Your browser doesn't support javascript.
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Int J Mol Sci ; 23(24)2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: covidwho-20234933

RESUMEN

Fusion among different cell populations represents a rare process that is mediated by both intrinsic and extracellular events. Cellular hybrid formation is relayed by orchestrating tightly regulated signaling pathways that can involve both normal and neoplastic cells. Certain important cell merger processes are often required during distinct organismal and tissue development, including placenta and skeletal muscle. In a neoplastic environment, however, cancer cell fusion can generate new cancer hybrid cells. Following survival during a subsequent post-hybrid selection process (PHSP), the new cancer hybrid cells express different tumorigenic properties. These can include elevated proliferative capacity, increased metastatic potential, resistance to certain therapeutic compounds, and formation of cancer stem-like cells, all of which characterize significantly enhanced tumor plasticity. However, many parts within this multi-step cascade are still poorly understood. Aside from intrinsic factors, cell fusion is particularly affected by extracellular conditions, including an inflammatory microenvironment, viruses, pH and ionic stress, hypoxia, and exosome signaling. Accordingly, the present review article will primarily highlight the influence of extracellular events that contribute to cell fusion in normal and tumorigenic tissues.


Asunto(s)
Carcinogénesis , Células Madre Neoplásicas , Humanos , Fusión Celular , Línea Celular Tumoral , Células Híbridas , Carcinogénesis/metabolismo , Células Madre Neoplásicas/metabolismo , Microambiente Tumoral
2.
Respir Res ; 24(1): 113, 2023 Apr 15.
Artículo en Inglés | MEDLINE | ID: covidwho-2303662

RESUMEN

BACKGROUND: Centromere protein O (CENPO) is a newly discovered constitutive centromeric protein, associated with cell death. However, little is known about how CENPO expression is associated with human cancers or immune infiltration. Here, we assessed the function of CENPO in pan-cancer and further verified the results in lung adenocarcinoma (LUAD) through in vitro and in vivo experiments. METHODS: Sangerbox and TCGA databases were used to evaluate the CENPO expression level in different human cancer types. A subsequent evaluation of the potential role of CENPO as a diagnostic and prognostic biomarker in pancancer was conducted. The CENPO mutations were analyzed using the cBioPortal database and its function was analyzed using the LinkedOmics and CancerSEA databases. The TIMER2 and TISIDB websites were used to find out how CENPO affects immune infiltration. The expression level of CENPO in LUAD was revealed by TCGA database and immunohistochemical (IHC) staining. Targetscan, miRWalk, miRDB, miRabel, LncBase databases, and Cytoscape tool were used to identify microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) that regulate expression and construct ceRNA network. Subsequently, loss-of-function assays were performed to identify the functions of CENPO on the malignant behavior and tumor growth of LUAD in vitro and in vivo experiments. RESULTS: In most cancers, CENPO was upregulated and mutated, which predicted a poorer prognosis. Furthermore, infiltration of CENPO and myeloid-derived suppressor cells (MDSC) showed a significant positive correlation, while T-cell NK infiltration showed a significant negative correlation in most cancers. CENPO was expressed at high levels in LUAD and was correlated with p-TNM stage. Furthermore, CENPO knockdown suppressed the malignant phenotypes of LUAD cells, manifested by slower proliferation, cycle in G2, increased apoptosis, decreased migration, and attenuated tumorigenesis. Furthermore, CENPO knockdown decreased CDK1/6, PIK3CA, and inhibited mTOR phosphorylation, suggesting that the mTOR signaling pathway may be involved in CENPO-mediated regulation of LUAD development. CONCLUSIONS: In pan-cancer, especially LUAD, CENPO may be a potential biomarker and oncogene. Furthermore, CENPO has been implicated in immune cell infiltration in pan-cancer and represents a potential immunotherapeutic target for tumor therapy.


Asunto(s)
Adenocarcinoma , Neoplasias Pulmonares , Humanos , Carcinogénesis , Muerte Celular , Óxidos N-Cíclicos , Neoplasias Pulmonares/genética , Pronóstico , Proteínas Cromosómicas no Histona
3.
J Biol Chem ; 299(5): 104658, 2023 05.
Artículo en Inglés | MEDLINE | ID: covidwho-2270108

RESUMEN

Eukaryotic initiation factor 3d (eIF3d), a known RNA-binding subunit of the eIF3 complex, is a 66 to 68-kDa protein with an RNA-binding motif and a cap-binding domain. Compared with other eIF3 subunits, eIF3d is relatively understudied. However, recent progress in studying eIF3d has revealed a number of intriguing findings on its role in maintaining eIF3 complex integrity, global protein synthesis, and in biological and pathological processes. It has also been reported that eIF3d has noncanonical functions in regulating translation of a subset of mRNAs by binding to 5'-UTRs or interacting with other proteins independent of the eIF3 complex and additional functions in regulating protein stability. The noncanonical regulation of mRNA translation or protein stability may contribute to the role of eIF3d in biological processes such as metabolic stress adaptation and in disease onset and progression including severe acute respiratory syndrome coronavirus 2 infection, tumorigenesis, and acquired immune deficiency syndrome. In this review, we critically evaluate the recent studies on these aspects of eIF3d and assess prospects in understanding the function of eIF3d in regulating protein synthesis and in biological and pathological processes.


Asunto(s)
Progresión de la Enfermedad , Factor 3 de Iniciación Eucariótica , Biosíntesis de Proteínas , Caperuzas de ARN , Humanos , COVID-19 , Factor 3 de Iniciación Eucariótica/metabolismo , Caperuzas de ARN/metabolismo , Síndrome de Inmunodeficiencia Adquirida , Carcinogénesis , Regiones no Traducidas 5'/genética
4.
Asian Pac J Cancer Prev ; 23(7): 2191-2197, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: covidwho-2282302

RESUMEN

The widespread infectious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) significantly impacts major human diseases. It is undoubtedly evident that cancer patients are more susceptible to the infection and at a higher risk of severe COVID-19 than the general population. Moreover, the rise in cancers incidence is waiting in the Globe as a long-term effect of post-COVID-19 complications. Multiple mostly unknown mechanisms participate and determine the oncogenic impact of virus-induced transformation. Imbalance in oncogenesis is considered critical in cancer development. Modified immunogenicity and metabolic features emerge as pivotal in COVID-19 pathogenesis and the organism system's response. The molecular mechanisms of the onset of the metabolic disorder have not yet been fully elucidated. The pathology is complicated, multifactorial, and emerging in various processes. Preventive anticancer therapy taking into account the change in metabolic processes, helps them respond better to anti-COVID-19 treatment than relying only on antiviral drugs. The modified therapeutic algorithm was provided to reduce the likelihood of post-acute complications in patients with preexisting pathologies and the onset of other chronic pathologies and cancers.


Asunto(s)
COVID-19 , Antivirales/uso terapéutico , COVID-19/complicaciones , Carcinogénesis , Humanos , ARN Viral , SARS-CoV-2
5.
Arch Iran Med ; 25(7): 450-455, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: covidwho-2270276

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARSCoV-2) continues to be a worldwide healthcare problem. While our knowledge of the interaction of cancer and its management with COVID-19 mortality is gradually evolving, there are still many unanswered questions regarding the impact of COVID-19 on cancer and its prognosis. Several factors activated during COVID-19 have been implicated in tumorigenesis and the development of metastasis. Inflammation, hypoxia, reduced levels of angiotensin converting enzyme 2, elevated levels of Interleukin 6 and some other cytokines that are hallmarks of COVID-19 are capable of inducing tumor relapse and metastasis. On the other hand, there are reports that COVID-19 has been associated with cancer cure. Understanding the interaction between COVID-19 and tumor cells is essential for evaluating the potential long-term risks of COVID-19 in cancer patients, and for scheduling necessary preventive and therapeutic interventions. In this review, we briefly overview the potential impacts that COVID-19 might have on tumorigenesis and cancer relapse, as well as the role that COVID-19 might play in cancer remission and cure.


Asunto(s)
COVID-19 , Enfermedades Pulmonares , Humanos , SARS-CoV-2 , Recurrencia , Carcinogénesis
6.
Curr Oncol ; 30(2): 1924-1944, 2023 02 05.
Artículo en Inglés | MEDLINE | ID: covidwho-2225090

RESUMEN

As per a recent study conducted by the WHO, 15.4% of all cancers are caused by infectious agents of various categories, and more than 10% of them are attributed to viruses. The emergence of COVID-19 has once again diverted the scientific community's attention toward viral diseases. Some researchers have postulated that SARS-CoV-2 will add its name to the growing list of oncogenic viruses in the long run. However, owing to the complexities in carcinogenesis of viral origin, researchers across the world are struggling to identify the common thread that runs across different oncogenic viruses. Classical pathways of viral oncogenesis have identified oncogenic mediators in oncogenic viruses, but these mediators have been reported to act on diverse cellular and multiple omics pathways. In addition to viral mediators of carcinogenesis, researchers have identified various host factors responsible for viral carcinogenesis. Henceforth owing to viral and host complexities in viral carcinogenesis, a singular mechanistic pathway remains yet to be established; hence there is an urgent need to integrate concepts from system biology, cancer microenvironment, evolutionary perspective, and thermodynamics to understand the role of viruses as drivers of cancer. In the present manuscript, we provide a holistic view of the pathogenic pathways involved in viral oncogenesis with special emphasis on alteration in the tumor microenvironment, genomic alteration, biological entropy, evolutionary selection, and host determinants involved in the pathogenesis of viral tumor genesis. These concepts can provide important insight into viral cancers, which can have an important implication for developing novel, effective, and personalized therapeutic options for treating viral cancers.


Asunto(s)
COVID-19 , Neoplasias , Humanos , SARS-CoV-2 , Virus Oncogénicos , Neoplasias/genética , Carcinogénesis , Genómica , Microambiente Tumoral
7.
researchsquare; 2023.
Preprint en Inglés | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-2484556.v1

RESUMEN

β-propiolactone (BPL) is a common inactivator agent used in vaccines. Due to BPL carcinogenicity, complete hydrolysis of it is necessary to prevent cytotoxicity in mammalian cells. As a result, more attention should be paid to it at the clinic and it is important to measure its trace amounts. BPL analysis is challenging due to its instability. A simple and fast gas chromatography-mass spectrometry (GC-MS) method was developed for quantitation of residual BPL in inactivated covid-19 vaccines. Caprolactone was used as internal standard in samples solutions, the analysis was performed after extraction of analyte from vaccine media by ethyl acetate. The validity of the method was studied with a linearity of r2 > 0.99 over the concentration range of 0.2–20 µg/mL with the limit of detection and the limit of quantification of 0.68 and 2.06 µg/mL, respectively. The target analyte BPL was not detected in the samples, demonstrating the test samples were qualified. The established method can be used for quality control of inactivated covid-19 vaccines.


Asunto(s)
COVID-19 , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Carcinogénesis
8.
medrxiv; 2022.
Preprint en Inglés | medRxiv | ID: ppzbmed-10.1101.2022.12.22.22283851

RESUMEN

Background Previously developed TaME-seq method for deep sequencing of HPV, allowed simultaneous identification of the HPV DNA consensus sequence, low-frequency variable sites, and chromosomal integration events. The method has been successfully validated and applied to the study of five carcinogenic high-risk (HR) HPV types (HPV16, 18, 31, 33, and 45). Here, we present TaME-seq2 with an updated laboratory workflow and bioinformatics pipeline. The HR-HPV type repertoire was expanded with HPV51, 52, and 59. As a proof-of-concept, TaME-seq2 was applied on SARS-CoV-2 positive samples showing the method's flexibility to a broader range of viruses, both DNA and RNA. Results Compared to TaME-seq version 1, the bioinformatics pipeline of TaME-seq2 is approximately 40x faster. In total, 23 HPV-positive samples and seven SARS-CoV-2 clinical samples passed the threshold of 300x mean depth and were submitted to further analysis. The mean number of variable sites per 1000 bp was ~ 1.5x higher in SARS-CoV-2 than in HPV-positive samples. Reproducibility and repeatability of the method were tested on a subset of samples. A viral integration breakpoint followed by a partial genomic deletion was found in within-run replicates of HPV59-positive sample. Identified viral consensus sequence in two separate runs was >99.9 % identical between replicates, differing by a couple of nucleotides identified in only one of the replicates. Conversely, the number of identical minor nucleotide variants (MNVs) differed greatly between replicates, probably caused by PCR-introduced bias. The total number of detected MNVs, calculated gene variability and mutational signature analysis, were unaffected by the sequencing run. Conclusion TaME-seq2 proved well suited for consensus sequence identification, and the detection of low-frequency viral genome variation and viral-chromosomal integrations. The repertoire of TaME-seq2 now encompasses seven HR-HPV types. Our goal is to further include all HR-HPV types in the TaME-seq2 repertoire. Moreover, with a minor modification of previously developed primers, the same method was successfully applied for the analysis of SARS-CoV-2 positive samples, implying the ease of adapting TaME-seq2 to other viruses.


Asunto(s)
Carcinogénesis
9.
researchsquare; 2022.
Preprint en Inglés | PREPRINT-RESEARCHSQUARE | ID: ppzbmed-10.21203.rs.3.rs-2404996.v1

RESUMEN

Background Previously developed TaME-seq method for deep sequencing of HPV, allowed simultaneous identification of the HPV DNA consensus sequence, low-frequency variable sites, and chromosomal integration events. The method has been successfully validated and applied to the study of five carcinogenic high-risk (HR) HPV types (HPV16, 18, 31, 33, and 45). Here, we present TaME-seq2 with an updated laboratory workflow and bioinformatics pipeline. The HR-HPV type repertoire was expanded with HPV51, 52, and 59. As a proof-of-concept, TaME-seq2 was applied on SARS-CoV-2 positive samples showing the method's flexibility to a broader range of viruses, both DNA and RNA. Results Compared to TaME-seq version 1, the bioinformatics pipeline of TaME-seq2 is approximately 40x faster. In total, 23 HPV-positive samples and seven SARS-CoV-2 clinical samples passed the threshold of 300x mean depth and were submitted to further analysis. The mean number of variable sites per 1000 bp was ~ 1.5x higher in SARS-CoV-2 than in HPV-positive samples.  Reproducibility and repeatability of the method were tested on a subset of samples. A viral integration breakpoint followed by a partial genomic deletion was found in within-run replicates of HPV59-positive sample. Identified viral consensus sequence in two separate runs was >99.9 % identical between replicates, differing by a couple of nucleotides identified in only one of the replicates. Conversely, the number of identical minor nucleotide variants (MNVs) differed greatly between replicates, probably caused by PCR-introduced bias. The total number of detected MNVs, calculated gene variability and mutational signature analysis, were unaffected by the sequencing run. Conclusion TaME-seq2 proved well suited for consensus sequence identification, and the detection of low-frequency viral genome variation and viral-chromosomal integrations. The repertoire of TaME-seq2 now encompasses seven HR-HPV types. Our goal is to further include all HR-HPV types in the TaME-seq2 repertoire. Moreover, with a minor modification of previously developed primers, the same method was successfully applied for the analysis of SARS-CoV-2 positive samples, implying the ease of adapting TaME-seq2 to other viruses.


Asunto(s)
Carcinogénesis
10.
J Microbiol ; 60(11): 1106-1112, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: covidwho-2075669

RESUMEN

Due to the evolutionary arms race between hosts and viruses, viruses must adapt to host translation systems to rapidly synthesize viral proteins. Highly expressed genes in hosts have a codon bias related to tRNA abundance, the primary RNA translation rate determinant. We calculated the relative synonymous codon usage (RSCU) of three hepatitis viruses (HAV, HBV, and HCV), SARS-CoV-2, 30 human tissues, and hepatocellular carcinoma (HCC). After comparing RSCU between viruses and human tissues, we calculated the codon adaptation index (CAI) of viral and human genes. HBV and HCV showed the highest correlations with HCC and the normal liver, while SARS-CoV-2 had the strongest association with lungs. In addition, based on HCC RSCU, the CAI of HBV and HCV genes was the highest. HBV and HCV preferentially adapt to the tRNA pool in HCC, facilitating viral RNA translation. After an initial trigger, rapid HBV/HCV translation and replication may change normal liver cells into HCC cells. Our findings reveal a novel perspective on virus-mediated oncogenesis.


Asunto(s)
COVID-19 , Carcinoma Hepatocelular , Hepatitis B , Hepatitis C , Neoplasias Hepáticas , Humanos , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/genética , Virus de la Hepatitis B/genética , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/genética , Hepatitis B/complicaciones , Hepatitis B/genética , Transcriptoma , SARS-CoV-2 , Codón , Carcinogénesis , ARN de Transferencia , Hepatitis C/genética
11.
Biomed Pharmacother ; 156: 113783, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: covidwho-2060453

RESUMEN

Pentraxin-3 (PTX3) is the prototype of the long pentraxin subfamily, an acute-phase protein consisting of a C-terminal pentraxin domain and a unique N-terminal domain. PTX3 was initially isolated from human umbilical vein endothelial cells and human FS-4 fibroblasts. It was subsequently found to be also produced by synoviocytes, chondrocytes, osteoblasts, smooth muscle cells, myeloid dendritic cells, epithelial cells, and tumor cells. Various modulatory factors, such as miRNAs, cytokines, drugs, and hypoxic conditions, could regulate the expression level of PTX3. PTX3 is essential in regulating innate immunity, inflammation, angiogenesis, and tissue remodeling. Besides, PTX3 may play dual (pro-tumor and anti-tumor) roles in oncogenesis. PTX3 is involved in the occurrence and development of many non-cancerous diseases, including COVID-19, and might be a potential biomarker indicating the prognosis, activity,and severity of diseases. In this review, we summarize and discuss the potential roles of PTX3 in the oncogenesis and pathogenesis of non-cancerous diseases and potential targeted therapies based on PTX3.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Proteína C-Reactiva/genética , Proteína C-Reactiva/metabolismo , Inflamación/metabolismo , Inmunidad Innata , Carcinogénesis
12.
Int J Mol Sci ; 23(15)2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: covidwho-1994080

RESUMEN

Landmark discoveries in molecular oncology have provided a wide-angle overview of the heterogenous and therapeutically challenging nature of cancer. The power of modern 'omics' technologies has enabled researchers to deeply and comprehensively characterize molecular mechanisms underlying cellular functions. Interestingly, high-throughput technologies have opened new horizons for the design and scientific fool-proof evaluation of the pharmacological properties of targeted chemical compounds to tactfully control the activities of the oncogenic protein networks. Groundbreaking discoveries have galvanized the expansion of the repertoire of available pharmacopoeia to therapeutically target a myriad of deregulated oncogenic pathways. Natural product research has undergone substantial broadening, and many of the drugs which constitute the backbone of modern pharmaceuticals have been derived from the natural cornucopia. Baicalein has gradually gained attention because of its unique ability to target different oncogenic signal transduction cascades in various cancers. We have partitioned this review into different sub-sections to provide a broader snapshot of the oncogenic pathways regulated by baicalein. In this review, we summarize baicalein-mediated targeting of WNT/ß-catenin, AKT/mTOR, JAK/STAT, MAPK, and NOTCH pathways. We also critically analyze how baicalein regulates non-coding RNAs (microRNAs and long non-coding RNAs) in different cancers. Finally, we conceptually interpret baicalein-mediated inhibition of primary and secondary growths in xenografted mice.


Asunto(s)
Flavanonas , MicroARNs , Neoplasias , Animales , Carcinogénesis , Flavanonas/farmacología , Flavanonas/uso terapéutico , Ratones , MicroARNs/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal
13.
J Nutr Biochem ; 108: 109092, 2022 10.
Artículo en Inglés | MEDLINE | ID: covidwho-1983513

RESUMEN

Both obesity and cancer are complex medical conditions that are considered public health problems. The influence of obesity on the predisposition to develop various types of cancer has been observed in a wide variety of studies. Due to their importance as public health problems, and the close relationship between both conditions, it is important to be able to understand and associate them mechanistically. In this review article, we intend to go a little further, by finding relationships between lifestyle, which can lead a person to develop obesity, and how it influences at the cellular and molecular level, affecting gene expression to favor signaling pathways or transcriptional programs involved in cancer. We describe how products of metabolism and intermediate metabolism can affect chromatin structure, participating in the regulation (or dysregulation) of gene expression, and we show an analysis of genes that are responsive to diets high in sugar and fat, and how their epigenetic landscape is altered.


Asunto(s)
Epigénesis Genética , Pandemias , Carcinogénesis/genética , Dieta/efectos adversos , Humanos , Obesidad/metabolismo
14.
Mol Cell Proteomics ; 21(7): 100247, 2022 07.
Artículo en Inglés | MEDLINE | ID: covidwho-1907570

RESUMEN

Since the discovery of oncogenes, there has been tremendous interest to understand their mechanistic basis and to develop broadly actionable therapeutics. Some of the most frequently activated oncogenes driving diverse cancers are c-MYC, EGFR, HER2, AKT, KRAS, BRAF, and MEK. Using a reductionist approach, we explored how cellular proteomes are remodeled in isogenic cell lines engineered with or without these driver oncogenes. The most striking discovery for all oncogenic models was the systematic downregulation of scores of antiviral proteins regulated by type 1 interferon. These findings extended to cancer cell lines and patient-derived xenograft models of highly refractory pancreatic cancer and osteosarcoma driven by KRAS and MYC oncogenes. The oncogenes reduced basal expression of and autocrine stimulation by type 1 interferon causing remarkable convergence on common phenotypic and functional profiles. In particular, there was dramatically lower expression of dsRNA sensors including DDX58 (RIG-I) and OAS proteins, which resulted in attenuated functional responses when the oncogenic cells were treated with the dsRNA mimetic, polyI:C, and increased susceptibility to infection with an RNA virus shown using SARS-CoV-2. Our reductionist approach provides molecular and functional insights connected to immune evasion hallmarks in cancers and suggests therapeutic opportunities.


Asunto(s)
COVID-19 , Interferón beta , Oncogenes , Proteómica , Animales , Factores de Restricción Antivirales , COVID-19/inmunología , Carcinogénesis , Línea Celular Tumoral , Humanos , Interferón beta/inmunología , Proteínas Proto-Oncogénicas p21(ras)/genética , SARS-CoV-2
15.
Front Immunol ; 13: 840785, 2022.
Artículo en Inglés | MEDLINE | ID: covidwho-1809395

RESUMEN

The COVID-19 pandemic caused by SARS-CoV-2 infection has placed health systems under excessive pressure and especially elderly people with cancer. Glioblastoma multiforme (GBM) is a malignant brain tumor with an increasing incidence in elderly individuals, and thereby GBM patients are a vulnerable population during the COVID-19 outbreak. Accumulating studies have implied that SARS-CoV-2 might invade the brain directly via coronavirus receptors. However, little is known about SARS-CoV-2 infection in the clinical development of GBM. Here, we explored the oncogenic roles of six coronavirus receptors (ACE2, DPP4, ANPEP, AXL, TMPRSS2, and ENPEP) in GBM using bioinformatics and experimental approaches. We found that ANPEP and ENPEP were significantly increased at both the mRNA and protein levels in GBM compared with normal brain tissue. Kaplan-Meier survival curves and Cox regression analysis demonstrated that high expressions of ANPEP and ENPEP are associated with poor prognosis and survival. Moreover, all receptors are positively correlated with the immune infiltration levels of monocyte. Furthermore, we identified 245 genes between COVID-19 and coronavirus receptors-correlated genes in GBM and performed a thorough analysis of their protein-protein interaction network, functional signaling pathway and molecular process. Our work explores for the first time the association of coronavirus receptors with GBM and suggests ANPEP and ENPEP as potential therapeutic targets of GBM irrespective of COVID-19.


Asunto(s)
COVID-19 , Glioblastoma , Anciano , Enzima Convertidora de Angiotensina 2 , Carcinogénesis , Glioblastoma/genética , Humanos , Pandemias , Receptores de Coronavirus , SARS-CoV-2
16.
PLoS Pathog ; 17(9): e1009947, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1470670

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) preferentially infects and causes Kaposi's sarcoma (KS) in male patients. However, the biological mechanisms are largely unknown. This study was novel in confirming the extensive nuclear distribution of the androgen receptor (AR) and its co-localization with viral oncoprotein of latency-associated nuclear antigen in KS lesions, indicating a transcription way of AR in KS pathogenesis. The endogenous AR was also remarkably higher in KSHV-positive B cells than in KSHV-negative cells and responded to the ligand treatment of 5α-dihydrotestosterone (DHT), the agonist of AR. Then, the anti-AR antibody-based chromatin immunoprecipitation (ChIP)-associated sequencing was used to identify the target viral genes of AR, revealing that the AR bound to multiple regions of lytic genes in the KSHV genome. The highest peak was enriched in the core promoter sequence of polyadenylated nuclear RNA (PAN), and the physical interaction was verified by ChIP-polymerase chain reaction (PCR) and the electrophoretic mobility shift assay (EMSA). Consistently, male steroid treatment significantly transactivated the promoter activity of PAN in luciferase reporter assay, consequently leading to extensive lytic gene expression and KSHV production as determined by real-time quantitative PCR, and the deletion of nuclear localization signals of AR resulted in the loss of nuclear transport and transcriptional activity in the presence of androgen and thus impaired the expression of PAN RNA. Oncogenically, this study identified that the AR was a functional prerequisite for cell invasion, especially under the context of KSHV reactivation, through hijacking the PAN as a critical effector. Taken together, a novel mechanism from male sex steroids to viral noncoding RNA was identified, which might provide a clue to understanding the male propensity in KS.


Asunto(s)
ARN Mensajero/metabolismo , ARN Viral/metabolismo , Receptores Androgénicos/metabolismo , Sarcoma de Kaposi/metabolismo , Caracteres Sexuales , Carcinogénesis/metabolismo , Femenino , Herpesvirus Humano 8 , Humanos , Masculino , ARN no Traducido/metabolismo
17.
Sci Rep ; 11(1): 12948, 2021 06 21.
Artículo en Inglés | MEDLINE | ID: covidwho-1279894

RESUMEN

COVID 19 disease has become a global catastrophe over the past year that has claimed the lives of over two million people around the world. Despite the introduction of vaccines against the disease, there is still a long way to completely eradicate it. There are concerns about the complications following infection with SARS-CoV-2. This research aimed to evaluate the possible correlation between infection with SARS-CoV viruses and cancer in an in-silico study model. To do this, the relevent dataset was selected from GEO database. Identification of differentially expressed genes among defined groups including SARS-CoV, SARS-dORF6, SARS-BatSRBD, and H1N1 were screened where the |Log FC| ≥ 1and p < 0.05 were considered statistically significant. Later, the pathway enrichment analysis and gene ontology (GO) were used by Enrichr and Shiny GO databases. Evaluation with STRING online was applied to predict the functional interactions of proteins, followed by Cytoscape analysis to identify the master genes. Finally, analysis with GEPIA2 server was carried out to reveal the possible correlation between candidate genes and cancer development. The results showed that the main molecular function of up- and down-regulated genes was "double-stranded RNA binding" and actin-binding, respectively. STRING and Cytoscape analysis presented four genes, PTEN, CREB1, CASP3, and SMAD3 as the key genes involved in cancer development. According to TCGA database results, these four genes were up-regulated notably in pancreatic adenocarcinoma. Our findings suggest that pancreatic adenocarcinoma is the most probably malignancy happening after infection with SARS-CoV family.


Asunto(s)
Adenocarcinoma/etiología , COVID-19/complicaciones , Carcinogénesis/genética , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/complicaciones , Neoplasias Pancreáticas/etiología , SARS-CoV-2 , Síndrome Respiratorio Agudo Grave/complicaciones , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo , COVID-19/genética , COVID-19/metabolismo , COVID-19/virología , Caspasa 3/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Regulación de la Expresión Génica , Ontología de Genes , Humanos , Gripe Humana/genética , Gripe Humana/metabolismo , Gripe Humana/virología , Fosfohidrolasa PTEN/genética , Mapas de Interacción de Proteínas , Riesgo , Síndrome Respiratorio Agudo Grave/genética , Síndrome Respiratorio Agudo Grave/metabolismo , Síndrome Respiratorio Agudo Grave/virología , Transducción de Señal/genética , Proteína smad3/genética , Regulación hacia Arriba/genética
18.
Redox Biol ; 45: 102042, 2021 09.
Artículo en Inglés | MEDLINE | ID: covidwho-1275680

RESUMEN

Hypochlorous acid (HOCl) is the active oxidizing principle underlying drinking water disinfection, also delivered by numerous skin disinfectants and released by standard swimming pool chemicals used on a global scale, a topic of particular relevance in the context of the ongoing COVID-19 pandemic. However, the cutaneous consequences of human exposure to HOCl remain largely unknown, posing a major public health concern. Here, for the first time, we have profiled the HOCl-induced stress response in reconstructed human epidermis and SKH-1 hairless mouse skin. In addition, we have investigated the molecular consequences of solar simulated ultraviolet (UV) radiation and HOCl combinations, a procedure mimicking co-exposure experienced for example by recreational swimmers exposed to both HOCl (pool disinfectant) and UV (solar radiation). First, gene expression elicited by acute topical HOCl exposure was profiled in organotypic human reconstructed epidermis. Next, co-exposure studies (combining topical HOCl and UV) performed in SKH-1 hairless mouse skin revealed that the HOCl-induced cutaneous stress response blocks redox and inflammatory gene expression elicited by subsequent acute UV exposure (Nos2, Ptgs2, Hmox1, Srxn1), a finding consistent with emerging clinical evidence in support of a therapeutic role of topical HOCl formulations for the suppression of inflammatory skin conditions (e.g. atopic dermatitis, psoriasis). Likewise, in AP-1 transgenic SKH-1 luciferase-reporter mice, topical HOCl suppressed UV-induced inflammatory signaling assessed by bioluminescent imaging and gene expression analysis. In the SKH-1 high-risk mouse model of UV-induced human keratinocytic skin cancer, topical HOCl blocked tumorigenic progression and inflammatory gene expression (Ptgs2, Il19, Tlr4), confirmed by immunohistochemical analysis including 3-chloro-tyrosine-epitopes. These data illuminate the molecular consequences of HOCl-exposure in cutaneous organotypic and murine models assessing inflammatory gene expression and modulation of UV-induced carcinogenesis. If translatable to human skin these observations provide novel insights on molecular consequences of chlorination stress relevant to environmental exposure and therapeutic intervention.


Asunto(s)
COVID-19 , Neoplasias Cutáneas , Animales , Carcinogénesis , Expresión Génica , Humanos , Ácido Hipocloroso , Ratones , Ratones Transgénicos , Pandemias , SARS-CoV-2 , Piel , Rayos Ultravioleta/efectos adversos
19.
Virus Res ; 301: 198464, 2021 08.
Artículo en Inglés | MEDLINE | ID: covidwho-1246220

RESUMEN

The spread of SARS-CoV-2 and the increasing mortality rates of COVID-19 create an urgent need for treatments, which are currently lacking. Although vaccines have been approved by the FDA for emergency use in the U.S., patients will continue to require pharmacologic intervention to reduce morbidity and mortality as vaccine availability remains limited. The rise of new variants makes the development of therapeutic strategies even more crucial to combat the current pandemic and future outbreaks. Evidence from several studies suggests the host immune response to SARS-CoV-2 infection plays a critical role in disease pathogenesis. Consequently, host immune factors are becoming more recognized as potential biomarkers and therapeutic targets for COVID-19. To develop therapeutic strategies to combat current and future coronavirus outbreaks, understanding how the coronavirus hijacks the host immune system during and after the infection is crucial. In this study, we investigated immunological patterns or characteristics of the host immune response to SARS-CoV-2 infection that may contribute to the disease severity of COVID-19 patients. We analyzed large bulk RNASeq and single cell RNAseq data from COVID-19 patient samples to immunoprofile differentially expressed gene sets and analyzed pathways to identify human host protein targets. We observed an immunological profile of severe COVID-19 patients characterized by upregulated cytokines, interferon-induced proteins, and pronounced T cell lymphopenia, supporting findings by previous studies. We identified a number of host immune targets including PERK, PKR, TNF, NF-kB, and other key genes that modulate the significant pathways and genes identified in COVID-19 patients. Finally, we identified genes modulated by COVID-19 infection that are implicated in oncogenesis, including E2F transcription factors and RB1, suggesting a mechanism by which SARS-CoV-2 infection may contribute to oncogenesis. Further clinical investigation of these targets may lead to bonafide therapeutic strategies to treat the current COVID-19 pandemic and protect against future outbreaks and viral escape variants.


Asunto(s)
COVID-19/inmunología , Inmunidad , Pandemias , SARS-CoV-2/inmunología , COVID-19/epidemiología , COVID-19/virología , Carcinogénesis , Citocinas/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , SARS-CoV-2/genética , Regulación hacia Arriba , Tratamiento Farmacológico de COVID-19
20.
OMICS ; 25(6): 358-371, 2021 06.
Artículo en Inglés | MEDLINE | ID: covidwho-1243453

RESUMEN

About a tenth of all cancers are caused by viruses or associated with viral infection. Recent global events including the coronavirus disease-2019 (COVID-19) pandemic means that human encounter with viruses is increased. Cancer development in individuals with viral infection can take many years after infection, demonstrating that the involvement of viruses in cancer development is a long and complex process. This complexity emanates from individual genetic heterogeneity and the many steps involved in cancer development owing to viruses. The process of tumorigenesis is driven by the complex interaction between several viral factors and host factors leading to the creation of a tumor microenvironment (TME) that is ideal and promotes tumor formation. Viruses associated with human cancers ensure their survival and proliferation through activation of several cellular processes including inflammation, migration, and invasion, resistance to apoptosis and growth suppressors. In addition, most human oncoviruses evade immune detection and can activate signaling cascades including the PI3K-Akt-mTOR, Notch and Wnt pathways associated with enhanced proliferation and angiogenesis. This expert review examines and synthesizes the multiple biological factors related to oncoviruses, and the signaling cascades activated by these viruses contributing to viral oncogenesis. In particular, I examine and review the Epstein-Barr virus, human papillomaviruses, and Kaposi's sarcoma herpes virus in a context of cancer pathogenesis. I conclude with a future outlook on therapeutic targeting of the viruses and their associated oncogenic pathways within the TME. These anticancer strategies can be in the form of, but not limited to, antibodies and inhibitors.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Neoplasias/virología , Infecciones por Papillomavirus/virología , Infecciones por Retroviridae/virología , Retroviridae/fisiología , Sarcoma de Kaposi/virología , Infecciones Tumorales por Virus/virología , Alphapapillomavirus/fisiología , Carcinogénesis , Transformación Celular Viral , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/fisiología , Herpesvirus Humano 8/fisiología , Humanos , Terapia Molecular Dirigida , Neoplasias/patología , Neoplasias/terapia , Infecciones por Papillomavirus/patología , Infecciones por Retroviridae/patología , Sarcoma de Kaposi/patología , Transducción de Señal , Microambiente Tumoral , Infecciones Tumorales por Virus/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA