Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 189
Filtrar
1.
Viruses ; 16(3)2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38543781

RESUMEN

Approximately 12% of human cancers worldwide are associated with infectious agents, which are classified by the International Agency for Research on Cancer (IARC) as Group 1 within the agents that are carcinogenic to humans. Most of these agents are viruses. Group 1 oncogenic viruses include hepatitis C virus, hepatitis B virus (HBV), human T-cell lymphotropic virus type 1, Epstein-Barr virus, Kaposi sarcoma-associated herpesvirus, human immunodeficiency virus-1 and high-risk human papillomaviruses (HPVs). In addition, some human polyomaviruses are suspected of inducing cancer prevalently in hosts with impaired immune responses. Merkel cell polyomavirus has been associated with Merkel cell carcinoma and included by the IARC in Group 2A (i.e., probably carcinogenic to humans). Linking viruses to human cancers has allowed for the development of diagnostic, prophylactic and therapeutic measures. Vaccination significantly reduced tumours induced by two oncogenic viruses as follows: HBV and HPV. Herein, we focus on mucosal alpha HPVs, which are responsible for the highest number of cancer cases due to tumour viruses and against which effective prevention strategies have been developed to reduce the global burden of HPV-related cancers.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Neoplasias , Infecciones por Papillomavirus , Virus , Humanos , Virus Oncogénicos/fisiología , Virus del Papiloma Humano , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/epidemiología , Infecciones por Papillomavirus/prevención & control , Herpesvirus Humano 4 , Carcinogénesis , Virus de la Hepatitis B
2.
Microb Pathog ; 183: 106292, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37557930

RESUMEN

Cancer is a serious public health problem globally. Many human cancers are induced by viruses. Understanding of the mechanisms by which oncogenic (tumorigenic) viruses induce cancer is essential in the prevention and control of cancer. This review covers comprehensive characteristics and molecular mechanisms of the main virus-attributed cancers caused by human papillomavirus, hepatitis B virus, hepatitis C virus, Epstein-Barr virus, human herpesvirus type 8, human T-cell lymphotropic virus, human polyomaviruses, Merkel cell polyomavirus, and HIV. Oncogenic viruses employ biological processes to replicate and avoid detection by host cell immune systems. Tumorigenic infectious agents activate oncogenes in a variety of ways, allowing the pathogen to block host tumour suppressor proteins, inhibit apoptosis, enhance cell proliferation, and promote invasion of host cells. Furthermore, this review assesses many pathways of viruses linked to cancer, including host cellular communication perturbation, DNA damage mechanisms, immunity, and microRNA targets that promote the beginning and progression of cancer. The current cancer prevention is primarily focused on non-communicable diseases, but infection-attributable cancer also needs attention to significantly reduce the rising cancer burden and related deaths.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Neoplasias , Humanos , Infecciones por Virus de Epstein-Barr/complicaciones , Herpesvirus Humano 4/genética , Virus Oncogénicos/fisiología , Hepacivirus
3.
J Cell Biochem ; 123(2): 161-182, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34520596

RESUMEN

Viruses are known to cause various diseases in human and also infect other species such as animal plants, fungi, and bacteria. Replication of viruses depends upon their interaction with hosts. Human cells are prone to such unwanted viral infections. Disintegration and reconstitution require host machinery and various macromolecules like DNA, RNA, and proteins are invaded by viral particles. E3 ubiquitin ligases are known for their specific function, that is, recognition of their respective substrates for intracellular degradation. Still, we do not understand how ubiquitin proteasome system-based enzymes E3 ubiquitin ligases do their functional interaction with different viruses. Whether E3 ubiquitin ligases help in the elimination of viral components or viruses utilize their molecular capabilities in their intracellular propagation is not clear. The first time our current article comprehends fundamental concepts and new insights on the different viruses and their interaction with various E3 Ubiquitin Ligases. In this review, we highlight the molecular pathomechanism of viruses linked with E3 Ubiquitin Ligases dependent mechanisms. An enhanced understanding of E3 Ubiquitin Ligase-mediated removal of viral proteins may open new therapeutic strategies against viral infections.


Asunto(s)
Ubiquitina-Proteína Ligasas/fisiología , Proteínas Virales/fisiología , Virosis/enzimología , Replicación Viral/fisiología , Transformación Celular Viral/fisiología , Proteínas Cullin/fisiología , Endosomas/virología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Inflamación/enzimología , Inflamación/virología , Neoplasias/enzimología , Neoplasias/virología , Virus Oncogénicos/fisiología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteínas de Motivos Tripartitos/fisiología , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Virosis/inmunología , Virosis/virología , Replicación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
4.
BMC Immunol ; 22(1): 65, 2021 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-34583647

RESUMEN

Adoptive transfer of T cells genetically engineered with a T cell receptor (TCR) is a promising cancer treatment modality that requires the identification of TCRs with good characteristics. Most T cell cloning methods involve a stringent singularization process, which necessitates either tedious hands-on operations or high cost. We present an efficient and nonstringent cloning approach based on existing techniques. We hypothesize that after elimination of most nonspecific T cells, a clonotype with high quality could outcompete other clonotypes and finally form a predominant population. This TCR identification method can be used to clone virus-specific TCRs efficiently from cancer patients and is easily adoptable by any laboratory.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Neoplasias/inmunología , Neoplasias/terapia , Virus Oncogénicos/fisiología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Virosis/inmunología , Línea Celular , Células Clonales , Citometría de Flujo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/trasplante
5.
mBio ; 12(4): e0170621, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34425696

RESUMEN

The methylation of RNA at the N6 position of adenosine (m6A) orchestrates multiple biological processes to control development, differentiation, and cell cycle, as well as various aspects of the virus life cycle. How the m6A RNA modification pathway is regulated to finely tune these processes remains poorly understood. Here, we discovered the m6A reader YTHDF2 as a caspase substrate via proteome-wide prediction, followed by in vitro and in vivo validations. We further demonstrated that cleavage-resistant YTHDF2 blocks, while cleavage-mimicking YTHDF2 fragments promote, the replication of a common human oncogenic virus, Epstein-Barr virus (EBV). Intriguingly, our study revealed a feedback regulation between YTHDF2 and caspase-8 via m6A modification of CASP8 mRNA and YTHDF2 cleavage during EBV replication. Further, we discovered that caspases cleave multiple components within the m6A RNA modification pathway to benefit EBV replication. Our study establishes that caspase disarming of the m6A RNA modification machinery fosters EBV replication. IMPORTANCE The discovery of an N6-methyladenosine (m6A) RNA modification pathway has fundamentally altered our understanding of the central dogma of molecular biology. This pathway is controlled by methyltransferases (writers), demethylases (erasers), and specific m6A binding proteins (readers). Emerging studies have linked the m6A RNA modification pathway to the life cycle of various viruses. However, very little is known regarding how this pathway is subverted to benefit viral replication. In this study, we established an unexpected linkage between cellular caspases and the m6A modification pathway, which is critical to drive the reactivation of a common tumor virus, Epstein-Barr virus (EBV).


Asunto(s)
Adenosina/metabolismo , Caspasas/metabolismo , Herpesvirus Humano 4/genética , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , Replicación Viral/genética , Adenosina/química , Caspasas/genética , Línea Celular , Infecciones por Virus de Epstein-Barr , Herpesvirus Humano 4/fisiología , Humanos , Metilación , Virus Oncogénicos/genética , Virus Oncogénicos/fisiología
7.
Commun Biol ; 4(1): 682, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083759

RESUMEN

An outbreak of the novel coronavirus SARS-CoV-2, the causative agent of Coronavirus Disease-2019 (COVID-19), a respiratory disease, has infected almost one hundred million people since the end of 2019, killed over two million, and caused worldwide social and economic disruption. Because the mechanisms of SARS-CoV-2 infection of host cells and its pathogenesis remain largely unclear, there are currently no antiviral drugs with proven efficacy. Besides severe respiratory and systematic symptoms, several comorbidities increase risk of fatal disease outcome. Therefore, it is required to investigate the impacts of COVID-19 on pre-existing diseases of patients, such as cancer and other infectious diseases. In the current study, we report that SARS-CoV-2 encoded proteins and some currently used anti-COVID-19 drugs are able to induce lytic reactivation of Kaposi's sarcoma-associated herpesvirus (KSHV), one of major human oncogenic viruses, through manipulation of intracellular signaling pathways. Our data indicate that those KSHV + patients especially in endemic areas exposure to COVID-19 or undergoing the treatment may have increased risks to develop virus-associated cancers, even after they have fully recovered from COVID-19.


Asunto(s)
Antivirales/farmacología , COVID-19/complicaciones , Herpesvirus Humano 8/fisiología , SARS-CoV-2/fisiología , Sarcoma de Kaposi/etiología , Activación Viral , Azitromicina/farmacología , Benzamidinas/farmacología , Línea Celular , Guanidinas/farmacología , Infecciones por Herpesviridae/inducido químicamente , Infecciones por Herpesviridae/etiología , Herpesvirus Humano 8/efectos de los fármacos , Humanos , Virus Oncogénicos/efectos de los fármacos , Virus Oncogénicos/fisiología , SARS-CoV-2/efectos de los fármacos , Sarcoma de Kaposi/inducido químicamente , Proteínas Virales/metabolismo , Activación Viral/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
8.
Biochemistry (Mosc) ; 85(10): 1191-1201, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33202204

RESUMEN

The disruption of apoptotic cell death process is closely associated with the etiology of various diseases, including cancer. Permanent viral infections can cause different types of cancers. Oncogenic viruses manipulate both external and internal apoptosis pathways, and inhibit the activity of proapoptotic proteins and signaling pathways, which facilitates carcinogenesis. Ineffective immune surveillance or immune response suppression can induce uncontrolled virus propagation and host cell proliferation. In this review, we discuss current data that provide insights into mechanisms of apoptotic death suppression by viruses and their role in oncogenesis.


Asunto(s)
Apoptosis , Carcinogénesis , Virus Oncogénicos/fisiología , Infecciones Tumorales por Virus/virología , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
9.
Life Sci ; 258: 118134, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32717272

RESUMEN

Cancer can arise due to mutations in numerous pathways present in our body and thus has many alternatives for getting aggravated. Due to this attribute, it gets difficult to treat cancer patients with monotherapy alone and has a risk of not being eliminated to the full extent. This necessitates the introduction of combinatorial therapy as it employs cancer treatment using more than one method and shows a greater success rate. Combinatorial therapy involves a complementary combination of two different therapies like a combination of radio and immunotherapy or a combination of drugs that can target more than one pathway of cancer formation like combining CDK targeting drugs with Growth factors targeting drugs. In this review, we discuss the various aspects of cancer which include, its causes; four regulatory mechanisms namely: apoptosis, cyclin-dependent kinases, tumor suppressor genes, and growth factors; some of the pathways involved; treatment: monotherapy and combinatorial therapy and combinatorial drug formulation in chemotherapy. The present review gives a holistic account of the different mechanisms of therapies and also drug combinations that may serve to not only complement the monotherapy but can also surpass the resistance against monotherapy agents.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias/tratamiento farmacológico , Progresión de la Enfermedad , Composición de Medicamentos , Genes Supresores de Tumor , Humanos , Neoplasias/genética , Neoplasias/patología , Virus Oncogénicos/fisiología
10.
J Infect Dis ; 221(8): 1331-1341, 2020 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-31111897

RESUMEN

Collectively, viruses are the principal cause of cancers arising in patients with immune dysfunction, including human immunodeficiency virus (HIV)-positive patients. Kaposi sarcoma (KS) etiologically linked to Kaposi sarcoma-associated herpesvirus (KSHV) continues to be the most common AIDS-associated tumor. The involvement of the oral cavity represents one of the most common clinical manifestations of this tumor. HIV infection incurs an increased risk among individuals with periodontal diseases and oral carriage of a variety of pathogenic bacteria. However, whether interactions involving periodontal bacteria and oncogenic viruses in the local environment facilitate replication or maintenance of these viruses in the oral cavity of HIV-positive patients remain largely unknown. We previously showed that pathogen-associated molecular patterns (PAMPs) from specific periodontal bacteria promoted KSHV entry into oral cells and subsequent establishment of latency. In the current study, we demonstrate that Staphylococcus aureus, one of common pathogens causing infection in HIV-positive patients, and its PAMPs can effectively induce KSHV lytic reactivation from infected oral cells, through the Toll-like receptor reactive oxygen species and cyclin D1-Dicer-viral microRNA axis. This investigation provides further clinical evidence about the relevance of coinfection due to these 2 pathogens in the oral cavities of a cohort HIV-positive patients and reveals novel mechanisms through which these coinfecting pathogens potentially promote virus-associated cancer development in the unique niche of immunocompromised patients.


Asunto(s)
Coinfección/microbiología , Coinfección/virología , Boca/microbiología , Boca/virología , Virus Oncogénicos/fisiología , Activación Viral/fisiología , Adulto , Anciano , Línea Celular , Femenino , Infecciones por VIH/microbiología , Infecciones por VIH/virología , Infecciones por Herpesviridae/microbiología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidad , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Virus Oncogénicos/genética , Sarcoma de Kaposi/virología , Staphylococcus aureus/patogenicidad , Latencia del Virus/fisiología , Replicación Viral/genética , Adulto Joven
11.
Rev Med Virol ; 30(1): e2088, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31788897

RESUMEN

Cancer progression is critically associated with modulation of host cell signaling pathways. Activator protein-1 (AP-1) signaling is one such pathway whose deregulation renders the host more susceptible to cancer development. Oncogenic viruses, including hepatitis B virus, hepatitis C virus, human papilloma virus, Epstein-Barr virus, human T-cell lymphotropic virus type 1, and Kaposi's sarcoma-associated herpes virus, are common causes of cancer. This review discusses how these oncoviruses by acting through various aspects of the host cell signaling machinery such as the AP-1 pathway might affect oncoviral tumorigenesis, replication, and pathogenesis. The review also briefly considers how the pathway might be targeted during infections with these oncogenic viruses.


Asunto(s)
Transformación Celular Viral , Neoplasias/etiología , Neoplasias/metabolismo , Virus Oncogénicos/fisiología , Transducción de Señal , Factor de Transcripción AP-1/metabolismo , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/virología , Animales , Susceptibilidad a Enfermedades , Interacciones Huésped-Patógeno , Humanos , Neoplasias/patología
12.
Rev Med Virol ; 30(1): e2085, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31743548

RESUMEN

Oncogenic viruses are one of the most important causes of cancer worldwide. The pathogens contribute to the establishment of human malignancies by affecting various cellular events. Epigenetic mechanisms, such as histone modification methylation/demethylation, are one of the most critical events manipulated by oncogenic viruses to drive tumorigenesis. Histone modifications are mediated by histone acetylation and deacetylation, regulated by histone acetyltransferases (HATs) and histone deacetylases (HDACs), respectively. Dysregulation of HDACs activity affects viral tumorigenesis in several ways, such as manipulating tumor suppressor and viral gene expression. The present review aims to describe the vital interactions between both cancer-caused/associated viruses and the HDAC machinery, particularly by focusing on those viruses involved in gastrointestinal tumors, as some of the most common viral-mediated cancers.


Asunto(s)
Transformación Celular Viral , Susceptibilidad a Enfermedades , Histona Desacetilasas/metabolismo , Interacciones Huésped-Patógeno , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Regulación de la Expresión Génica , Histona Acetiltransferasas/metabolismo , Humanos , Neoplasias/patología , Virus Oncogénicos/fisiología , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/virología
13.
Curr Opin Virol ; 39: 60-69, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31766001

RESUMEN

Oncogenic viruses, like all viruses, relies on host metabolism to provide the metabolites and energy needed for virus replication. Many DNA tumor viruses and retroviruses will reprogram metabolism during infection. Additionally, some viral oncogenes may alter metabolism independent of virus replication. Virus infection and cancer development share many similarities regarding metabolic reprogramming as both processes demand increased metabolic activity to produce biomass: cell proliferation in the case of cancer and virion production in the case of infection. This review discusses the parallels in metabolic reprogramming between human oncogenic viruses and oncogenesis.


Asunto(s)
Carcinogénesis , Reprogramación Celular , Redes y Vías Metabólicas , Virus Oncogénicos/fisiología , Biomasa , Proliferación Celular , Hepacivirus/fisiología , Virus de la Hepatitis B/fisiología , Herpesviridae/fisiología , Humanos , Poliomavirus de Células de Merkel/fisiología , Neoplasias/metabolismo , Papillomaviridae/fisiología , Retroviridae , Virión/metabolismo , Replicación Viral
14.
Viruses ; 11(11)2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31739606

RESUMEN

Jaagsiekte sheep retrovirus (JSRV) and enzootic nasal tumor virus (ENTV) are small-ruminant betaretroviruses that share high nucleotide and amino acid identity, utilize the same cellular receptor, hyaluronoglucosaminidase 2 (Hyal2) for entry, and transform tissues with their envelope (Env) glycoprotein; yet, they target discrete regions of the respiratory tract-the lung and nose, respectively. This distinct tissue selectivity makes them ideal tools with which to study the pathogenesis of betaretroviruses. To uncover the genetic determinants of tropism, we constructed JSRV-ENTV chimeric viruses and produced lentivectors pseudotyped with the Env proteins from JSRV (Jenv) and ENTV (Eenv). Through the transduction and infection of lung and nasal turbinate tissue slices, we observed that Hyal2 expression levels strongly influence ENTV entry, but that the long terminal repeat (LTR) promoters of these viruses are likely responsible for tissue-specificity. Furthermore, we show evidence of ENTV Env expression in chondrocytes within ENTV-infected nasal turbinate tissue, where Hyal2 is highly expressed. Our work suggests that the unique tissue tropism of JSRV and ENTV stems from the combined effort of the envelope glycoprotein-receptor interactions and the LTR and provides new insight into the pathogenesis of ENTV.


Asunto(s)
Productos del Gen env/genética , Retrovirus Ovino Jaagsiekte/fisiología , Virus Oncogénicos/fisiología , Adenomatosis Pulmonar Ovina/virología , Secuencias Repetidas Terminales , Infecciones Tumorales por Virus/virología , Tropismo Viral , Animales , Línea Celular , Orden Génico , Genoma Viral , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Virus Reordenados/genética , Ovinos
15.
Rev Med Virol ; 29(6): e2076, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31407416

RESUMEN

Seven oncogenic viruses are known for tumorigenesis and contribute to 12% of all human cancers. The oncogenic factors, the target tissue, and pathology of cancer vary among these viruses with several mechanisms proposed for the initiation and development of cancer. Aneuploidy in cells is associated with anomalies in chromosome number that can be a hallmark of cancer, a disease defined by expanded proliferative potential. In this review, we summarize the different mechanisms of aneuploidy and furthermore discuss recent findings of the role of viral oncoproteins in inducing cellular aneuploidy that might facilitate tumorigenesis. Improved understanding of viral oncogenesis may help to find new strategies for controlling virus-associated cancers.


Asunto(s)
Aneuploidia , Transformación Celular Viral , Virus Oncogénicos/fisiología , Animales , Humanos , Neoplasias/etiología , Neoplasias/metabolismo , Neoplasias/patología , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/virología
17.
Crit Rev Oncol Hematol ; 137: 108-114, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-31014506

RESUMEN

Human cancers attributed to viral infections represent a growing proportion of the global cancer burden, with these types of cancers being the leading cause of morbidity and mortality in some regions. The concept that viruses play a causal role in human cancers is not new, but the mechanism thereof, while well described for some viruses, still remains elusive and complex for others, especially in the case of HIV-associated B-cell derived cancers. In the last decade, compelling evidence has demonstrated that cellular microRNAs are deregulated in cancers, with an increasing number of studies identifying microRNAs as potential biomarkers for human cancer diagnosis, prognosis and therapeutic targets or tools. Recent research demonstrates that viruses and viral components manipulate host microRNA expressions to their advantage, and the emerging picture suggests that the virus/microRNA pathway interaction is defined by a plethora of complex mechanisms. In this review, we highlight the current knowledge on virus/microRNA pathway interactions in the context of cancer and provide new insights on HIV as an oncogenic virus.


Asunto(s)
VIH/fisiología , MicroARNs/genética , Neoplasias/genética , Neoplasias/virología , Virus Oncogénicos/fisiología , Animales , Carcinogénesis , Humanos , MicroARNs/biosíntesis , Pronóstico
18.
Philos Trans R Soc Lond B Biol Sci ; 374(1773): 20190041, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-30955496

RESUMEN

One out of 10 cancers is estimated to arise from infections by a handful of oncogenic viruses. These infectious cancers constitute an opportunity for primary prevention through immunization against the viral infection, for early screening through molecular detection of the infectious agent, and potentially for specific treatments, by targeting the virus as a marker of cancer cells. Accomplishing these objectives will require a detailed understanding of the natural history of infections, the mechanisms by which the viruses contribute to disease, the mutual adaptation of viruses and hosts, and the possible viral evolution in the absence and in the presence of the public health interventions conceived to target them. This issue showcases the current developments in experimental tissue-like and animal systems, mathematical models and evolutionary approaches to understand DNA oncoviruses. Our global aim is to provide proximate explanations to the present-day interface and interactions between virus and host, as well as ultimate explanations about the adaptive value of these interactions and about the evolutionary pathways that have led to the current malignant phenotype of oncoviral infections. This article is part of the theme issue 'Silent cancer agents: multi-disciplinary modelling of human DNA oncoviruses'.


Asunto(s)
Infecciones por Virus ADN/virología , Virus ADN/patogenicidad , Virus Oncogénicos/patogenicidad , Infecciones Tumorales por Virus/virología , Animales , Virus ADN/fisiología , Evolución Molecular , Humanos , Virus Oncogénicos/fisiología , Virulencia
19.
Philos Trans R Soc Lond B Biol Sci ; 374(1773): 20180304, 2019 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-30955500

RESUMEN

Most known oncogenic viruses of humans use DNA as their genomic material. Research over the past quarter century has revealed that their oncogenicity results largely from direct interference with barriers to oncogenesis. In contrast to viruses that have been accepted causes of particular cancers, candidate viral causes tend to have fewer viral than cellular genomes in the tumours. These low viral loads have caused researchers to conclude that the associated viruses are not primary causes of the associated cancers. Consideration of differential survival, reproduction and infiltration of cells in a tumour suggest, however, that viral loads could be low even when viruses are primary causes of cancer. Resolution of this issue has important implications for human health because medical research tends to be effective at preventing and controlling infectious diseases. Mathematical models may clarify the problem and help guide future research by assessing whether low viral loads are likely outcomes of the differential survival, reproduction, and infiltration of cells in a tumour and, more generally, the extent to which viruses contribute to cancer. This article is part of the theme issue 'Silent cancer agents: multi-disciplinary modelling of human DNA oncoviruses'.


Asunto(s)
Evolución Biológica , Carcinogénesis , Neoplasias/virología , Virus Oncogénicos/fisiología , Humanos
20.
Cell Rep ; 26(13): 3657-3671.e5, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917319

RESUMEN

Aurora kinase B (AURKB), a central regulator of chromosome segregation and cytokinesis, is aberrantly expressed in various cancer cells. However, the relationship of AURKB and oncogenic viruses in cancer progression remains unclear. Here, we reveal that N-cleaved isoforms of AURKB exist in several oncovirus-associated tumor cells and patient cancer tissues, including Kaposi's sarcoma-associated herpesvirus (KSHV), Epstein-Barr virus (EBV), and human papillomavirus virus (HPV). Mechanistically, in KSHV-infected tumor cells, the latent viral antigen LANA cleaves AURKB at Asp76 in a serine protease-dependent manner. The N'-AURKB relocalizes to the spindle pole and promotes the metaphase-to-telophase transition in mitotic cells. Introduction of N'-AURKB but not C'-AURKB promotes colony formation and malignant growth of tumor cells in vitro and in vivo using a murine xenograft model. Altogether, our findings uncover a proteolytic cleavage mechanism by which oncoviruses induce cancer cell segregation and tumorigenesis.


Asunto(s)
Aurora Quinasa B/metabolismo , Carcinogénesis , Neoplasias/virología , Virus Oncogénicos/fisiología , Adulto , Anciano , Animales , Segregación Cromosómica , Femenino , Células HEK293 , Células HeLa , Herpesvirus Humano 8/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Persona de Mediana Edad , Mitosis , Neoplasias/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA