Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 191
Filtrar
1.
Microb Pathog ; 183: 106292, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37557930

RESUMO

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.


Assuntos
Infecções por Vírus Epstein-Barr , Neoplasias , Humanos , Infecções por Vírus Epstein-Barr/complicações , Herpesvirus Humano 4/genética , Vírus Oncogênicos/fisiologia , Hepacivirus
2.
J Cell Biochem ; 123(2): 161-182, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34520596

RESUMO

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.


Assuntos
Ubiquitina-Proteína Ligases/fisiologia , Proteínas Virais/fisiologia , Viroses/enzimologia , Replicação Viral/fisiologia , Transformação Celular Viral/fisiologia , Proteínas Culina/fisiologia , Endossomos/virologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Inflamação/enzimologia , Inflamação/virologia , Neoplasias/enzimologia , Neoplasias/virologia , Vírus Oncogênicos/fisiologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteínas com Motivo Tripartido/fisiologia , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Viroses/imunologia , Viroses/virologia , Replicação Viral/efeitos dos fármacos , Tratamento Farmacológico da COVID-19
4.
BMC Immunol ; 22(1): 65, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34583647

RESUMO

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.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/imunologia , Neoplasias/terapia , Vírus Oncogênicos/fisiologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T/imunologia , Viroses/imunologia , Linhagem Celular , Células Clonais , Citometria de Fluxo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/transplante
5.
Rev Med Virol ; 30(1): e2088, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31788897

RESUMO

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.


Assuntos
Transformação Celular Viral , Neoplasias/etiologia , Neoplasias/metabolismo , Vírus Oncogênicos/fisiologia , Transdução de Sinais , Fator de Transcrição AP-1/metabolismo , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/virologia , Animais , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Humanos , Neoplasias/patologia
6.
Rev Med Virol ; 30(1): e2085, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31743548

RESUMO

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.


Assuntos
Transformação Celular Viral , Suscetibilidade a Doenças , Histona Desacetilases/metabolismo , Interações Hospedeiro-Patógeno , Neoplasias/etiologia , Neoplasias/metabolismo , Animais , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Humanos , Neoplasias/patologia , Vírus Oncogênicos/fisiologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/virologia
7.
Semin Immunol ; 32: 14-24, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28602713

RESUMO

During the last few decades we have become accustomed to the idea that viruses can cause tumors. It is much less considered and discussed, however, that most people infected with oncoviruses will never develop cancer. Therefore, the genetic and environmental factors that tip the scales from clearance of viral infection to development of cancer are currently an area of active investigation. Microbiota has recently emerged as a potentially critical factor that would affect this balance by increasing or decreasing the ability of viral infection to promote carcinogenesis. In this review, we provide a model of microbiome contribution to the development of oncogenic viral infections and viral associated cancers, give examples of this process in human tumors, and describe the challenges that prevent progress in the field as well as their potential solutions.


Assuntos
Microbiota/imunologia , Neoplasias/imunologia , Vírus Oncogênicos/fisiologia , Viroses/imunologia , Animais , Carcinogênese , Interação Gene-Ambiente , Humanos , Neoplasias/complicações , Carga Viral , Viroses/complicações
8.
J Infect Dis ; 221(8): 1331-1341, 2020 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-31111897

RESUMO

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.


Assuntos
Coinfecção/microbiologia , Coinfecção/virologia , Boca/microbiologia , Boca/virologia , Vírus Oncogênicos/fisiologia , Ativação Viral/fisiologia , Adulto , Idoso , Linhagem Celular , Feminino , Infecções por HIV/microbiologia , Infecções por HIV/virologia , Infecções por Herpesviridae/microbiologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/patogenicidade , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , Vírus Oncogênicos/genética , Sarcoma de Kaposi/virologia , Staphylococcus aureus/patogenicidade , Latência Viral/fisiologia , Replicação Viral/genética , Adulto Jovem
9.
Rev Med Virol ; 29(6): e2076, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31407416

RESUMO

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.


Assuntos
Aneuploidia , Transformação Celular Viral , Vírus Oncogênicos/fisiologia , Animais , Humanos , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/virologia
10.
Biochemistry (Mosc) ; 85(10): 1191-1201, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33202204

RESUMO

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.


Assuntos
Apoptose , Carcinogênese , Vírus Oncogênicos/fisiologia , Infecções Tumorais por Vírus/virologia , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Humanos , Mitocôndrias/metabolismo , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Cancer Immunol Immunother ; 68(5): 849-859, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30465060

RESUMO

Cancer immunotherapy has greatly advanced in recent years. Most immunotherapeutic strategies are based on the use of immune checkpoint blockade to unleash antitumor immune responses or on the induction or adoptive transfer of immune effector cells. We aim to develop therapeutic vaccines based on recombinant Semliki Forest virus vectors to induce tumor-specific effector immune cells. In this review, we describe our ongoing work on SFV-based vaccines targeted against human papillomavirus- and hepatitis C virus-related infections and malignancies, focusing on design, delivery, combination strategies, preclinical efficacy and product development for a first-in-man clinical trial with an HPV-specific vaccine.


Assuntos
Alphavirus/genética , Vacinas Anticâncer/imunologia , Neoplasias/terapia , Vírus Oncogênicos/fisiologia , Viroses/terapia , Animais , Ensaios Clínicos como Assunto , Vetores Genéticos , Humanos , Imunização , Neoplasias/imunologia , Viroses/imunologia
12.
Rev Med Virol ; 28(2)2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29345394

RESUMO

Transforming growth factor-ß (TGF-ß) signaling pathway is a key network in cell signaling that controls vital processes such as proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and migration, thus acting as a double-edged sword in normal development and diseases, in particular organ fibrosis, vascular disorders, and cancer. Early in tumorigenesis, the pathway exerts anti-tumor effects through suppressing cell cycle and inducing apoptosis, while during late stages, it functions as a tumor promoter by enhancing tumor invasiveness and metastasis. This signaling pathway can be perturbed by environmental and genetic factors such as microbial interference and mutation, respectively. In this way, the present review describes the modulation of the TGF-ß pathway by oncogenic human viral pathogens and other viruses. The main mechanisms by which viruses interferes with TGF-ß signaling seems to be through (1) the alteration of either TGF-ß protein expression or activation, (2) the modulation of the TGF-ß receptors or SMADs factors (by interfering with their levels and functions), (3) the alteration of none-SMAD pathways, and (4) indirect interaction with the pathway by the modulation of transcriptional co-activator/repressor and regulators of the pathway. Given the axial role of this pathway in tumorigenesis, it can be regarded as an attractive target for cancer therapy. Hence, further investigations on this subject may represent molecular targets among either TGF-ß signaling molecules or viral factors for the treatment and management of viral infection consequences such as cancer.


Assuntos
Interações Hospedeiro-Patógeno , Neoplasias/etiologia , Neoplasias/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fenômenos Fisiológicos Virais , Animais , Transformação Celular Viral , Humanos , Vírus Oncogênicos/fisiologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/virologia
13.
Rev Med Virol ; 28(5): e1988, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29956408

RESUMO

The Notch pathway is a conserved signaling pathway and a form of direct cell-cell communication related to many biological processes during development and adulthood. Deregulation of the Notch pathway is involved in many diseases, including cancer. Almost 20% of all cancer cases have an infectious etiology, with viruses responsible for at least 1.5 million new cancer cases per year. Seven groups of viruses have been classified as oncogenic: hepatitis B and C viruses (HBV and HCV respectively), Epstein-Barr virus (EBV), Kaposi sarcoma-associated herpesvirus (KSHV), human T lymphotropic virus (HTLV-1), human papillomavirus (HPV), and Merkel cell polyomavirus (MCPyV). These viruses share the ability to manipulate a variety of cell pathways that are critical in proliferation and differentiation, leading to malignant transformation. Viral proteins interact directly or indirectly with different members of the Notch pathway, altering their normal function. This review focuses exclusively on the direct interactions of viral oncoproteins with Notch elements, providing a deeper understanding of the dual behavior of the Notch pathway as activator or suppressor of neoplasia in virus-related cancers.


Assuntos
Transformação Celular Neoplásica/metabolismo , Transformação Celular Viral , Vírus Oncogênicos/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Biomarcadores , Suscetibilidade a Doenças , Humanos , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Ligação Proteica , Receptores Notch/química
14.
Mol Cell Proteomics ; 16(4 suppl 1): S65-S74, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28104704

RESUMO

Oncogenic viruses are responsible for about 15% human cancers. This article explores the promise and challenges of viral proteomics in the study of the oncogenic human DNA viruses, HPV, McPyV, EBV and KSHV. These viruses have coevolved with their hosts and cause persistent infections. Each virus encodes oncoproteins that manipulate key cellular pathways to promote viral replication and evade the host immune response. Viral proteomics can identify cellular pathways perturbed by viral infection, identify cellular proteins that are crucial for viral persistence and oncogenesis, and identify important diagnostic and therapeutic targets.


Assuntos
Neoplasias/virologia , Proteínas Oncogênicas/metabolismo , Vírus Oncogênicos/fisiologia , Proteômica/métodos , Infecções por Vírus de DNA/imunologia , Infecções por Vírus de DNA/virologia , Interações Hospedeiro-Patógeno , Humanos , Vírus Oncogênicos/metabolismo , Proteínas Virais/metabolismo , Replicação Viral
16.
Immunol Cell Biol ; 95(4): 364-371, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27999434

RESUMO

Persistent viral infections are associated with the majority of human cancers where infectious agents have been recognized as the primary etiological agent. These viruses contribute to the malignant transformation of human cells either through the expression of oncogenic proteins or chronic inflammation. In spite of the high prevalence of these viral infections in humans, only a small proportion of these individuals who may have an underlying immune defect develop malignant disease. Furthermore, many of these viruses have evolved unique mechanisms to avoid the host immune system to successfully establish latent infection with limited gene expression. Technological advances in delineating the role of cellular immune responses in the control of viral infections and ability to rapidly expand these effector cells in vitro have provided an important platform for the development of novel immunotherapeutic strategies to treat virus-associated cancers. While autologous T cell therapies have provided promising results, development of 'off-the-shelf' third-party allogeneic virus-specific T cell therapies have emerged as powerful tools to treat many of the virus-associated diseases. It is anticipated that adoptive T cell therapy in combination with newly emerging immune checkpoint inhibitors and therapeutic vaccines will provide opportunities to successfully treat advanced metastatic virus-associated cancers which are currently not amenable to standard therapeutic strategies.


Assuntos
Imunoterapia Adotiva/métodos , Neoplasias/prevenção & controle , Vírus Oncogênicos/fisiologia , Linfócitos T/imunologia , Infecções Tumorais por Vírus/terapia , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/imunologia , Antígenos Virais/imunologia , Transformação Celular Neoplásica , Terapia Combinada , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Humanos , Evasão da Resposta Imune , Neoplasias/etiologia , Neoplasias/imunologia , Medicina de Precisão , Linfócitos T/transplante , Infecções Tumorais por Vírus/imunologia , Latência Viral
17.
Semin Cancer Biol ; 34: 70-80, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25837157

RESUMO

When stringent criteria have been used, the Epstein Barr virus (EBV), the Kaposi's sarcoma herpesvirus (KSHV), human immunodeficiency virus type 1 (HIV-1) and human hepatitis C virus (HCV) have been identified with sufficient evidence to be causative agents of non-Hodgkin's Lymphomas. Initially, single viral infection was considered fully responsible for the oncogenic properties of each virus, while it is now established that in many cases, multiple viral agents collaborate as cofactors in inducing lymphomas, especially in the presence of HIV-dependent immunodeficiency. Viruses cooperate by using their specific pathogenetic mechanisms in different combinations. The aim of this review is to describe the cooperation between different viruses in the development of lymphomas including the evidences supporting their pathogenetic role. Viral cooperation, a mechanism by which different viruses coinfecting human tissues have synergistic or regulatory effects on carcinogenesis, targets neoplastic B cells as well as cells of the microenvironment, such as reactive T-cells, B cells and macrophages, as well as non-immune cells such as endothelial cells, that contribute to tumor microenvironment. The most important viral genes involved in cooperation include HIV-1 tat and vpu, EBV LMP-1 and EBNA-2 and KSHV KIE2, Rta and LANA. Lymphomagenesis related to viral cooperation represents an interesting topic where microenvironmental abnormalities may be particularly relevant, particularly because antiviral targeted therapies and therapies producing the reconstitution of the immune system may constitute areas of interest aiming at improving the outcome of virus associated lymphomas. While the immune component of the lymphoma microenvironment can be easily studied by immunological and molecular techniques, the definition of the non-immune component of the lymphoma microenvironment is largely incomplete and may be the issue of future investigations. Understanding the pathogenetic role of viral infection in specific malignancies and defining microenvironmental abnormalities and mechanisms of viral carcinogenesis are important steps toward precise diagnosis and accurate treatment strategies for HIV-associated cancers.


Assuntos
Transformação Celular Viral , Linfoma/etiologia , Linfoma/patologia , Vírus Oncogênicos/fisiologia , Microambiente Tumoral , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/virologia , Animais , Coinfecção , Humanos , Vigilância Imunológica , Linfoma/diagnóstico , Linfoma/tratamento farmacológico , Linfoma/metabolismo , Transtornos Linfoproliferativos/etiologia , Transtornos Linfoproliferativos/metabolismo , Transtornos Linfoproliferativos/patologia , Técnicas de Diagnóstico Molecular , Transplante/efeitos adversos , Microambiente Tumoral/imunologia , Infecções Tumorais por Vírus/diagnóstico , Infecções Tumorais por Vírus/tratamento farmacológico
18.
J Gen Virol ; 97(11): 2973-2988, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27655063

RESUMO

Marek's disease virus, or Gallid herpesvirus 2 (GaHV-2), is an avian alphaherpesvirus that induces T-cell lymphoma in chickens. During transcriptomic studies of the RL region of the genome, we characterized the 7.5 kbp gene of the ERL lncRNA (edited repeat-long, long non-coding RNA), which may act as a natural antisense transcript (NAT) of the major GaHV-2 oncogene meq and of two of the three miRNA clusters. During infections in vivo and in vitro, we detected hyperediting of the ERL lncRNA that appeared to be directly correlated with ADAR1 expression levels. The ERL lncRNA was expressed equally during the lytic and latent phases of infection and during viral reactivation, but its hyperediting increased only during the lytic infection of chicken embryo fibroblasts. We also showed that chicken ADAR1 expression was controlled by the JAK/STAT IFN-response pathway, through an inducible promoter containing IFN-stimulated response elements that were functional during stimulation with IFN-α or poly(I:C). Like the human and murine miR-155-5p, the chicken gga-miR-155-5p and the GaHV-2 analogue mdv1-miR-M4-5p deregulated this pathway by targeting and repressing expression of suppressor of cytokine signalling 1, leading to the upregulation of ADAR1. Finally, we hypothesized that the natural antisense transcript role of the ERL lncRNA could be disrupted by its hyperediting, particularly during viral lytic replication, and that the observed deregulation of the innate immune system by mdv1-miR-M4-5p might contribute to the viral cycle.


Assuntos
Adenosina Desaminase/metabolismo , Herpesvirus Galináceo 2/genética , Doença de Marek/enzimologia , Doença de Marek/virologia , Vírus Oncogênicos/genética , RNA Longo não Codificante/genética , RNA Viral/genética , Adenosina Desaminase/genética , Animais , Galinhas , Fibroblastos/enzimologia , Fibroblastos/metabolismo , Fibroblastos/virologia , Regulação Viral da Expressão Gênica , Herpesvirus Galináceo 2/fisiologia , Doença de Marek/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Vírus Oncogênicos/fisiologia , Regiões Promotoras Genéticas , Edição de RNA , RNA Longo não Codificante/metabolismo , RNA Viral/metabolismo
19.
Rev Med Virol ; 25(5): 320-41, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26258805

RESUMO

MicroRNAs (miRNAs) function as key regulators in immune responses and cancer development. In the contexts of infection with oncogenic viruses, miRNAs are engaged in viral persistence, latency establishment and maintenance, and oncogenesis. In this review, we summarize the potential roles and mechanisms of viral and cellular miRNAs in the host-pathogen interactions during infection with selected tumor viruses and HIV, which include (i) repressing viral replication and facilitating latency establishment by targeting viral transcripts, (ii) evading innate and adaptive immune responses via toll-like receptors, RIG-I-like receptors, T-cell receptor, and B-cell receptor pathways by targeting signaling molecules such as TRAF6, IRAK1, IKKε, and MyD88, as well as downstream targets including regulatory cytokines such as tumor necrosis factor α, interferon γ, interleukin 10, and transforming growth factor ß, (iii) antagonizing intrinsic and extrinsic apoptosis pathways by targeting pro-apoptotic or anti-apoptotic gene transcripts such as the Bcl-2 family and caspase-3, (iv) modulating cell proliferation and survival through regulation of the Wnt, PI3K/Akt, Erk/MAPK, and Jak/STAT signaling pathways, as well as the signaling pathways triggered by viral oncoproteins such as Epstein-Barr Virus LMP1, by targeting Wnt-inhibiting factor 1, SHIP, pTEN, and SOCSs, and (v) regulating cell cycle progression by targeting cell cycle inhibitors such as p21/WAF1 and p27/KIP1. Further elucidation of the interaction between miRNAs and these key biological events will facilitate our understanding of the pathogenesis of viral latency and oncogenesis and may lead to the identification of miRNAs as novel targets for developing new therapeutic or preventive interventions.


Assuntos
Regulação da Expressão Gênica , HIV/imunologia , HIV/fisiologia , Interações Hospedeiro-Patógeno , MicroRNAs/metabolismo , Vírus Oncogênicos/imunologia , Vírus Oncogênicos/fisiologia , Carcinogênese , Humanos , Viroses/imunologia
20.
Arch Pharm (Weinheim) ; 349(2): 73-90, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26754591

RESUMO

Cancerous transformation comprises different events that are both genetic and epigenetic. The ultimate goal for such events is to maintain cell survival and proliferation. This transformation occurs as a consequence of different features such as environmental and genetic factors, as well as some types of infection. Many viral infections are considered to be causative agents of a number of different malignancies. To convert normal cells into cancerous cells, oncogenic viruses must function at the epigenetic level to communicate with their host cells. Oncogenic viruses encode certain epigenetic factors that lead to the immortality and proliferation of infected cells. The epigenetic effectors produced by oncogenic viruses constitute appealing targets to prevent and treat malignant diseases caused by these viruses. In this review, we highlight the importance of epigenetic reprogramming for virus-induced oncogenesis, with special emphasis on viral epigenetic oncoproteins as therapeutic targets. The discovery of molecular components that target epigenetic pathways, especially viral factors, is also discussed.


Assuntos
Epigênese Genética , Neoplasias/virologia , Vírus Oncogênicos/fisiologia , Infecções Tumorais por Vírus/virologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antivirais/farmacologia , Antivirais/uso terapêutico , Transformação Celular Viral , Humanos , Neoplasias/prevenção & controle , Vírus Oncogênicos/genética , Vírus Oncogênicos/patogenicidade , Infecções Tumorais por Vírus/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA