in 2022 in West Bengal and a shocking number of cases arose throughout India and in other under-developed areas. This is going to be a big threat to public health since no vaccine, awareness or protocol policies were introduced. Early detection, immediate isolation and proper policy developments are the key factors in overcoming the situation. Therefore, we performed this rapid review and discussed probable mitigation strategies, updated research on vaccine development, and treatment strategies to control the outbreaks of mutated HAdV. DESIGN: This is a narrative review of publicly available information. METHODS: Here, we extracted updated information and data using the terms HAdV outbreaks, mutations, species, risks and prevention from Google Scholar and PubMed. We considered relevant articles that have discussed prevention strategies, ongoing research, and antiviral drugs for managing HAdV outbreaks. RESULTS: Early detection from throat swabs, isolation and symptomatic treatments are required to minimise viral infections. A massive test needs to be performed to find the affected people. The cases should be immediately isolated. It is recommended to treat high-touch surfaces with heat- or bleach-containing cleaners to prevent the spread of infection. Oxygen support and many broad-spectrum antivirals have been used to treat HAdV. Several studies showed antibody neutralisation and interactions between the natural killer cell receptor KIR3DS1 and HLA-F in infected cells, indicating possible therapeutic options in the future. HAdV-4 and HAdV-7 vaccines have been limitedly approved for administration to military personnel. CONCLUSION: Isolation, certain safety measures, broad-spectrum antiviral drugs and further research on new vaccines could be useful to prevent this virus from producing a worldwide pandemic. Also, the authorities should ensure the proper therapeutic interventions and nursing care facilities for the infected children. PATIENT OR PUBLIC CONTRIBUTION: Patient or public contribution was not relevant to our work.
Assuntos
Surtos de Doenças , Humanos , Índia/epidemiologia , Surtos de Doenças/prevenção & controle , Medição de Risco , Infecções por Adenovirus Humanos/epidemiologia , Infecções por Adenovirus Humanos/prevenção & controle , Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , MutaçãoRESUMO
We developed an in vivo HSC gene therapy approach that consists of HSC mobilization and intravenous injection of HSC-tropic HDAd vectors. To achieve therapeutically relevant numbers of corrected cells, we incorporated in vivo expansion of transduced cells. We used an HDAd vector for a multiplex adenine base editing approach to (1) remove the region within CD33 that is recognized by gemtuzumab ozogamicin (GO) (Mylotarg), and (2) create therapeutic edits within the HBG1/2 promoters to reactivate γ-globin/HbF. In vitro studies with HDAd-transduced human CD34+ cells showed editing of both targeted sites and a 2- to 3-fold GO-mediated expansion of edited erythroid/myeloid progenitors. After erythroid in vitro differentiation, up to 40% of erythrocytes were HbF positive. For in vivo studies, mice were transplanted with human CD34+ cells. After engraftment, HSCs were mobilized with G-CSF/AMD3100 followed by an intravenous HDAd injection and GO-mediated in vivo selection. Two months later, editing in human cells within the bone marrow was significantly higher in GO-treated mice. The percentage of HbF+ human erythroid cells was 2.5-fold greater compared with untreated mice. These data indicate that in vivo GO selection can increase edited erythroid cells.
RESUMO
BACKGROUND: Human adenoviruses (HAdVs) frequently cause common respiratory or gastrointestinal infections among children, adults, individuals with immune deficiencies, and other vulnerable populations with varying degree of symptoms, ranging from mild to server, and in some cases, even fatalities. Despite the significant clinical impact of HAdVs, there is currently no approved vaccine available. METHODS: This study explores the potential of the adenovirus type 5 fiber knob (Ad5-FK) to stimulate the production of Ad-specific neutralizing antibodies and T-cell responses in mice. Based on structure predictions, we first expressed Ad5-FK in E. coli and confirmed the assembly of FK into its trimeric form. After testing the binding capability of the trimeric FK to susceptible cells, the immunogenicity of the protein in combination with the c-di-AMP adjuvant was assessed in BALB/c mice. RESULTS: The purified Ad5-FK exhibited self-trimerization and maintained correct conformation akin to the authentic FK structure. This facilitated effective binding to susceptible HEK293 cells. Notably, the protein demonstrated significant inhibition of HEK293 cells infection by rAd5-GFP. Immunization of BALB/c mice with Ad5-FK, or Ad5-FK mixed with c-di-AMP yielded FK-specific antibodies with potent neutralization capacity. Significantly, Ad5-FK was found to elicit a vigorous CD4+ T-cell response in the immunized mice. CONCLUSION: Our findings underscore the efficacy of FK-based vaccine in eliciting anti-Ad humoral immune response and CD4 T-cell immune reactions essential for protection against viral infections.
Assuntos
Adenovírus Humanos , Anticorpos Neutralizantes , Anticorpos Antivirais , Camundongos Endogâmicos BALB C , Animais , Anticorpos Neutralizantes/imunologia , Humanos , Anticorpos Antivirais/imunologia , Camundongos , Células HEK293 , Adenovírus Humanos/imunologia , Adenovírus Humanos/genética , Linfócitos T/imunologia , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Feminino , Vacinação , Vacinas contra Adenovirus/imunologia , Vacinas contra Adenovirus/administração & dosagem , Infecções por Adenoviridae/imunologia , Infecções por Adenoviridae/prevenção & controle , Infecções por Adenovirus Humanos/imunologia , Infecções por Adenovirus Humanos/prevenção & controle , Infecções por Adenovirus Humanos/virologiaRESUMO
BACKGROUND: Severe adenovirus pneumonia in children has a high mortality rate, but research on risk prediction models is lacking. Such models are essential as they allow individualized predictions and assess whether children will likely progress to severe disease. METHODS: A retrospective analysis was performed on children with adenovirus pneumonia who were hospitalized at the Children's Hospital of Nanjing Medical University from January 2017 to March 2024. The patients were grouped according to clinical factors, and the groups were compared using Ridge regression and multiple logistic regression to identify risk factors associated with severe adenovirus pneumonia. A prediction model was constructed, and its value in clinical application was evaluated. RESULTS: 699 patients were included in the study, with 284 in the severe group and 415 in the general group. Through the screening of 44 variables, the final risk factors for severe adenovirus pneumonia in children as the levels of neutrophils (OR = 1.086, 95% CI: 1.054â1.119, P < 0.001), D-dimer (OR = 1.005, 95% CI: 1.003â1.007, P < 0.001), fibrinogen degradation products (OR = 1.341, 95% CI: 1.034â1.738, P = 0.027), B cells (OR = 1.076, 95%CI: 1.046â1.107, P < 0.001), and lactate dehydrogenase (OR = 1.008, 95% CI: 1.005â1.011, P < 0.001). The value of the area under the receiver operating characteristic curve was 0.974, the 95% CI was 0.963-0.985, and the P-value of the Hosmer-Lemeshow test was 0.547 (P > 0.05), indicating that the model had strong predictive power. CONCLUSION: In this study, the clinical variables of children with adenovirus pneumonia were retrospectively analyzed to identify risk factors for severe disease. A prediction model for severe disease was constructed and evaluated, showing good application value.
Assuntos
Pneumonia Viral , Humanos , Estudos Retrospectivos , Masculino , Feminino , Fatores de Risco , Pré-Escolar , Lactente , Pneumonia Viral/epidemiologia , Pneumonia Viral/diagnóstico , Pneumonia Viral/terapia , Medição de Risco , Índice de Gravidade de Doença , Infecções por Adenovirus Humanos/diagnóstico , Infecções por Adenovirus Humanos/epidemiologia , Criança , China/epidemiologia , Modelos LogísticosRESUMO
The liver plays a crucrial role in detoxification, metabolism, and nutrient storage. Because liver cancer ranks among the top three leading causes of death globally, there is an urgent need for developing treatment strategies for liver cancer. Although traditional approaches such as radiation, chemotherapy, surgical removal, and transplantation are widely practiced, the number of patients with liver cancer continues to increase rapidly each year. Some novel therapeutics for liver cancer have been studied for many years. In the past decade, oncolytic therapy has emerged, in which viruses selectively infect and destroy cancer cells while sparing normal cells. However, oncolytic virotherapy for liver cancer remains relatively obscure due to the aggressive nature of the disease and the limited effectiveness of treatment. To keep pace with the latest developments in oncolytic tumor therapy for liver cancer, this review summarizes basic science studies and clinical trials conducted within 5 years, focusing on the efficacy and safety profiles of the five most commonly used oncolytic viruses: herpes simplex virus, adenovirus, influenza virus, vaccinia virus, and coxsackievirus.
RESUMO
Systemic delivery of oncolytic adenovirus (oAd) for cancer gene therapy must overcome several limitations such as rapid clearance from the blood, nonspecific accumulation in the liver, and insufficient delivery to the tumor tissues. In the present report, a tumor microenvironment-triggered artificial lipid envelope composed of a pH-responsive sulfamethazine-based polymer (PUSSM)-conjugated phospholipid (DOPE-HZ-PUSSM) and another lipid decorated with epidermal growth factor receptor (EGFR) targeting peptide (GE11) (GE11-DOPE) was utilized to encapsulate replication-incompetent Ad (dAd) or oAd coexpressing short-hairpin RNA (shRNA) against Wnt5 (shWnt5) and decorin (dAd/LP-GE-PS or oAd/LP-GE-PS, respectively). In vitro studies demonstrated that dAd/LP-GE-PS transduced breast cancer cells in a pH-responsive and EGFR-specific manner, showing a higher level of transduction than naked Ad under a mildly acidic pH of 6.0 in EGFR-positive cell lines. In vivo biodistribution analyses revealed that systemic administration of oAd/LP-GE-PS leads to a significantly higher level of intratumoral virion accumulation compared to naked oAd, oAd encapsulated in a liposome without PUSSM or EGFR targeting peptide moiety (oAd/LP), or oAd encapsulated in a liposome with EGFR targeting peptide alone (oAd/LP-GE) in an EGFR overexpressing MDA-MB-468 breast tumor xenograft model, showing that both pH sensitivity and EGFR targeting ability were integral to effective systemic delivery of oAd. Further, systemic administration of all liposomal oAd formulations (oAd/LP, oAd/LP-GE, and oAd/LP-GE-PS) showed significantly attenuated hepatic accumulation of the virus compared to naked oAd. Collectively, our findings demonstrated that pH-sensitive and EGFR-targeted liposomal systemic delivery of oAd can be a promising strategy to address the conventional limitations of oAd to effectively treat EGFR-positive cancer in a safe manner.
Assuntos
Adenoviridae , Receptores ErbB , Terapia Genética , Terapia Viral Oncolítica , Vírus Oncolíticos , Humanos , Animais , Vírus Oncolíticos/genética , Camundongos , Adenoviridae/genética , Terapia Viral Oncolítica/métodos , Terapia Genética/métodos , Receptores ErbB/metabolismo , Feminino , Concentração de Íons de Hidrogênio , Linhagem Celular Tumoral , Camundongos Nus , Camundongos Endogâmicos BALB C , Fosfatidiletanolaminas/química , Lipossomos/química , Lipídeos/química , Distribuição Tecidual , PeptídeosRESUMO
Genetic modification in vivo could provide direct functions of genes that could potentially contribute to diverse areas of research including genetics, developmental biology, and physiology. It has been reported that genes of interest could be introduced via recombinant adenovirus type 5 (Ad5) in poultry. Successful gene delivery to mammal fetuses in utero promises substantial progress in clinical and developmental biology, but it is limited because of difficulties in injecting specific sites and invasiveness. On the other hand, developing avian embryos are easily accessible by making a window on the eggshell. Therefore, the objective of this study is to determine permissive embryonic stages for gene transfer into specific avian tissue/organs by injection of Ad5 containing the green fluorescent protein (GFP) gene into blood vessels. At 2 d of post-injection, a strong GFP signal was predominantly identified in the heart of chicken embryos injected at Hamilton-Hamburger (HH) 14, 15, 16 and17 stages with the percentages (44%, 53%, 25%, and 14%, respectively) of GFP positive embryos. In quail embryos, the injection at the HH 15 resulted in heart-specific expression of GFP. Western blot analysis revealed that GFP was exclusively expressed in the avian hearts. These results suggest that the GFP gene is specifically delivered to the avian embryonic hearts when Ad5 is injected through the blood vessel at HH 14-17. This adenoviral transduction of genes of interest in avian embryonic hearts can provide new models for understanding functions of genetic factors on embryonic heart development and unravel genetic etiology of congenital heart diseases.
RESUMO
Gizzard erosion and ulceration syndrome (GEUS) is caused by a fowl adenovirus serotype 1 (FAdV-1) and was first reported in laying hens in Japan in 1993. This syndrome has emerged as an epizootic in Morocco since 2014, causing significant economic losses for the poultry industry, but no involvement of a FAdV has been confirmed. Thus, the objective of this work was to assess GEUS cases that occurred in the country and to determine the role of FAdVs in their occurrence. Investigations were based on a retrospective reassessment of tissue sections and paraffin blocks of gizzards and livers from GEUS cases between 2014 and 2021 coupled with a prospective search of cases in 2022. Gizzards and livers were fixed in 10% neutral buffered formalin for histopathologic examinations according to standard methods and stored at -20 C for molecular analysis. After deparaffinizing, 10-µm-thick tissue sections along with fresh organs were subjected to DNA extraction using a commercial kit. A primer pair specific for the Hexon gene of FAdVs was used in conventional PCR; in contrast, for real-time PCR, a primer pair targeting the 52K gene was employed. In total, 24 flock cases with characteristic GEUS were assessed between 2014 and 2022. They were nine broiler cases aged between 11 and 39 days, 11 layer cases with an age between 17 and 29 wk, two cases in meat-type breeders aged 10 and 27 wk, and two flock cases of turkey poults aged 22 and 23 days. In most cases, microscopic lesions were consistent with an ulcerative and lymphoplasmocytic ventriculitis, and pathognomonic viral intranuclear inclusion bodies within degenerate epithelial cells were identified in four broiler flock cases, four layer cases, and one case in breeders and hence were highly suggestive of a FAdV infection. Among these nine cases that were positive at the histopathologic examination, six cases were found to be FAdV-PCR positive; another four cases were negative to histology but FAdV-PCR positive. Furthermore, a sequencing analysis was conducted, providing the initial evidence of the implication of FAdV-1 from species A as the cause of GEUS in Moroccan poultry. Additionally, a phylogenetic analysis was executed to facilitate a comparison between the strains investigated in this study and those identified in diverse geographic regions and across various time periods.
Síndrome de erosión y ulceración de la molleja en parvadas avícolas marroquíes y caracterización molecular de los adenovirus aviares (FAdV). El síndrome de erosión y ulceración de la molleja (GEUS) es causado por un adenovirus del pollo de serotipo 1 (FAdV-1) y se reportó por primera vez en gallinas de postura en Japón en 1993. Este síndrome se ha convertido en una epizootia en Marruecos desde 2014, causando importantes pérdidas económicas a la industria avícola, pero no se ha confirmado la participación de ningún adenovirus del pollo. Por lo tanto, el objetivo de este trabajo fue evaluar los casos de GEUS ocurridos en el país y determinar el papel de adenovirus del pollo en su presentación. Las investigaciones se basaron en una reevaluación retrospectiva de secciones de tejido y bloques de parafina de mollejas e hígados de casos de GEUS entre 2014 y 2021, junto con una investigación prospectiva de casos en el año 2022. Las mollejas y los hígados se fijaron en formalina al 10% amortiguada y neutra para exámenes histopatológicos de acuerdo con métodos estándar y se almacenaron a -20 C para análisis moleculares. Después de la desparafinación, las secciones de tejido de 10 µm de espesor junto con órganos frescos se sometieron a extracción de ADN utilizando un estuche comercial. Para realizar un método de PCR convencional, se utilizó un par de iniciadores específicos para el gene de hexon de los adenovirus del pollo, mientras que, para el método de PCR en tiempo real, se empleó un par de iniciadores dirigidos al gene 52K. En total, se evaluaron 24 casos de parvadas con la presentación característica del síndrome de erosión y ulceración de la molleja entre los años 2014 y 2022. Se trató de nueve casos de pollos de engorde con edades comprendidas entre los 11 y 39 días, 11 casos de ponedoras con una edad de entre 17 y 29 semanas, dos casos en reproductoras pesadas de 10 y 27 semanas, y dos lotes de pavitos de 22 y 23 días. En la mayoría de los casos, las lesiones microscópicas fueron consistentes con una ventriculitis ulcerativa y linfoplasmocítica, y se identificaron cuerpos de inclusión intranucleares virales patognomónicos dentro de células epiteliales degeneradas en cuatro casos de parvadas de pollos de engorde, cuatro casos de ponedoras y un caso en reproductoras y por lo tanto, fueron altamente sugestivos de la infección por adenovirus de pollo. Entre los nueve casos que fueron positivos en el examen histopatológico, se encontró que seis casos fueron positivos para adenovirus de pollo mediante PCR; otros cuatro casos fueron negativos a la histología pero positivos a la presencia de adenovirus del pollo mediante PCR. Además, se realizó un análisis de secuenciación que proporcionó la evidencia inicial del papel de adenovirus del pollo especie A como causante del síndrome de erosión y ulceración de la molleja en la avicultura de Marruecos. Además, se realizó un análisis filogenético para facilitar una comparación entre las cepas investigadas en este estudio y las identificadas en diversas regiones geográficas y en varios períodos de tiempo.
Assuntos
Infecções por Adenoviridae , Galinhas , Moela das Aves , Doenças das Aves Domésticas , Animais , Infecções por Adenoviridae/veterinária , Infecções por Adenoviridae/virologia , Infecções por Adenoviridae/patologia , Infecções por Adenoviridae/epidemiologia , Doenças das Aves Domésticas/virologia , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/epidemiologia , Moela das Aves/patologia , Moela das Aves/virologia , Marrocos/epidemiologia , Adenovirus A das Aves/genética , Adenovirus A das Aves/isolamento & purificação , Filogenia , Estudos Retrospectivos , Úlcera/veterinária , Úlcera/virologia , Úlcera/patologia , Aviadenovirus/isolamento & purificação , Aviadenovirus/genética , Aviadenovirus/classificaçãoRESUMO
BACKGROUND: Non-enveloped viruses, which lack a lipid envelope, display higher resistance to disinfectants, soaps and sanitizers compared to enveloped viruses. The capsids of these viruses are highly stable and symmetric protein shells that resist inactivation by commonly employed virucidal agents. This group of viruses include highly transmissible human pathogens such as Rotavirus, Poliovirus, Foot and Mouth Disease Virus, Norovirus and Adenovirus; thus, devising appropriate strategies for chemical disinfection is essential. RESULTS: In this study, we tested a mild, hypoallergenic combination of a denaturant, alcohol, and organic acid (3.2% citric acid, 1% urea and 70% ethanol, pH4) on two representative non-enveloped viruses - Human Adenovirus 5 (HAdV5) and Feline Calicivirus (FCV)- and evaluated the pathways of capsid neutralization using biophysical methods. The conformational shifts in the capsid upon chemical treatment were studied using Differential Scanning Calorimetry (DSC), while the morphological alterations were visualized concurrently using Transmission Electron Microscopy (TEM). We found that while treatment of purified HAdV5 particles with a formulation resulted in thermal instability and, large scale aggregation; similar treatment of FCV particles resulted in complete collapse of the capsids. Further, while individual components of the formulation caused significant damage to the capsids, a synergistic action of the whole formulation was evident against both non-enveloped viruses tested. CONCLUSIONS: The distinct effects of the chemical treatment on the morphology of HAdV5 and FCV suggests that non-enveloped viruses with icosahedral geometry can follow different morphological pathways to inactivation. Synergistic effect of whole formulation is more effective compared to individual components. Molecular level understanding of inactivation pathways may result in the design and development of effective mass-market formulations for rapid neutralization of non-enveloped viruses.
Assuntos
Adenovírus Humanos , Calicivirus Felino , Capsídeo , Inativação de Vírus , Inativação de Vírus/efeitos dos fármacos , Calicivirus Felino/efeitos dos fármacos , Calicivirus Felino/fisiologia , Adenovírus Humanos/efeitos dos fármacos , Adenovírus Humanos/fisiologia , Adenovírus Humanos/química , Adenovírus Humanos/ultraestrutura , Capsídeo/efeitos dos fármacos , Capsídeo/química , Capsídeo/ultraestrutura , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/química , Desinfetantes/farmacologia , Humanos , Microscopia Eletrônica de Transmissão , Ureia/farmacologia , Ureia/química , Ureia/análogos & derivados , Ácido Cítrico/farmacologia , Ácido Cítrico/química , Etanol/farmacologia , Animais , Varredura Diferencial de CalorimetriaRESUMO
Recombinant oncolytic adenovirus offers a novel and promising cancer treatment approach, but its standalone efficacy remains limited. This study investigates a combination treatment strategy by co-administering recombinant oncolytic Adv-loaded silk hydrogel with a PD-L1 inhibitor for patients with bladder cancer to enhance treatment outcomes. Bladder cancer tissues from mice were collected and subjected to single-cell sequencing, identifying CRB3 as a key gene in malignant cells. Differential expression and functional enrichment analyses were performed, validating CRB3's inhibitory role through in vitro experiments showing suppression of bladder cancer cell proliferation, migration, and invasion. Recombinant oncolytic adenoviruses encoding CRB3 and GM-CSF were constructed and encapsulated in silk hydrogel to enhance drug loading and release efficiency. In vivo experiments demonstrated that the nano-composite hydrogel significantly inhibited tumor growth and increased immune infiltration in tumor tissues. Co-administration of adenovirus silk hydrogel (Adv-CRB3@gel) with a PD-L1 inhibitor significantly enhanced T-cell infiltration and tumor killing. The combination of recombinant oncolytic Adv-loaded nano-composite hydrogel encoding CRB3 and GM-CSF with a PD-L1 inhibitor improves bladder cancer treatment outcomes by effectively recruiting T cells, providing a novel therapeutic strategy.
Assuntos
Adenoviridae , Antígeno B7-H1 , Hidrogéis , Terapia Viral Oncolítica , Vírus Oncolíticos , Seda , Neoplasias da Bexiga Urinária , Neoplasias da Bexiga Urinária/terapia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Camundongos , Adenoviridae/genética , Humanos , Linhagem Celular Tumoral , Hidrogéis/química , Terapia Viral Oncolítica/métodos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Seda/química , Terapia Combinada , Vírus Oncolíticos/genética , Inibidores de Checkpoint Imunológico/farmacologia , Feminino , Proliferação de Células/efeitos dos fármacos , Camundongos Endogâmicos BALB C , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genéticaRESUMO
BACKGROUND: Birds are known to harbour many pathogens, including circovirus, herpesviruses, adenoviruses and Chlamydia psittaci. Some of these pose zoonotic risks, while others, such as beak and feather disease virus (BFDV), have a significant impact on the conservation of endangered bird species. OBJECTIVES: This study was aimed to determine the faecal virome of a group of apparently healthy Monk parakeet using high-throughput sequencing. METHODS: Fresh faecal samples were collected from four Monk parakeets at a pet shop in Melbourne, Australia. Virus enrichment and nucleic acid extraction were performed on the faecal samples, followed by high-throughput sequencing at the Australian Genome Research Facility (AGRF). RESULTS: Utilising an established pipeline for high-throughput sequencing data analysis, this study revealed the presence of three viruses of the families Circoviridae, Parvoviridae and Adenoviridae. Subsequent sequence comparison and phylogenetic analyses further confirmed that the detected viruses belong to the genera Chaphamaparvovirus (unassigned species), Circovirus (species Circovirus parrot) and Siadenovirus (species Siadenovirus viridis). CONCLUSION: Despite non-pathogenicity, the existence of multiple viruses within a bird species underscores the risk of these viruses spreading into the pet trade. Detection and a better understanding of avian viruses are crucial for the establishment of appropriate management and biosecurity measures in the domestic and international bird trade, which ultimately supports the conservation of vulnerable bird species.
Assuntos
Fezes , Periquitos , Animais , Fezes/virologia , Fezes/microbiologia , Periquitos/virologia , Doenças das Aves/virologia , Doenças das Aves/epidemiologia , Doenças das Aves/microbiologia , Austrália , Adenoviridae/isolamento & purificação , Adenoviridae/classificação , Adenoviridae/genética , Parvoviridae/isolamento & purificação , Parvoviridae/genética , Parvoviridae/classificação , Filogenia , Circovirus/genética , Circovirus/isolamento & purificação , Circovirus/classificação , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , Vitória , Circoviridae/isolamento & purificação , Circoviridae/genética , Circoviridae/classificação , Viroma , MetagenômicaRESUMO
In 2022, several cases of acute hepatitis of unknown etiology (AHUE) have been associated with Adeno-associated virus 2 (AAV-2) and the common childhood virus Adenovirus 41 (AdV-41). This outbreak has resulted in serious complications in patients which included 5 % of individuals requiring a liver transplant and 22 deaths. Before these AHUE cases, no previous information had been reported regarding the co-infections and co-occurrence of these two viruses in the human population. The present study utilized WBE tools to investigate the prevalence of AAV-2 and AdV-F (AdV-41 and AdV-40) in wastewater from two different waste-water treatment plants (WWTP) serving the city of Bloomington in Southern Indiana, USA. The concentrations of AAV-2 and AdV-F were quantified using digital PCR in weekly wastewater samples taken over the duration of 18 months. High levels of both viruses were observed in most of the samples where co-detection and correlation in the concentrations for AAV-2 and AdV-F were found to be significant (p < 0.01) throughout duration of the study. In addition, significant seasonal changes were observed in the viral concentrations of both viruses (P < 0.01), but these seasonal variations were different between WWTPs (p < 0.01). However, these seasonal variations in viral concentrations were similar for both viruses. The sequences of AdV-F and AAV were obtained from the wastewater samples and confirmed the detection of AAV-2, AdV-41, and AdV-40 in the samples analyzed. Even though our study was done after the 2022 outbreak of AHUE, our results demonstrated the persistence of infections with both viruses in the population. It also highlights the ongoing spread of both viruses in the population and the importance of WBE in surveillance of these viruses.
RESUMO
Luciferase (luc) bioluminescence (BL) is the most used light-emitting protein that has been engineered to be expressed in multiple cancer cell lines, allowing for the detection of tumor nodules in vivo as it can penetrate most tissues. The goal of this study was to develop an oncolytic adenovirus (OAd)-resistant human triple-negative breast cancer (TNBC) that could express luciferase. Thus, when combining an OAd with chemotherapies or targeted therapies, we would be able to monitor the ability of these compounds to enhance OAd antitumor efficacy using BL in real time. The TNBC cell line HCC1937 was stably transfected with the plasmid pGL4.50[luc2/CMV/Hygro] (HCC1937/luc2). Once established, HCC1937/luc2 was orthotopically implanted in the 4th mammary gland fat pad of NSG (non-obese diabetic severe combined immunodeficiency disease gamma) female mice. Bioluminescence imaging (BLI) revealed that the HCC1937/luc2 cell line developed orthotopic breast tumor and lung metastasis over time. However, the integration of luc plasmid modified the HCC1937 phenotype, making HCC1937/luc2 more sensitive to OAdmCherry compared to the parental cell line and blunting the interferon (IFN) antiviral response. Testing two additional luc cell lines revealed that this was not a universal response; however, proper controls would need to be evaluated, as the integration of luciferase could affect the cells' response to different treatments.
Assuntos
Luciferases , Neoplasias de Mama Triplo Negativas , Animais , Humanos , Feminino , Camundongos , Linhagem Celular Tumoral , Luciferases/metabolismo , Luciferases/genética , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Adenoviridae/genética , Terapia Viral Oncolítica/métodos , Medições Luminescentes/métodos , Camundongos SCID , Camundongos Endogâmicos NOD , Vírus Oncolíticos/genéticaRESUMO
BACKGROUND: Rabies is a zoonotic viral encephalitis that is endemic in many countries and confers a high mortality. Licensed vaccines require several doses to ensure efficacy. To investigate a logistically favorable approach, we assessed the safety and immunogenicity of ChAd155-RG, a novel investigational rabies vaccine using a replication-defective chimpanzee adenovirus vector. METHODS: We conducted a first-in-human, phase 1, randomized, double-blind, dose-escalation trial comparing ChAd155-RG with a licensed inactivated vaccine (RabAvert) in healthy adults. Participants received either RabAvert at standard dosing or ChAd155-RG at a low dose for one immunization or a high dose for one or two immunizations. To assess safety, we evaluated reactogenicity, unsolicited adverse events, and thrombotic events. To measure immunogenicity, we measured rabies viral neutralizing antibody (VNA) titers and anti-ChAd155 neutralizing antibodies. RESULTS: Mild to moderate systemic reactogenicity and transient lymphopenia and neutropenia were more common among recipients of ChAd155-RG compared with those who received RabAvert. No thrombotic events or serious adverse events were reported. Only the groups receiving RabAvert or two doses of high-dose ChAd155-RG achieved 100 % seroconversion, and seroprotection was most durable in the RabAvert group. Most participants had preexisting anti-vector antibodies, which were boosted by ChAd155-RG. Baseline and post-vaccination anti-vector antibody titers were negatively associated with post-vaccination rabies VNA titers. CONCLUSIONS: In this phase 1 clinical trial, a novel rabies vaccine using a simian adenovirus vector was safe and tolerable, but generated lower, less durable rabies VNA titers than a standard inactivated rabies virus vaccine, which may be due to preexisting, anti-vector immunity.
RESUMO
Orthoflaviviruses are single-stranded RNA viruses characterized by highly efficient self-amplification of RNA in host cells, which makes them attractive vehicles for vaccines. Numerous preclinical and clinical studies have demonstrated the efficacy and safety of orthoflavivirus replicon vectors for vaccine development. In this study, we constructed Tembusu virus (TMUV) replicon-based single-round infectious particles (SRIPs) as vaccine development platform. To evaluate the potential of TMUV SRIPs as vaccines, we generated SRIPs that express the heterologous Fowl adenovirus 4 (FAdV-4) fiber2 protein and fiber2 head domain, named TMUVRP-fiber2 and TMUVRP-fiber2H, respectively. To assess the immunogenicity of the TMUV SRIPs, SPF chicks were intramuscularly inoculated twice. Our results showed that the TMUVRP-fiber2 vaccines elicited high levels of neutralizing antibodies. Challenge experiments showed that TMUVRP-fiber2 provided full protection against virulent FAdV-4 and significantly reduced viral shedding. Moreover, the immunogenicity of TMUVRP-fiber2H was significantly lower than that of TMUVRP-fiber2, which was reflected in the neutralizing antibody titer, survival rate, and virus shedding after challenge. Therefore, our results suggested that TMUV SRIPs are a promising novel platform for the development of vaccines for existing and emerging poultry diseases.
RESUMO
Turtles are an evolutionarily unique and morphologically distinctive order of reptiles, and many species are globally endangered. Although a high diversity of adenoviruses in scaled reptiles is well-documented, turtle adenoviruses remain largely understudied. To investigate their molecular diversity, we focused on the identification and characterisation of adenoviruses in turtle-derived organ, swab and egg samples. Since reptile circoviruses have been scarcely reported and no turtle circoviruses have been documented to date, we also screened our samples for circoviruses. Host-virus coevolution is a common feature of these viral families, so we aimed to investigate possible signs of this as well. Two screening projects were conducted: one on Brazilian samples collected from animals in their natural habitat, and the other on Hungarian pet shop samples. Nested PCR systems were used for the detection of adeno- and circoviruses and purified PCR products were Sanger sequenced. Phylogenetic trees for the viruses were reconstructed based on the adenoviral DNA polymerase and hexon genes, circoviral Rep genes, and for the turtle hosts based on mitochondrial cytochrome b amino acid sequences. During the screening, testadeno-, siadeno-, and circovirus strains were detected. The circovirus strains were classified into the genus Circovirus, exhibiting significant evolutionary divergence but forming a monophyletic clade within a group of fish circoviruses. The phylogenetic tree of turtles reflected their taxonomic relationships, showing a deep bifurcation between suborders and distinct monophyletic clades corresponding to families. A similar clustering pattern was observed among the testadenovirus strains in their phylogenetic tree. As a result, this screening of turtle samples revealed at least three new testadenoviruses, including the first sea turtle adenovirus, evidence of coevolution between testadenoviruses and their hosts, and the first turtle associated circoviruses. These findings underscore the need for further research on viruses in turtles, and more broadly in reptiles, to better understand their viral diversity and the evolutionary processes shaping host-virus interactions.
RESUMO
Various viral proteins are post-translationally modified by SUMO-conjugation during the human adenovirus (HAdV) replication cycle. This modification leads to diverse consequences for target proteins as it influences their intracellular localization or cell transformation capabilities. SUMOylated HAdV proteins include the multifunctional oncoprotein E1B-55K. Our previous research, along with that of others, has demonstrated a substantial influence of yet another adenoviral oncoprotein, E4orf6, on E1B-55K SUMOylation levels. Protein SUMOylation can be reversed by cellular sentrin/SUMO-specific proteases (SENPs). In this study, we investigated the interaction of E1B-55K with cellular SENPs to understand deSUMOylation activities and their consequences for cell transformation mediated by this adenoviral oncoprotein. We show that E1B-55K interacts with and is deSUMOylated by SENP 1, independently of E4orf6. Consistent with these results, we found that SENP 1 prevents E1A/E1B-dependent focus formation in rodent cells. We anticipate these findings to be the groundwork for future studies on adenovirus-host interactions, the mechanisms that underlie E1B-55K SUMOylation, as well as the role of this major adenoviral oncoprotein in HAdV-mediated cell transformation.
Assuntos
Proteínas E1B de Adenovirus , Adenovírus Humanos , Cisteína Endopeptidases , Sumoilação , Adenovírus Humanos/fisiologia , Adenovírus Humanos/metabolismo , Adenovírus Humanos/genética , Humanos , Cisteína Endopeptidases/metabolismo , Cisteína Endopeptidases/genética , Animais , Proteínas E1B de Adenovirus/metabolismo , Proteínas E1B de Adenovirus/genética , Transformação Celular Viral , Interações Hospedeiro-Patógeno , Linhagem Celular , Células HEK293 , Processamento de Proteína Pós-Traducional , CamundongosRESUMO
This study aimed to assess the impact of COVID-19 on the prevalence of adenovirus (AdV) infection in children. This study retrospectively analyzed the changes in the epidemiological and clinical features of AdV-associated respiratory infections in children in Hangzhou, China, between January 2019 and July 2024. A total of 771 316 samples were included in the study, and the positive rate was 6.10% (47 050/771 316). Among them, the positive rate of AdV infection was highest in 2019, reaching 11.29% (26 929/238 333), while the positive rates in the remaining years were between 2% and 9%. In terms of seasonal epidemic characteristics, the summer of 2019 was the peak of AdV incidence, with the positive rate peaking at around 16.95% (7275/45 268), followed by a gradual decline and a low-level epidemic in winter, with a positive rate of 8.79% (8094/92 060). However, during the period 2020-2024, the AdV epidemic season did not show any significant regularity. Gender analysis revealed that the positive rate of male patients was generally greater than that of female patients. In different age groups, the population susceptible to AdV changed before and after the epidemic. In the early and middle stages of the COVID-19 epidemic, the susceptible population was mainly 2-5 years old, whereas in the later stages of the epidemic, the susceptible population was 5-18 years old. In addition, the main clinical symptoms of AdV-positive children from 2019-2024 were respiratory tract symptoms and fever. In summary, the COVID-19 epidemic has had a certain impact on the prevalence of AdV. These findings provide an important basis and reference for the prevention and diagnosis of AdV, especially in the context of increasing age- and gender-specific public health strategies.