Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1263: 45-54, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32588322

RESUMO

Oncomodulatory viruses can affect the tumor microenvironment (TME) by triggering inflammation, suppressing apoptosis, initiating angiogenesis, altering tumor metabolism, and stimulating tumor cell signaling pathways, leading to tumor growth, proliferation, and invasion. The higher incidence of malignancies among people with HIV (PWH), despite the widespread use of antiretroviral therapy (ART), suggests a more complex relation than HIV-associated immune deregulation. Viral cooperation can have synergistic effect on tumorigenesis. The most relevant oncogenes involved in viral cooperation include the HIV-1-related Tat and Vpu genes, EBV LMP-1 and EBNA-2 genes, and Kaposi's sarcoma herpesvirus (KSHV) KIE2, Rta, and LANA genes. The TME in HIV-related malignancies is highly angiogenic and characterized by high microvessel density compared to sporadic cases. Tat protein, found in patients with HIV infection regardless of their immune status, has been widely implicated in the increased angiogenesis and has been a target of interest for therapeutic strategies. Similarly, HIV-1 matrix protein p17 can be detected in the plasma and tissues of PWH, including those treated with ART. Studies have found that p17 can cause dysregulation of the biological activity of different immune cells, is involved in aberrant angiogenesis, and exhibits an IL8 chemokine activity, activating multiple intracellular signaling pathways, promoting angiogenic responses in endothelial cells, and forming capillary like structures. In addition, several studies have demonstrated difference in the cellular immune components within the TME in patients with or without HIV infection, as well as cases in pre- and post-ART era. In this chapter, we review the existing literature about the role tumor microenvironment plays in the pathogenesis of HIV-related malignancies. Understanding the functions of each component of the TME and determining how these cellular and noncellular components contribute to tumorigenesis will impact the advancement of interventions and treatment in clinical oncology among PWH.


Assuntos
HIV/patogenicidade , Sarcoma de Kaposi/virologia , Microambiente Tumoral , Infecções por HIV/virologia , Humanos , Oncogenes
2.
Cancers (Basel) ; 14(5)2022 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-35267456

RESUMO

Human cytomegalovirus (HCMV) infects 40-70% of adults in developed countries. Detection of HCMV DNA and/or proteins in breast tumors varies considerably, ranging from 0-100%. In this study, nested PCR to detect HCMV glycoprotein B (gB) DNA in breast tumors was shown to be sensitive and specific in contrast to the detection of DNA for immediate early genes. HCMV gB DNA was detected in 18.4% of 136 breast tumors while 62.8% of 94 breast cancer patients were seropositive for HCMV. mRNA for the HCMV immediate early gene was not detected in any sample, suggesting viral latency in breast tumors. HCMV seropositivity was positively correlated with age, body mass index and menopause. Patients who were HCMV seropositive or had HCMV DNA in their tumors were 5.61 (CI 1.77-15.67, p = 0.003) or 5.27 (CI 1.09-28.75, p = 0.039) times more likely to develop Stage IV metastatic tumors, respectively. Patients with HCMV DNA in tumors experienced reduced relapse-free survival (p = 0.042). Being both seropositive with HCMV DNA-positive tumors was associated with vascular involvement and metastasis. We conclude that determining the seropositivity for HCMV and detection of HCMV gB DNA in the breast tumors could identify breast cancer patients more likely to develop metastatic cancer and warrant special treatment.

3.
Virus Res ; 276: 197829, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31790777

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive form of primary human gliomas. While chemotherapy using the DNA alkylating agent temozolomide (TMZ) is a first line treatment for GBMs, the development of resistance to TMZ is a common limitation to successful treatment. Human Cytomegalovirus (HCMV) is a ubiquitous ß-herpesvirus that establishes a lifelong infection latent infection in host haematopoetic cells, where lytic replication of the virus is silenced. HCMV can also establish a persistent infection in hosts, where low levels of virus are lytically produced. Furthermore, multiple studies have identified HCMV DNA and/or proteins in human GBM samples, and have shown that acute infection with HCMV confers a glioblastoma stem cell (GSC) phenotype, further supporting an oncomodulatory role for HCMV in GBM progression and severity. In this current study, we examined the long-term effects of HCMV persistence to cell viability, cell proliferation, and the development of TMZ resistance over time using a glioblastoma cell line known as LN-229. Persistent HCMV infections were established and maintained in this cell line for 30 weeks without the addition of new virus. Here, we report that HCMV persistence in this cell line resulted in increased cell viability, increased cell proliferation, and a marked resistance to the DNA alkylating agent, TMZ, over time, suggesting that low levels of lytically replicating HCMV could contribute to tumor progression in GBM.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Infecções por Citomegalovirus/complicações , Citomegalovirus/fisiologia , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/etiologia , Temozolomida/farmacologia , Antineoplásicos Alquilantes/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Infecções por Citomegalovirus/virologia , Glioblastoma/tratamento farmacológico , Humanos , Temozolomida/uso terapêutico
4.
Cancer Lett ; 384: 79-85, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-27777041

RESUMO

Glioblastoma multiforme (GBM) is the most aggressive neoplastic brain tumor in humans with a median survival of less than 2 years. It is therefore critical to understand the mechanism of glioma progression and to identify future targets for intervention. We investigate the mechanisms of cytomegalovirus as an oncomodulatory agent implicated in glioma progression, as well as immunosuppression. This review provides a comprehensive evaluation of recent investigative developments concerning the role of CMV in cellular processes during glioma growth. The manners in which CMV and its viral products interact with regulatory cellular signaling pathways in the host are of primary interest. Here, we examine some of the most significant oncomodulatory effects that CMV can confer in brain tumors, including the inhibition of apoptosis and promoting the growth of glioma stem cells, which are tightly linked to tumor survival and recurrence.


Assuntos
Neoplasias Encefálicas/virologia , Transformação Celular Viral , Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Glioma/virologia , Infecções Tumorais por Vírus/virologia , Animais , Apoptose , Neoplasias Encefálicas/epidemiologia , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/patologia , Ciclo Celular , Proliferação de Células , Citomegalovirus/imunologia , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/metabolismo , Progressão da Doença , Glioma/epidemiologia , Glioma/imunologia , Glioma/patologia , Interações Hospedeiro-Patógeno , Humanos , Mediadores da Inflamação/metabolismo , Evasão Tumoral , Infecções Tumorais por Vírus/epidemiologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/metabolismo
5.
Front Oncol ; 5: 61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25853089

RESUMO

The hypothesis that cytomegalovirus (CMV) modulates cancer is evolving. Originally discovered in glioblastoma in 2002, the number of cancers, where intratumoral CMV antigen is detected, has increased in recent years suggesting that CMV actively affects the pathobiology of certain tumors. These findings are controversial as several groups have also reported inability to replicate these results. Regardless, several clinical trials for glioblastoma are underway or have been completed that target intratumoral CMV with anti-viral drugs or immunotherapy. Therefore, a better understanding of the possible pathobiology of CMV in cancer needs to be ascertained. We have developed genetic, syngeneic, and orthotopic malignant glioma mouse models to study the role of CMV in cancer development and progression. These models recapitulate for the most part intratumoral CMV expression as seen in human tumors. Additionally, we discovered that CMV infection in Trp53(-/+) mice promotes pleomorphic rhabdomyosarcomas. These mouse models are not only a vehicle for studying pathobiology of the viral-tumor interaction but also a platform for developing and testing cancer therapeutics.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA