Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EBioMedicine ; 80: 104056, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35596973

RESUMEN

BACKGROUND: Human cytomegalovirus (HCMV) infection has been actively implicated in complex neoplastic processes. Beyond oncomodulation, the molecular mechanisms that might underlie HCMV-induced oncogenesis are being extensively studied. Polycomb repressive complex 2 (PRC2) proteins, in particular enhancer of zeste homolog 2 (EZH2) are associated with cancer progression. Nevertheless, little is known about EZH2 activation in the context of HCMV infection and breast oncogenesis. METHODS: Herein, we identified EZH2 as a downstream target for HCMV-induced Myc upregulation upon acute and chronic infection with high-risk strains using a human mammary epithelial model. FINDINGS: We detected polyploidy and CMV-transformed HMECs (CTH) cells harboring HCMV and dynamically undergoing the giant cells cycle. Acquisition of embryonic stemness markers positively correlated with EZH2 and Myc expression. EZH2 inhibitors curtail sustained CTH cells' malignant phenotype. Besides harboring polyploid giant cancer cells (PGCCs), tumorigenic breast biopsies were characterized by an enhanced EZH2 and Myc expression, with a strong positive correlation between EZH2 and Myc expression, and between PGCC count and EZH2/Myc expression in the presence of HCMV. Further, we isolated two HCMV strains from EZH2HighMycHigh basal-like tumors which replicate in MRC5 cells and transform HMECs toward CTH cells after acute infection. INTERPRETATION: Our data establish a potential link between HCMV-induced Myc activation, the subsequent EZH2 upregulation, and polyploidy induction. These data support the proposed tumorigenesis properties of EZH2/Myc, and allow the isolation of two oncogenic HCMV strains from EZH2HighMycHigh basal breast tumors while identifying EZH2 as a potential therapeutic target in the management of breast cancer, particularly upon HCMV infection. FUNDING: This work was supported by grants from the University of Franche-Comté (UFC) (CR3300), the Région Franche-Comté (2021-Y-08292 and 2021-Y-08290) and the Ligue contre le Cancer (CR3304) to Georges Herbein. Zeina Nehme is a recipient of a doctoral scholarship from the municipality of Habbouch. Sandy Haidar Ahmad is recipient of a doctoral scholarship from Lebanese municipality. Ranim El Baba is a recipient of a doctoral scholarship from Hariri foundation for sustainable human development.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Proteína Potenciadora del Homólogo Zeste 2 , Glándulas Mamarias Humanas , Neoplasias , Proteínas Proto-Oncogénicas c-myc , Carcinogénesis , Citomegalovirus/genética , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/patología , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/virología , Células Gigantes/metabolismo , Células Gigantes/patología , Células Gigantes/virología , Humanos , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Glándulas Mamarias Humanas/virología , Poliploidía , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Regulación hacia Arriba
2.
Vet Microbiol ; 261: 109207, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34419774

RESUMEN

Mammary gland-derived Escherichia coli (E. coli) is an important pathogen causing dairy cow mastitis. YdiV, with EAL-like domains, inhibits flagellum biogenesis and motility and affects c-di-GMP (eubacterial signaling molecule) concentration changes in bacteria. However, the pathophysiological role of ydiV in host-pathogen cross-talk still needs to be elucidated. In this study, firstly constructed the ydiV mutant (NJ17ΔydiV) and ydiV complementary (cNJ17ΔydiV) E. coli strains to infect mouse mammary epithelial cells (EpH4-Ev) and macrophages (RAW264.7), as well as mouse mammary glands, respectively. Then biological characteristics, adaptor molecules in related signaling pathways, proinflammatory cytokines and the extent of host cell damage was evaluated. Compared with E. coli NJ17 infected mice, the bacterial load in the mammary gland of NJ17ΔydiV was significantly lower and the extent of the damage was alleviated. Notably, the deletion of ydiV significantly aggravated cell damage in RAW264.7 cells and compared with the wild-type strain, NJ17ΔydiV significantly activated the STING/TBK1/IRF3 pathway in macrophages. In EpH4-Ev cells, although STING did not sense E. coli NJ17 invasion, IRF3 was activated by the NJ17ΔydiV strain. Taken together, ydiV deletion significantly affects a variety of biological characteristics and induces severe cell damage, while the STING/TBK1/IRF3 pathway actively participated in pathogen elimination in the host. This study highlights a new role for ydiV in E. coli infection and provides a foundation for further studies to better understand host-bacteria interactions and potential prophylactic strategies for infectious diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Proteínas de Escherichia coli/metabolismo , Escherichia coli/genética , Escherichia coli/inmunología , Interacciones Microbiota-Huesped/inmunología , Evasión Inmune/genética , Animales , Carga Bacteriana , Proteínas Portadoras/genética , Línea Celular , Células Epiteliales/inmunología , Células Epiteliales/virología , Proteínas de Escherichia coli/genética , Femenino , Interacciones Microbiota-Huesped/genética , Humanos , Factor 3 Regulador del Interferón/inmunología , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/virología , Proteínas de la Membrana/inmunología , Ratones , Mutación , Proteínas Serina-Treonina Quinasas/inmunología , Células RAW 264.7
3.
Elife ; 102021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34223819

RESUMEN

Early events in retrovirus transmission are determined by interactions between incoming viruses and frontline cells near entry sites. Despite their importance for retroviral pathogenesis, very little is known about these events. We developed a bioluminescence imaging (BLI)-guided multiscale imaging approach to study these events in vivo. Engineered murine leukemia reporter viruses allowed us to monitor individual stages of retrovirus life cycle including virus particle flow, virus entry into cells, infection and spread for retroorbital, subcutaneous, and oral routes. BLI permitted temporal tracking of orally administered retroviruses along the gastrointestinal tract as they traversed the lumen through Peyer's patches to reach the draining mesenteric sac. Importantly, capture and acquisition of lymph-, blood-, and milk-borne retroviruses spanning three routes was promoted by a common host factor, the I-type lectin CD169, expressed on sentinel macrophages. These results highlight how retroviruses co-opt the immune surveillance function of tissue-resident sentinel macrophages for establishing infection.


Asunto(s)
Infecciones por Retroviridae/diagnóstico por imagen , Infecciones por Retroviridae/transmisión , Retroviridae/fisiología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Humanos , Virus de la Leucemia Murina , Estadios del Ciclo de Vida , Ganglios Linfáticos , Macrófagos/virología , Masculino , Glándulas Mamarias Humanas/diagnóstico por imagen , Glándulas Mamarias Humanas/virología , Ratones , Retroviridae/genética , Infecciones por Retroviridae/metabolismo , Infecciones por Retroviridae/patología , Lectina 1 Similar a Ig de Unión al Ácido Siálico/genética , Bazo/diagnóstico por imagen , Virión , Internalización del Virus
4.
Mol Biol Rep ; 47(6): 4383-4392, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32410141

RESUMEN

The ACE2 gene is a receptor of SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) for COVID-19 (coronavirus disease 2019). To analyze the expression profiles and clinical significances for this gene in humans, RNA-seq data representing 27 different tissues were analyzed using NCBI; total RNA was extracted from different tissues of mouse and semi-quantitative reverse transcriptional-polymerase chain reaction (Q-RT-PCR) was carried out. Immunohistochemistry expression profiles in normal tissues and cancer tissues and TCGA survival analysis in renal and liver cancer were conducted. ACE2 was highly conserved in different species. In normal tissues, ACE2 expression distributions were organ-specific, mainly in the kidney, male testis and female breast, and cardiovascular and gastrointestinal systems. High level of expression in testis, cardiovascular and gastrointestinal system indicated that SARS-CoV-2 might not only attack the lungs, but also affect other organs, particularly the testes, thus it may severely damage male sexual development for younger male and lead to infertility in an adult male, if he contracted COVID-19. On the other side, high expression of ACE2 was correlated with increased survival rate in renal and liver cancer, indicating that ACE2 is a prognostic marker in both renal cancer and liver cancers. Thus, the ACE2 is a functional receptor for SARS-CoV-2 and has a potential anti-tumor role in cancer. Taken together, this study may not only provide potential clues for further medical pathogenesis of COVID-19 and male fertility, but also indicate the clinical significance of the role of the ACE2 gene in cancer.


Asunto(s)
Betacoronavirus/patogenicidad , Infecciones por Coronavirus/epidemiología , Neoplasias Renales/genética , Neoplasias Hepáticas/genética , Peptidil-Dipeptidasa A/genética , Neumonía Viral/epidemiología , Receptores Virales/genética , Glicoproteína de la Espiga del Coronavirus/genética , Adulto , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/efectos de los fármacos , Betacoronavirus/genética , COVID-19 , Infecciones por Coronavirus/diagnóstico , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/genética , Bases de Datos Genéticas , Femenino , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Humanos , Riñón/metabolismo , Riñón/patología , Riñón/virología , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Neoplasias Renales/virología , Hígado/metabolismo , Hígado/patología , Hígado/virología , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Masculino , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Glándulas Mamarias Humanas/virología , Ratones , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/diagnóstico , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/genética , Unión Proteica , Receptores Virales/metabolismo , SARS-CoV-2 , Análisis de Secuencia de ARN , Transducción de Señal , Glicoproteína de la Espiga del Coronavirus/metabolismo , Análisis de Supervivencia , Testículo/metabolismo , Testículo/patología , Testículo/virología
5.
Viruses ; 11(10)2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31619008

RESUMEN

Zika virus (ZIKV) belongs to the large category of arboviruses. Surprisingly, several human-to-human transmissions of ZIKV have been notified, either following sexual intercourse or from the mother to fetus during pregnancy. Importantly, high viral loads have been detected in the human breast milk of infected mothers, and the existence of breastfeeding as a new mode of mother-to-child transmission of ZIKV was recently hypothesized. However, the maternal origin of infectious particles in breast milk is currently unknown. Here, we show that ZIKV disseminates to the mammary glands of infected mice after both systemic and local exposure with differential kinetics. Ex vivo, we demonstrate that primary human mammary epithelial cells were sensitive and permissive to ZIKV infection in this study. Moreover, by using in vitro models, we prove that mammary luminal- and myoepithelial-phenotype cell lines are both able to produce important virus progeny after ZIKV exposure. Our data suggest that the dissemination of ZIKV to the mammary glands and subsequent infection of the mammary epithelium could be one mechanism of viral excretion in human breast milk.


Asunto(s)
Células Epiteliales/virología , Glándulas Mamarias Humanas/virología , Tropismo Viral , Replicación Viral , Virus Zika/crecimiento & desarrollo , Animales , Línea Celular , Femenino , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Glándulas Mamarias Humanas/citología , Ratones , Leche Humana/virología , Embarazo , ARN Viral , Carga Viral , Virus Zika/genética , Virus Zika/fisiología
6.
J Virol ; 92(1)2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29046454

RESUMEN

Increasing evidence suggests that repetitive elements may play a role in host gene regulation, particularly through the donation of alternative promoters, enhancers, splice sites, and termination signals. Elevated transcript expression of the endogenous retrovirus group HERV-K (HML-2) is seen in many human cancers, although the identities of the individual proviral loci contributing to this expression as well as their mechanisms of activation have been unclear. Using high-throughput next-generation sequencing techniques optimized for the capture of HML-2 expression, we characterized the HML-2 transcriptome and means of activation in an in vitro model of human mammary epithelial cell transformation. Our analysis showed significant expression originating from 15 HML-2 full-length proviruses, through four modes of transcription. The majority of expression was in the antisense orientation and from proviruses integrated within introns. We found two instances of long terminal repeat (LTR)-driven provirus transcription but no evidence to suggest that these active 5' LTRs were influencing nearby host gene expression. Importantly, LTR-driven transcription was restricted to tumorigenic cells, suggesting that LTR promoter activity is dependent upon the transcriptional environment of a malignant cell.IMPORTANCE Here, we use an in vitro model of human mammary epithelial cell transformation to assess how malignancy-associated shifts in the transcriptional milieu of a cell may impact HML-2 activity. We found 15 proviruses to be significantly expressed through four different mechanisms, with the majority of transcripts being antisense copies of proviruses located within introns. We saw active 5' LTR use in tumorigenic cells only, suggesting that the cellular environment of a cancer cell is a critical component for induction of LTR promoter activity. These findings have implications for future studies investigating HML-2 as a target for immunotherapy or as a biomarker for disease.


Asunto(s)
Transformación Celular Viral , Retrovirus Endógenos/genética , Células Epiteliales/virología , Glándulas Mamarias Humanas/citología , Glándulas Mamarias Humanas/virología , Transcripción Genética , Línea Celular Tumoral , Regulación de la Expresión Génica , Genoma Humano , Genoma Viral , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Regiones Promotoras Genéticas , Provirus/genética , Secuencias Repetidas Terminales , Transcriptoma
7.
PLoS Pathog ; 11(10): e1005173, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26448646

RESUMEN

Seasonal influenza viruses are typically restricted to the human upper respiratory tract whereas influenza viruses with greater pathogenic potential often also target extra-pulmonary organs. Infants, pregnant women, and breastfeeding mothers are highly susceptible to severe respiratory disease following influenza virus infection but the mechanisms of disease severity in the mother-infant dyad are poorly understood. Here we investigated 2009 H1N1 influenza virus infection and transmission in breastfeeding mothers and infants utilizing our developed infant-mother ferret influenza model. Infants acquired severe disease and mortality following infection. Transmission of the virus from infants to mother ferrets led to infection in the lungs and mother mortality. Live virus was also found in mammary gland tissue and expressed milk of the mothers which eventually led to milk cessation. Histopathology showed destruction of acini glandular architecture with the absence of milk. The virus was localized in mammary epithelial cells of positive glands. To understand the molecular mechanisms of mammary gland infection, we performed global transcript analysis which showed downregulation of milk production genes such as Prolactin and increased breast involution pathways indicated by a STAT5 to STAT3 signaling shift. Genes associated with cancer development were also significantly increased including JUN, FOS and M2 macrophage markers. Immune responses within the mammary gland were characterized by decreased lymphocyte-associated genes CD3e, IL2Ra, CD4 with IL1ß upregulation. Direct inoculation of H1N1 into the mammary gland led to infant respiratory infection and infant mortality suggesting the influenza virus was able to replicate in mammary tissue and transmission is possible through breastfeeding. In vitro infection studies with human breast cells showed susceptibility to H1N1 virus infection. Together, we have shown that the host-pathogen interactions of influenza virus infection in the mother-infant dyad initiate immunological and oncogenic signaling cascades within the mammary gland. These findings suggest the mammary gland may have a greater role in infection and immunity than previously thought.


Asunto(s)
Animales Lactantes/virología , Interacciones Huésped-Parásitos/fisiología , Glándulas Mamarias Animales/virología , Glándulas Mamarias Humanas/virología , Infecciones por Orthomyxoviridae/transmisión , Animales , Animales Recién Nacidos , Western Blotting , Línea Celular , Modelos Animales de Enfermedad , Femenino , Hurones , Humanos , Inmunohistoquímica , Subtipo H1N1 del Virus de la Influenza A , Gripe Humana/virología , Lactancia , Glándulas Mamarias Animales/patología , Microscopía Confocal , Leche/virología , Madres , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Orthomyxoviridae/patología , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcriptoma
8.
PLoS One ; 10(9): e0134304, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26332838

RESUMEN

BACKGROUND: Age, reproductive history, hormones, genetics, and lifestyle are known risk factors for breast cancer, but the agents that initiate cellular changes from normal to malignant are not understood. We previously detected bovine leukemia virus (BLV), a common oncogenic virus of cattle, in the breast epithelium of humans. The objective of this study was to determine whether the presence of BLV DNA in human mammary epithelium is associated with breast cancer. METHODS: This was a case-control study of archival formalin fixed paraffin embedded breast tissues from 239 donors, received 2002-2008 from the Cooperative Human Tissue Network. Case definition as breast cancer versus normal (women with no history of breast cancer) was established through medical records and examination of tissues by an anatomical pathologist. Breast exposure to BLV was determined by in situ-PCR detection of a biomarker, BLV DNA, localized within mammary epithelium. RESULTS: The frequency of BLV DNA in mammary epithelium from women with breast cancer (59%) was significantly higher than in normal controls (29%) (multiply- adjusted odds ratio = 3.07, confidence interval = 1.66-5.69, p = .0004, attributable risk = 37%). In women with premalignant breast changes the frequency of BLV DNA was intermediate (38%) between that of women with breast cancer and normal controls (p for trend < .001). CONCLUSIONS: Among the specimens in this study, the presence of amplified BLV DNA was significantly associated with breast cancer. The odds ratio magnitude was comparable to those of well-established breast cancer risk factors related to reproductive history, hormones, and lifestyle and was exceeded only by risk factors related to genetics (familial breast cancer), high dose ionizing radiation, and age. These findings have the potential for primary and secondary prevention of breast cancer.


Asunto(s)
Neoplasias de la Mama/virología , Virus de la Leucemia Bovina/aislamiento & purificación , Glándulas Mamarias Humanas/virología , Adulto , Anciano , Animales , Biomarcadores , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Bovinos , Femenino , Humanos , Virus de la Leucemia Bovina/genética , Glándulas Mamarias Humanas/patología , Persona de Mediana Edad
9.
J Gen Virol ; 96(Pt 3): 650-662, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25491421

RESUMEN

Mouse mammary tumour virus (MMTV) is a member of the genus Betaretrovirus, infects rodent cells and uses mouse tranferrin receptor 1 for cell entry. Several MMTV strains have been shown to productively infect, in addition to murine cells, various heterologous cell lines including those of human origin, albeit less efficiently than murine cells. Here, we analysed whether MMTV from C3H mice [MMTV(C3H)], reported previously to be incapable of infecting human cells, could productively infect human cells. Using a recently described high-titre MMTV-based vector carrying MMTV(C3H) envelope protein (Env), we successfully transduced cells of human origin. Furthermore, WT MMTV(C3H) was able to infect human cells, albeit less efficiently than mouse cells. The established infection was, however, sufficient to enable virus spread to every cell in culture. The infectivity of WT MMTV(C3H) and MMTV-based vectors carrying MMTV(C3H)Env was blocked by heat inactivation, an inhibitor of reverse transcription (3'-azido-3'-deoxythymidine) and pre-incubation with neutralizing anti-MMTV antibodies that did not neutralize vectors pseudotyped with amphotropic murine leukemia virus Env, providing evidence for an authentic, receptor-mediated and reverse transcriptase-dependent infection process. Persistently infected human Hs578T cells produced infectious virions capable of infecting naïve human breast cells in culture, the infectivity of which could also be blocked by neutralizing anti-MMTV antibodies, demonstrating that virus particles released by the persistently infected Hs578T cells were related antigenically to the virus produced from murine cells. Taken together, our results show that MMTV(C3H), like MMTV(GR) and MMTV(RIII), is able not only to infect but also to replicate in cultured human breast cells.


Asunto(s)
Glándulas Mamarias Humanas/virología , Virus del Tumor Mamario del Ratón/fisiología , Infecciones Tumorales por Virus/virología , Animales , ADN Viral/genética , Regulación Viral de la Expresión Génica/fisiología , Vectores Genéticos , Humanos , Virus del Tumor Mamario del Ratón/clasificación , Ratones , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
10.
BMC Cancer ; 14: 942, 2014 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-25495285

RESUMEN

BACKGROUND: Breast cancer is a complex multifactorial genetic disease. Among other factors, race and, to an even greater extent, viruses are known to influence the development of this heterogeneous disease. It has been reported that MMTV-like (HMTV) gene sequences with a 90 to 98% homology to mouse mammary tumor virus are found in several populations with a prevalence range of 0 to 74%. In the Mexican population, 4.2% of patients with breast cancer exhibit the presence of HMTV (MMTV-like) sequences. The aim of this study was to evaluate the presence and current prevalence of retroviral HMTV (MMTV-like) sequences in breast cancer in Mexican women. METHODS: We used nested PCR and real-time PCR with a TaqMan probe. As a positive control, we used the C3H MMTV strain inserted into pBR322 plasmid. To confirm that we had identified the HMTV sequences, we sequenced the amplicons and compared these sequences with those of MMTV and HMTV (GenBank AF033807 and AF346816). RESULTS: A total of 12.4% of breast tumors were HMTV-positive, and 15.7% of the unaffected tissue samples from 458 patients were HMTV-positive. A total of 8.3% of the patients had both HMTV-positive tumor and adjacent tissues. The HMTV-positive samples presented 98% similarity to the reported HMTV sequence. CONCLUSIONS: These results confirm that the HMTV sequence is present in breast tumors and non-affected tissues in the Mexican population. HMTV should be considered a prominent causative agent of breast cancer.


Asunto(s)
Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/etiología , Virus del Tumor Mamario del Ratón , Infecciones por Retroviridae/complicaciones , Infecciones Tumorales por Virus/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama/patología , Estudios Transversales , ADN Viral , Femenino , Productos del Gen env/genética , Humanos , Glándulas Mamarias Humanas/virología , Virus del Tumor Mamario del Ratón/clasificación , Virus del Tumor Mamario del Ratón/genética , México/epidemiología , Ratones , Persona de Mediana Edad , Filogenia , Prevalencia , Estudios Prospectivos , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/virología
11.
Virology ; 460-461: 55-65, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25010270

RESUMEN

Urine and breast milk represent the main routes of human cytomegalovirus (HCMV) transmission but the contribution of renal and mammary epithelial cells to viral excretion remains unclear. We observed that kidney and mammary epithelial cells were permissive to HCMV infection and expressed immediate early, early and late antigens within 72 h of infection. During the first 24 h after infection, high titers of infectious virus were measured associated to the cells and in culture supernatants, independently of de novo synthesis of virus progeny. This phenomenon was not observed in HCMV-infected fibroblasts and suggested the sequestration and the release of HCMV by epithelial cells. This hypothesis was supported by confocal and electron microscopy analyses. The sequestration and progressive release of HCMV by kidney and mammary epithelial cells may play an important role in the excretion of the virus in urine and breast milk and may thereby contribute to HCMV transmission.


Asunto(s)
Infecciones por Citomegalovirus/virología , Citomegalovirus/fisiología , Células Epiteliales/virología , Riñón/virología , Glándulas Mamarias Humanas/virología , Línea Celular , Citomegalovirus/genética , Femenino , Fibroblastos/virología , Humanos , Riñón/citología , Glándulas Mamarias Humanas/citología , Replicación Viral
12.
Emerg Infect Dis ; 20(5): 772-82, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24750974

RESUMEN

Bovine leukemia virus (BLV), a deltaretrovirus, causes B-cell leukemia/lymphoma in cattle and is prevalent in herds globally. A previous finding of antibodies against BLV in humans led us to examine the possibility of human infection with BLV. We focused on breast tissue because, in cattle, BLV DNA and protein have been found to be more abundant in mammary epithelium than in lymphocytes. In human breast tissue specimens, we identified BLV DNA by using nested liquid-phase PCR and DNA sequencing. Variations from the bovine reference sequence were infrequent and limited to base substitutions. In situ PCR and immunohistochemical testing localized BLV to the secretory epithelium of the breast. Our finding of BLV in human tissues indicates a risk for the acquisition and proliferation of this virus in humans. Further research is needed to determine whether BLV may play a direct role in human disease.


Asunto(s)
ADN Viral , Virus de la Leucemia Bovina/genética , Glándulas Mamarias Humanas/virología , Provirus , Animales , Secuencia de Bases , Bovinos , Femenino , Genes Virales , Genoma Viral , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo
13.
Virol J ; 11: 65, 2014 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-24708706

RESUMEN

BACKGROUND: Small ruminant lentiviruses escaping efficient serological detection are still circulating in Swiss goats in spite of a long eradication campaign that essentially eliminated clinical cases of caprine arthritis encephalitis in the country. This strongly suggests that the circulating viruses are avirulent for goats.To test this hypothesis, we isolated circulating viruses from naturally infected animals and tested the in vitro and in vivo characteristics of these field isolates. METHODS: Viruses were isolated from primary macrophage cultures. The presence of lentiviruses in the culture supernatants was monitored by reverse transcriptase assay. Isolates were passaged in different cells and their cytopathogenic effects monitored by microscopy. Proviral load was quantified by real-time PCR using customized primer and probes. Statistical analysis comprised Analysis of Variance and Bonferroni Multiple Comparison Test. RESULTS: The isolated viruses belonged to the small ruminant lentiviruses A4 subtype that appears to be prominent in Switzerland. The 4 isolates replicated very efficiently in macrophages, displaying heterogeneous phenotypes, with two isolates showing a pronounced cytopathogenicity for these cells. By contrast, all 4 isolates had a poor replication capacity in goat and sheep fibroblasts. The proviral loads in the peripheral blood and, in particular, in the mammary gland were surprisingly high compared to previous observations. Nevertheless, these viruses appear to be of low virulence for goats except for the mammary gland were histopathological changes were observed. CONCLUSIONS: Small ruminant lentiviruses continue to circulate in Switzerland despite a long and expensive caprine arthritis encephalitis virus eradication campaign. We isolated 4 of these lentiviruses and confirmed their phylogenetic association with the prominent A4 subtype. The pathological and histopathological analysis of the infected animals supported the hypothesis that these A4 viruses are of low pathogenicity for goats, with, however, a caveat about the potentially detrimental effects on the mammary gland. Moreover, the high proviral load detected indicates that the immune system of the animals cannot control the infection and this, combined with the phenotypic plasticity observed in vitro, strongly argues in favour of a continuous and precise monitoring of these SRLV to avoid the risk of jeopardizing a long eradication campaign.


Asunto(s)
Virus de la Artritis-Encefalitis Caprina/genética , Virus de la Artritis-Encefalitis Caprina/patogenicidad , Enfermedades de las Cabras/virología , Infecciones por Lentivirus/veterinaria , Animales , Virus de la Artritis-Encefalitis Caprina/clasificación , Virus de la Artritis-Encefalitis Caprina/aislamiento & purificación , Sangre/virología , Células Cultivadas , Análisis por Conglomerados , Efecto Citopatogénico Viral , Fibroblastos/virología , Genotipo , Enfermedades de las Cabras/epidemiología , Cabras , Humanos , Infecciones por Lentivirus/epidemiología , Infecciones por Lentivirus/virología , Macrófagos/virología , Glándulas Mamarias Humanas/virología , Microscopía , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena de la Polimerasa , Provirus/genética , Provirus/aislamiento & purificación , ARN Viral/genética , Análisis de Secuencia de ADN , Ovinos , Suiza/epidemiología , Carga Viral
14.
Neoplasia ; 15(9): 1100-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24027434

RESUMEN

Epithelial-to-mesenchymal transition (EMT) facilitates the escape of epithelial cancer cells from the primary tumor site, which is a key event early in metastasis. Here, we explore how extrinsic, tumor microenvironmental cytokines cooperate with intrinsic, genetic changes to promote EMT in human mammary epithelial cells (HMECs). Viral transduction of transforming genetic events into HMECs routinely generated two distinct cell populations. One population retained epithelial characteristics, while an emergent population spontaneously acquired a mesenchymal morphology and properties associated with cancer stem cells (CSCs). Interestingly, the spontaneous mesenchymal/CSCs were unable to differentiate and lacked epithelial-mesenchymal plasticity. In contrast, exposure of the transformed HMECs retaining epithelial characteristics to exogenous transforming growth factor-ß (TGF-ß) generated a mesenchymal/CSC population with remarkable plasticity. The TGF-ß-induced mesenchymal/CSC population was dependent on the continued presence of TGF-ß. Removal of TGF-ß or pharmacologic or genetic inhibition of TGF-ß/SMAD signaling led to the reversion of mesenchymal/CSC to epithelial/non-CSC. Our results demonstrate that targeting exogenous cytokine signaling disrupts epithelial-mesenchymal plasticity and may be an effective strategy to inhibit the emergence of circulating tumor cells. The model of epithelial-mesenchymal plasticity we describe here can be used to identify novel tumor microenvironmental factors and downstream signaling that cooperate with intrinsic genetic changes to drive metastasis. Understanding the interaction between extrinsic and intrinsic factors that regulate epithelial-mesenchymal plasticity will allow the development of new therapies that target tumor microenvironmental signals to reduce metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Transición Epitelial-Mesenquimal/genética , Glándulas Mamarias Humanas/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Neoplasias de la Mama/genética , Proliferación Celular , Transformación Celular Neoplásica , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Humanos , Glándulas Mamarias Humanas/virología , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Transducción de Señal , Proteínas Smad/metabolismo , Células Tumorales Cultivadas , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Virus Res ; 163(2): 537-45, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22178805

RESUMEN

The caprine arthritis encephalitis virus (CAEV) long terminal repeat promoter was cloned and sequenced from mammary gland and carpal joint synovium isolated from a 15.5 year old, CAEV-infected Toggenburg doe with chronic mastitis and carpal arthritis. A deletion of the CAEV gamma activated site (GAS) was identified in the mammary gland but not the synovial isolate. Subsequent promoter-reporter gene construct experiments indicated that the GAS is necessary for interferon γ-mediated promoter activation. Utilizing a molecular clone of the classic isolate CAEV-CO, these findings were corroborated by a set of GAS mutant promoter-reporter constructs with and without the CAEV GAS. Results of experiments with U937 monocyte cell lines stably transfected with molecular clones of CAEV-CO GAS deletion mutants also indicated the GAS is necessary for IFNγ-mediated promoter activation. The mammary gland CAE viral isolate was propagated in caprine peripheral blood mononuclear cells and was assigned the name CAEV-MA. This is the first report describing two CAE viral isolates cloned from different anatomical locations in the same animal with and without the CAEV GAS, and is the first report detailing cytokine-induced CAEV promoter function in a naturally occurring ΔGAS promoter.


Asunto(s)
Virus de la Artritis-Encefalitis Caprina/patogenicidad , Interferón gamma/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Proteínas Virales/biosíntesis , Animales , Virus de la Artritis-Encefalitis Caprina/aislamiento & purificación , Línea Celular , Genes Reporteros , Cabras , Humanos , Leucocitos Mononucleares/virología , Glándulas Mamarias Humanas/virología , Datos de Secuencia Molecular , Monocitos/virología , Análisis de Secuencia de ADN , Eliminación de Secuencia , Membrana Sinovial/virología , Secuencias Repetidas Terminales/genética
16.
Mol Cancer ; 10: 97, 2011 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-21827643

RESUMEN

BACKGROUND: In normal cells proliferation and apoptosis are tightly regulated, whereas in tumor cells the balance is shifted in favor of increased proliferation and reduced apoptosis. Anticancer agents mediate tumor cell death via targeting multiple pathways of programmed cell death. We have reported that the non-pathogenic, tumor suppressive Adeno-Associated Virus Type 2 (AAV2) induces apoptosis in Human Papillomavirus (HPV) positive cervical cancer cells, but not in normal keratinocytes. In the current study, we examined the potential of AAV2 to inhibit proliferation of MCF-7 and MDA-MB-468 (both weakly invasive), as well as MDA-MB-231 (highly invasive) human breast cancer derived cell lines. As controls, we used normal human mammary epithelial cells (nHMECs) isolated from tissue biopsies of patients undergoing breast reduction surgery. RESULTS: AAV2 infected MCF-7 line underwent caspase-independent, and MDA-MB-468 and MDA-MB-231 cell lines underwent caspase-dependent apoptosis. Death of MDA-MB-468 cells was marked by caspase-9 activation, whereas death of MDA-MB-231 cells was marked by activation of both caspase-8 and caspase-9, and resembled a mixture of apoptotic and necrotic cell death. Cellular demise was correlated with the ability of AAV2 to productively infect and differentially express AAV2 non-structural proteins: Rep78, Rep68 and Rep40, dependent on the cell line. Cell death in the MCF-7 and MDA-MB-231 lines coincided with increased S phase entry, whereas the MDA-MB-468 cells increasingly entered into G2. AAV2 infection led to decreased cell viability which correlated with increased expression of proliferation markers c-Myc and Ki-67. In contrast, nHMECs that were infected with AAV2 failed to establish productive infection or undergo apoptosis. CONCLUSION: AAV2 regulated enrichment of cell cycle check-point functions in G1/S, S and G2 phases could create a favorable environment for Rep protein expression. Inherent Rep associated endonuclease activity and AAV2 genomic hair-pin ends have the potential to induce a cellular DNA damage response, which could act in tandem with c-Myc regulated/sensitized apoptosis induction. In contrast, failure of AAV2 to productively infect nHMECs could be clinically advantageous. Identifying the molecular mechanisms of AAV2 targeted cell cycle regulation of death inducing signals could be harnessed for developing novel therapeutics for weakly invasive as well as aggressive breast cancer types.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama/patología , Carcinoma/patología , Caspasas/fisiología , Dependovirus/fisiología , Glándulas Mamarias Humanas/metabolismo , Infecciones por Parvoviridae/patología , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/virología , Carcinoma/complicaciones , Carcinoma/metabolismo , Carcinoma/virología , Caspasas/metabolismo , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dependovirus/genética , Femenino , Regulación Viral de la Expresión Génica , Genoma Viral/fisiología , Humanos , Glándulas Mamarias Humanas/patología , Glándulas Mamarias Humanas/fisiopatología , Glándulas Mamarias Humanas/virología , Infecciones por Parvoviridae/complicaciones , Infecciones por Parvoviridae/metabolismo , Transducción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral/fisiología
17.
Breast ; 20(2): 184-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21146410

RESUMEN

Human papillomavirus (HPV) has been implicated in breast carcinogenesis. Consecutive and non-selected mastectomy specimens from Mexican patients harboring breast carcinomas were sampled in order to look for the presence of HPV DNA. HPV-16 was detected in 6 (10%) of 60 breast carcinomas. Two of these also had HPV genome in adjacent non-neoplastic mammary-tissues. Seven cases had HPV DNA only in non-neoplastic tissue specimens. HPV DNA was also detected in 4 (25%) of 10 tumor-bed specimens without residual neoplastic lesions that were obtained from patients who underwent neoadjuvant chemotherapy or neoadjuvant chemotherapy/radiotherapy. HPV-positive tumors tended to be smaller in size, than HPV-negative tumors (p=0.047). Histological distributions of HPV-positive and -negative cases showed no significant difference. Although all the HPV-16 DNA were found integrated, its low viral load rendered it difficult to incriminate this virus in breast carcinogenesis. However, the possibility that HPV infection occurred during carcinoma development cannot be ruled out.


Asunto(s)
Neoplasias de la Mama/virología , Carcinoma Ductal de Mama/virología , Carcinoma Lobular/virología , Carcinoma Papilar/virología , Papillomavirus Humano 16/aislamiento & purificación , Infecciones por Papillomavirus/complicaciones , Adulto , Anciano , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/terapia , Carcinoma Ductal de Mama/epidemiología , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/terapia , Carcinoma Lobular/epidemiología , Carcinoma Lobular/metabolismo , Carcinoma Lobular/terapia , Carcinoma Papilar/epidemiología , Carcinoma Papilar/metabolismo , Carcinoma Papilar/terapia , Femenino , Humanos , Glándulas Mamarias Humanas/virología , México , Persona de Mediana Edad , Pezones/virología , Infecciones por Papillomavirus/epidemiología , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Carga Viral
18.
Cell Host Microbe ; 8(6): 534-43, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-21147467

RESUMEN

Viruses, including retroviruses like human immunodeficiency virus (HIV) and mouse mammary tumor virus (MMTV), are transmitted from mother to infants through milk. Lymphoid cells and antibodies are thought to provide mammary gland and milk-borne immunity. In contrast, little is known about the role of mammary epithelial cells (MECs). The APOBEC3 family of retroviral restriction factors is highly expressed in macrophages and lymphoid and dendritic cells. We now show that APOBEC3 proteins are also expressed in mouse and human MECs. Lymphoid cell-expressed APOBEC3 restricts in vivo spread of MMTV to lymphoid and mammary tissue. In contrast, mammary gland-expressed APOBEC3 is packaged into MMTV virions and decreases the infectivity of milk-borne viruses. Moreover, APOBEC3G and other APOBEC3 genes are expressed in human mammary cells and have the potential to restrict viruses produced in this cell type. These data point to a role for APOBEC3 proteins in limiting infectivity of milk-transmitted viruses.


Asunto(s)
Citidina Desaminasa/fisiología , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Virus del Tumor Mamario del Ratón/fisiología , Leche/virología , Infecciones por Retroviridae/metabolismo , Desaminasas APOBEC , Animales , Células Cultivadas , Citidina Desaminasa/biosíntesis , Citosina Desaminasa/biosíntesis , Citosina Desaminasa/fisiología , Células Epiteliales/virología , Femenino , VIH-1/patogenicidad , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Linfocitos/metabolismo , Glándulas Mamarias Animales/virología , Glándulas Mamarias Humanas/virología , Virus del Tumor Mamario del Ratón/patogenicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/virología , Infecciones por Retroviridae/transmisión , Infecciones por Retroviridae/virología , Infecciones Tumorales por Virus/metabolismo , Infecciones Tumorales por Virus/transmisión , Infecciones Tumorales por Virus/virología , Virión/fisiología , Ensamble de Virus , Productos del Gen vif del Virus de la Inmunodeficiencia Humana/fisiología
20.
Gene Ther ; 16(12): 1477-82, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19693098

RESUMEN

Oncolytic herpes simplex viruses type 1 (oHSVs) such as G47Delta and G207 are genetically engineered for selective replication competence in cancer cells. Several factors can influence the overall effectiveness of oHSV tropism, including HSV-1 receptor expression, extracellular matrix milieu and cellular permissiveness. We have taken advantage of human prostate organ cultures derived from radical prostatectomies to investigate oHSV tropism. In this study, we show that both G47Delta and G207 specifically replicate in epithelial cells of the prostatic glands but not in the surrounding stroma. In contrast, both the epithelial and stromal cell compartments were readily infected by wild-type HSV-1. Analysis of oHSV replication in prostate surgical specimens 3 days post infection showed that G47Delta generated approximately 30-fold more viral progeny than did G207. This correlated with the enhanced expression of G47Delta-derived glycoprotein gB protein levels as compared with G207. In benign prostate tissues, G207 and G47Delta titers were notably reduced, whereas strain F titers were maintained at similar levels compared with prostate cancer specimens. Overall, our results show that these oncolytic herpes vectors show both target specificity and replication competence in human prostate cancer specimens and point to the utility of using human prostate organ cultures in assessing oHSV tropism and cellular specificity.


Asunto(s)
Herpesvirus Humano 1/genética , Virus Oncolíticos/genética , Técnicas de Cultivo de Órganos , Animales , Ingeniería Genética , Vectores Genéticos , Humanos , Masculino , Glándulas Mamarias Humanas/virología , Ratones , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA