Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 129
Filtrar
1.
Medicine (Baltimore) ; 103(31): e39057, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39093763

RESUMEN

BACKGROUND: The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, poses a huge threat to human health. Pancreatic cancer (PC) is a malignant tumor with high mortality. Research suggests that infection with SARS-CoV-2 may increase disease severity and risk of death in patients with pancreatic cancer, while pancreatic cancer may also increase the likelihood of contracting SARS-CoV-2, but the link is unclear. METHODS: This study investigated the transcriptional profiles of COVID-19 and PC patients, along with their respective healthy controls, using bioinformatics and systems biology approaches to uncover the molecular mechanisms linking the 2 diseases. Specifically, gene expression data for COVID-19 and PC patients were obtained from the Gene Expression Omnibus datasets, and common differentially expressed genes (DEGs) were identified. Gene ontology and pathway enrichment analyses were performed on the common DEGs to elucidate the regulatory relationships between the diseases. Additionally, hub genes were identified by constructing a protein-protein interaction network from the shared DEGs. Using these hub genes, we conducted regulatory network analyses of microRNA/transcription factors-genes relationships, and predicted potential drugs for treating COVID-19 and PC. RESULTS: A total of 1722 and 2979 DEGs were identified from the transcriptome data of PC (GSE119794) and COVID-19 (GSE196822), respectively. Among these, 236 common DEGs were found between COVID-19 and PC based on protein-protein interaction analysis. Functional enrichment analysis indicated that these shared DEGs were involved in pathways related to viral genome replication and tumorigenesis. Additionally, 10 hub genes, including extra spindle pole bodies like 1, holliday junction recognition protein, marker of proliferation Ki-67, kinesin family member 4A, cyclin-dependent kinase 1, topoisomerase II alpha, cyclin B2, ubiquitin-conjugating enzyme E2 C, aurora kinase B, and targeting protein for Xklp2, were identified. Regulatory network analysis revealed 42 transcription factors and 23 microRNAs as transcriptional regulatory signals. Importantly, lucanthone, etoposide, troglitazone, resveratrol, calcitriol, ciclopirox, dasatinib, enterolactone, methotrexate, and irinotecan emerged as potential therapeutic agents against both COVID-19 and PC. CONCLUSION: This study unveils potential shared pathogenic mechanisms between PC and COVID-19, offering novel insights for future research and therapeutic strategies for the treatment of PC and SARS-CoV-2 infection.


Asunto(s)
COVID-19 , Biología Computacional , Neoplasias Pancreáticas , Mapas de Interacción de Proteínas , SARS-CoV-2 , Biología de Sistemas , Humanos , COVID-19/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/virología , Biología Computacional/métodos , Biología de Sistemas/métodos , SARS-CoV-2/genética , Mapas de Interacción de Proteínas/genética , Redes Reguladoras de Genes , MicroARNs/genética , MicroARNs/metabolismo , Perfilación de la Expresión Génica/métodos
2.
J Med Virol ; 96(7): e29809, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39016466

RESUMEN

Pancreatic cancer (PC) is a highly aggressive malignancy with a poor prognosis, making early diagnosis crucial for improving patient outcomes. While the gut microbiome, including bacteria and viruses, is believed to be essential in cancer pathogenicity, the potential contribution of the gut virome to PC remains largely unexplored. In this study, we conducted a comparative analysis of the gut viral compositional and functional profiles between PC patients and healthy controls, based on fecal metagenomes from two publicly available data sets comprising a total of 101 patients and 82 healthy controls. Our results revealed a decreasing trend in the gut virome diversity of PC patients with disease severity. We identified significant alterations in the overall viral structure of PC patients, with a meta-analysis revealing 219 viral operational taxonomic units (vOTUs) showing significant differences in relative abundance between patients and healthy controls. Among these, 65 vOTUs were enriched in PC patients, and 154 were reduced. Host prediction revealed that PC-enriched vOTUs preferentially infected bacterial members of Veillonellaceae, Enterobacteriaceae, Fusobacteriaceae, and Streptococcaceae, while PC-reduced vOTUs were more likely to infect Ruminococcaceae, Lachnospiraceae, Clostridiaceae, Oscillospiraceae, and Peptostreptococcaceae. Furthermore, we constructed random forest models based on the PC-associated vOTUs, achieving an optimal average area under the curve (AUC) of up to 0.879 for distinguishing patients from controls. Through additional 10 public cohorts, we demonstrated the reproducibility and high specificity of these viral signatures. Our study suggests that the gut virome may play a role in PC development and could serve as a promising target for PC diagnosis and therapeutic intervention. Future studies should further explore the underlying mechanisms of gut virus-bacteria interactions and validate the diagnostic models in larger and more diverse populations.


Asunto(s)
Heces , Microbioma Gastrointestinal , Metagenómica , Neoplasias Pancreáticas , Viroma , Humanos , Neoplasias Pancreáticas/virología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/microbiología , Microbioma Gastrointestinal/genética , Metagenómica/métodos , Heces/virología , Heces/microbiología , Virus/aislamiento & purificación , Virus/genética , Virus/clasificación , Metagenoma , Bacterias/aislamiento & purificación , Bacterias/clasificación , Bacterias/genética , Persona de Mediana Edad , Masculino , Femenino , Anciano , Estudios de Casos y Controles
4.
Viruses ; 13(6)2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071585

RESUMEN

Although the oncolytic parvovirus H-1PV has entered clinical trials, predicting therapeutic success remains challenging. We investigated whether the antiviral state in tumor cells determines the parvoviral oncolytic efficacy. The interferon/interferon-stimulated genes (IFN/ISG)-circuit and its major configurator, human endogenous retroviruses (HERVs), were evaluated using qRT-PCR, ELISA, Western blot, and RNA-Seq techniques. In pancreatic cancer cell lines, H-1PV caused a late global shutdown of innate immunity, whereby the concomitant inhibition of HERVs and IFN/ISGs was co-regulatory rather than causative. The growth-inhibitory IC50 doses correlated with the power of suppression but not with absolute ISG levels. Moreover, H-1PV was not sensitive to exogenous IFN despite upregulated antiviral ISGs. Such resistance questioned the biological necessity of the oncotropic ISG-shutdown, which instead might represent a surrogate marker for personalized oncolytic efficacy. The disabled antiviral homeostasis may modify the activity of other viruses, as demonstrated by the reemergence of endogenous AluY-retrotransposons. This way of suppression may compromise the interferogenicity of drugs having gemcitabine-like mechanisms of action. This shortcoming in immunogenic cell death induction is however amendable by immune cells which release IFN in response to H-1PV.


Asunto(s)
Parvovirus H-1/inmunología , Parvovirus H-1/patogenicidad , Homeostasis/inmunología , Inmunidad Innata , Interferones/inmunología , Neoplasias Pancreáticas/virología , Muerte Celular/inmunología , Línea Celular Tumoral , Citocinas , Humanos , Leucocitos Mononucleares/virología , Virus Oncolíticos/genética , Virus Oncolíticos/inmunología , Virus Oncolíticos/patogenicidad , Infecciones por Parvoviridae/complicaciones , Infecciones por Parvoviridae/virología
5.
Mol Cancer Ther ; 20(8): 1481-1494, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34045231

RESUMEN

Oncolytic vaccinia viruses have promising efficacy and safety profiles in cancer therapy. Although antitumor activity can be increased by manipulating viral genes, the relative efficacy of individual modifications has been difficult to assess without side-by-side comparisons. This study sought to compare the initial antitumor activity after intravenous administration of five vaccinia virus variants of the same Western Reserve backbone and thymidine kinase gene deletion in RIP-Tag2 transgenic mice with spontaneous pancreatic neuroendocrine tumors. Tumors had focal regions of infection at 5 days after all viruses. Natural killer (NK) cells were restricted to these sites of infection, but CD8+ T cells and tumor cell apoptosis were widespread and varied among the viruses. Antitumor activity of virus VV-A34, bearing amino acid substitution A34K151E to increase viral spreading, and virus VV-IL2v, expressing a mouse IL2 variant (mIL2v) with attenuated IL2 receptor alpha subunit binding, was similar to control virus VV-GFP. However, antitumor activity was significantly greater after virus VV-A34/IL2v, which expressed mIL2v together with A34K151E mutation and viral B18R gene deletion, and virus VV-GMCSF that expressed mouse GM-CSF. Both viruses greatly increased expression of CD8 antigens Cd8a/Cd8b1 and cytotoxicity genes granzyme A, granzyme B, Fas ligand, and perforin-1 in tumors. VV-A34/IL2v led to higher serum IL2 and greater tumor expression of death receptor ligand TRAIL, but VV-GMCSF led to higher serum GM-CSF, greater expression of leukocyte chemokines and adhesion molecules, and more neutrophil recruitment. Together, the results show that antitumor activity is similarly increased by viral expression of GM-CSF or IL2v combined with additional genetic modifications.


Asunto(s)
Apoptosis , Citocinas/metabolismo , Inmunidad , Tumores Neuroendocrinos/terapia , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/terapia , Virus Vaccinia/genética , Animales , Linfocitos T CD8-positivos/inmunología , Proliferación Celular , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Humanos , Interleucina-2/genética , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Tumores Neuroendocrinos/metabolismo , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/virología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/virología , Células Tumorales Cultivadas
6.
Commun Biol ; 4(1): 368, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33742099

RESUMEN

High expression levels of human epidermal growth factor receptor 2 (HER2) have been associated with poor prognosis in patients with pancreatic adenocarcinoma (PDAC). However, HER2-targeting immunotherapies have been unsuccessful to date. Here we increase the breadth, potency, and duration of anti-PDAC HER2-specific CAR T-cell (HER2.CART) activity with an oncolytic adeno-immunotherapy that produces cytokine, immune checkpoint blockade, and a safety switch (CAdTrio). Combination treatment with CAdTrio and HER2.CARTs cured tumors in two PDAC xenograft models and produced durable tumor responses in humanized mice. Modifications to the tumor immune microenvironment contributed to the antitumor activity of our combination immunotherapy, as intratumoral CAdTrio treatment induced chemotaxis to enable HER2.CART migration to the tumor site. Using an advanced PDAC model in humanized mice, we found that local CAdTrio treatment of primary tumor stimulated systemic host immune responses that repolarized distant tumor microenvironments, improving HER2.CART anti-tumor activity. Overall, our data demonstrate that CAdTrio and HER2.CARTs provide complementary activities to eradicate metastatic PDAC and may represent a promising co-operative therapy for PDAC patients.


Asunto(s)
Adenoviridae/patogenicidad , Carcinoma Ductal Pancreático/terapia , Inmunoterapia Adoptiva , Viroterapia Oncolítica , Virus Oncolíticos/patogenicidad , Neoplasias Pancreáticas/terapia , Receptores Quiméricos de Antígenos/inmunología , Linfocitos T/trasplante , Antígeno B7-H1/inmunología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/inmunología , Carcinoma Ductal Pancreático/virología , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Interleucina-12/genética , Masculino , Metástasis de la Neoplasia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/inmunología , Neoplasias Pancreáticas/virología , Receptor ErbB-2/genética , Receptores Quiméricos de Antígenos/genética , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Linfocitos T/inmunología , Carga Tumoral , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Am J Case Rep ; 21: e921269, 2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-32176677

RESUMEN

BACKGROUND Epstein-Barr virus (EBV)-positive diffuse large B-cell lymphoma (DLBCL) is a rare subtype of B-cell neoplasm that can have diverse presentations. When it involves the pancreas (i.e., pancreatic lymphoma), it can mimic metastatic pancreatic adenocarcinoma. Pancreatic lymphoma and adenocarcinoma often have similar clinical, laboratory, and radiographic features making the distinction challenging without pathological tissue examination. The differentiation of these 2 entities is important as the prognosis of pancreatic lymphoma is certainly more favorable with a chance of cure with chemoimmunotherapy. CASE REPORT We present an unusual case of EBV-positive DLBCL involving the pancreas that was initially believed to be metastatic pancreatic adenocarcinoma. The patient was treated with chemoimmunotherapy and had a remarkable response. This is the first known case of EBV-positive DLBCL involving the pancreas that was successfully treated with chemoimmunotherapy. CONCLUSIONS EBV-positive DLBCL can have diverse presentations, including a pancreatic mass with multi-organ involvement, which mimics metastatic pancreatic adenocarcinoma. The prognosis of EBV-positive DLBCL is thought to be worse than that of EBV-negative tumors. However, it remains certainly superior to that of its adenocarcinoma counterpart with conventional chemoimmunotherapy.


Asunto(s)
Infecciones por Virus de Epstein-Barr/patología , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/terapia , Adenocarcinoma/diagnóstico , Terapia Combinada , Diagnóstico Diferencial , Quimioterapia , Humanos , Inmunoterapia , Linfoma de Células B Grandes Difuso/virología , Masculino , Persona de Mediana Edad , Neoplasias Pancreáticas/virología , Pronóstico , Resultado del Tratamiento
9.
Cancer Epidemiol ; 65: 101691, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32088651

RESUMEN

BACKGROUND: A growing body of evidence has suggested an association between Hepatitis C virus (HCV) infection and risk of pancreatic cancer (PAC). Herein, we conducted a meta-analysis of available evidence to explore this association. METHODS: We systematically retrieved studies that investigated the association between HCV infection and risk of PAC. Pooled odds ratio (OR) with corresponding 95 % confidence interval (CI) of PAC for patients with HCV infection was calculated using the fixed- or random-effects model. RESULTS: A total of 16 studies (8 cohort and 8 case-control) were included in this meta-analysis. Combined, patients with HCV infection were more likely to develop PAC than people without it (pooled OR = 1.51, 95 % CI: 1.31, 1.74; I2 = 63.49 %, p-value for heterogeneity< 0.001). Studies that adjusted their results for diabetes, chronic pancreatitis, alcohol intake, and smoking showed lower ORs than studies that did not adjust for them. CONCLUSION: HCV infection was associated with increased risk of PAC, but this association was attenuated among studies that adjusted their results for potential risk factors for PAC. Future prospective cohort studies are needed to confirm this association.


Asunto(s)
Hepatitis C/epidemiología , Hepatitis C/patología , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/virología , Estudios de Casos y Controles , Estudios de Cohortes , Femenino , Humanos , Masculino , Factores de Riesgo
10.
J Virol ; 94(3)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31694943

RESUMEN

Vesicular stomatitis virus (VSV) based oncolytic viruses are promising agents against various cancers. We have shown that pancreatic ductal adenocarcinoma (PDAC) cell lines exhibit great diversity in susceptibility and permissibility to VSV. Here, using a directed evolution approach with our two previously described oncolytic VSV recombinants, VSV-p53wt and VSV-p53-CC, we generated novel oncolytic VSVs with an improved ability to replicate in virus-resistant PDAC cell lines. VSV-p53wt and VSV-p53-CC encode a VSV matrix protein (M) with a ΔM51 mutation (M-ΔM51) and one of two versions of a functional human tumor suppressor, p53, fused to a far-red fluorescent protein, eqFP650. Each virus was serially passaged 32 times (which accounts for more than 60 viral replication cycles) on either the SUIT-2 (moderately resistant to VSV) or MIA PaCa-2 (highly permissive to VSV) human PDAC cell lines. While no phenotypic changes were observed for MIA PaCa-2-passaged viruses, both SUIT-2-passaged VSV-p53wt and VSV-p53-CC showed improved replication in SUIT-2 and AsPC-1, another human PDAC cell line also moderately resistant to VSV, while remaining highly attenuated in nonmalignant cells. Surprisingly, two identical VSV glycoprotein (VSV-G) mutations, K174E and E238K, were identified in both SUIT-2-passaged viruses. Additional experiments indicated that the acquired G mutations improved VSV replication, at least in part due to improved virus attachment to SUIT-2 cells. Importantly, no mutations were found in the M-ΔM51 protein, and no deletions or mutations were found in the p53 or eqFP650 portions of virus-carried transgenes in any of the passaged viruses, demonstrating long-term genomic stability of complex VSV recombinants carrying large transgenes.IMPORTANCE Vesicular stomatitis virus (VSV)-based oncolytic viruses are promising agents against pancreatic ductal adenocarcinoma (PDAC). However, some PDAC cell lines are resistant to VSV. Here, using a directed viral evolution approach, we generated novel oncolytic VSVs with an improved ability to replicate in virus-resistant PDAC cell lines, while remaining highly attenuated in nonmalignant cells. Two independently evolved VSVs obtained 2 identical VSV glycoprotein mutations, K174E and E238K. Additional experiments indicated that these acquired G mutations improved VSV replication, at least in part due to improved virus attachment to SUIT-2 cells. Importantly, no deletions or mutations were found in the virus-carried transgenes in any of the passaged viruses. Our findings demonstrate long-term genomic stability of complex VSV recombinants carrying large transgenes and support further clinical development of oncolytic VSV recombinants as safe therapeutics for cancer.


Asunto(s)
Carcinoma Ductal Pancreático/virología , Neoplasias Pancreáticas/virología , Estomatitis Vesicular/virología , Virus de la Estomatitis Vesicular Indiana/genética , Replicación Viral/fisiología , Línea Celular Tumoral , Glicoproteínas/química , Glicoproteínas/genética , Humanos , Modelos Moleculares , Mutación , Viroterapia Oncolítica , Virus Oncolíticos/genética , Conformación Proteica , Proteínas Recombinantes , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Virus de la Estomatitis Vesicular Indiana/fisiología , Proteínas Virales de Fusión/química , Proteínas de la Matriz Viral , Proteínas Virales , Acoplamiento Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...