Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 252
Filtrar
1.
J Histotechnol ; 46(2): 80-89, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-35975713

RESUMEN

Transforming growth factor alpha (TGFα), a member of the epidermal growth factor (EGF) family, regulates cell proliferation, differentiation, and development, and involves follicular development and viability. In ovaries, TGFα is shown localized in granulosa cells (GCs) of primary follicles, theca cells (TCs) of pre-antral, antral and pre-ovulatory follicles. TGFα overexpression in mouse mammary tumor virus (MMTV-TGFα) transgenic mice causes mammary tumor after 50 weeks. However, follicular development and preservation of the ovarian follicle reserve-mediating follicle stimulating hormone (FSH) response are unknown. Mammalian target of rapamycin (mTOR) is a key regulator for cell proliferation, growth, differentiation, and apoptosis, and important for ovarian folliculogenesis and oocyte maturation. The study aim determines TGFα overexpression during folliculogenesis via mTOR signaling pathway in ovaries from 10-, 18-, 50-, and 82-week-old MMTV-TGFα mice. Histological analysis was performed, along with western blot for mTOR, p-mTOR, P70S6K, PCNA, and Caspase-3, and quantitative RNA (qRT-PCR) for mTOR and P70S6K. Developing follicles number decreased and atretic follicles number increased with aging in MMTV-TGFα mice ovary. Ovaries at 18 and 82 weeks had decreased PCNA and increased Caspase-3 protein expression levels as compared to 10-week ovaries. Protein expression levels of mTOR and p-mTOR decreased gradually from ovaries at 10-18 weeks, increased at 50 weeks and decreased again at 82 weeks. These results indicate that TGFα may be one regulator of healthy follicular development and affect mTOR signaling pathway during ovarian aging. Thus, over-expression of TGFα might lead to reduced ovarian reserve and premature ovarian insufficiency.


Asunto(s)
Ovario , Factor de Crecimiento Transformador alfa , Femenino , Ratones , Animales , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo , Caspasa 3/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Mamíferos/metabolismo
2.
Nucleic Acids Res ; 50(15): 8898-8918, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-35947647

RESUMEN

Integration into host target DNA (tDNA), a hallmark of retroviral replication, is mediated by the intasome, a multimer of integrase (IN) assembled on viral DNA (vDNA) ends. To ascertain aspects of tDNA recognition during integration, we have solved the 3.5 Å resolution cryo-EM structure of the mouse mammary tumor virus (MMTV) strand transfer complex (STC) intasome. The tDNA adopts an A-like conformation in the region encompassing the sites of vDNA joining, which exposes the sugar-phosphate backbone for IN-mediated strand transfer. Examination of existing retroviral STC structures revealed conservation of A-form tDNA in the analogous regions of these complexes. Furthermore, analyses of sequence preferences in genomic integration sites selectively targeted by six different retroviruses highlighted consistent propensity for A-philic sequences at the sites of vDNA joining. Our structure additionally revealed several novel MMTV IN-DNA interactions, as well as contacts seen in prior STC structures, including conserved Pro125 and Tyr149 residues interacting with tDNA. In infected cells, Pro125 substitutions impacted the global pattern of MMTV integration without significantly altering local base sequence preferences at vDNA insertion sites. Collectively, these data advance our understanding of retroviral intasome structure and function, as well as factors that influence patterns of vDNA integration in genomic DNA.


Asunto(s)
Integrasas , Integración Viral , Animales , Ratones , Integrasas/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , ADN Viral/genética , ADN Viral/química , Conformación Molecular , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/metabolismo
3.
J Virol ; 95(14): e0053121, 2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33952644

RESUMEN

Mouse mammary tumor virus (MMTV) encodes a Rem precursor protein that specifies both regulatory and accessory functions. Rem is cleaved at the endoplasmic reticulum (ER) membrane into a functional N-terminal signal peptide (SP) and the C terminus (Rem-CT). Rem-CT lacks a membrane-spanning domain and a known ER retention signal, and yet it was not detectably secreted into cell supernatants. Inhibition of intracellular trafficking by the drug brefeldin A (BFA), which interferes with the ER-to-Golgi secretory pathway, resulted in dramatically reduced intracellular Rem-CT levels that were not rescued by proteasomal or lysosomal inhibitors. A Rem mutant lacking glycosylation was cleaved into SP and Rem-CT but was insensitive to BFA, suggesting that unglycosylated Rem-CT does not reach this BFA-dependent compartment. Treatment with endoglycosidase H indicated that Rem-CT does not traffic through the Golgi apparatus. Analysis of wild-type Rem-CT and its glycosylation mutant by confocal microscopy revealed that both were primarily localized to the ER lumen. A small fraction of wild-type Rem-CT, but not the unglycosylated mutant, was colocalized with Rab5-positive (Rab5+) early endosomes. The expression of a dominant-negative (DN) form of ADP ribosylation factor 1 (Arf1) (containing a mutation of threonine to asparagine at position 31 [T31N]) mimicked the effects of BFA by reducing Rem-CT levels and increased Rem-CT association with early and late endosomes. Inhibition of the AAA ATPase p97/VCP rescued Rem-CT in the presence of BFA or DN Arf1 and prevented localization to Rab5+ endosomes. Thus, Rem-CT uses an unconventional p97-mediated scheme for trafficking to early endosomes. IMPORTANCE Mouse mammary tumor virus is a complex retrovirus that encodes a regulatory/accessory protein, Rem. Rem is a precursor protein that is processed at the endoplasmic reticulum (ER) membrane by signal peptidase. The N-terminal SP uses the p97/VCP ATPase to elude ER-associated degradation to traffic to the nucleus and serve a human immunodeficiency virus Rev-like function. In contrast, the function of the C-terminal glycosylated cleavage product (Rem-CT) is unknown. Since localization is critical for protein function, we used mutants, inhibitors, and confocal microscopy to localize Rem-CT. Surprisingly, Rem-CT, which lacks a transmembrane domain or an ER retention signal, was detected primarily within the ER and required glycosylation and the p97 ATPase for early endosome trafficking without passage through the Golgi apparatus. Thus, Rem-CT uses a novel intracellular trafficking pathway, potentially impacting host antiviral immunity.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Retículo Endoplásmico/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Proteínas Nucleares/metabolismo , Adenosina Trifosfatasas/antagonistas & inhibidores , Transporte Biológico/efectos de los fármacos , Brefeldino A/farmacología , Endosomas/metabolismo , Células HEK293 , Humanos , Microscopía Confocal , Proteínas Nucleares/antagonistas & inhibidores , Precursores de Proteínas/metabolismo , Proteínas del Envoltorio Viral/metabolismo
4.
Nucleic Acids Res ; 49(8): 4668-4688, 2021 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-33836091

RESUMEN

Retroviral RNA genome (gRNA) harbors cis-acting sequences that facilitate its specific packaging from a pool of other viral and cellular RNAs by binding with high-affinity to the viral Gag protein during virus assembly. However, the molecular intricacies involved during selective gRNA packaging are poorly understood. Binding and footprinting assays on mouse mammary tumor virus (MMTV) gRNA with purified Pr77Gag along with in cell gRNA packaging study identified two Pr77Gag binding sites constituting critical, non-redundant packaging signals. These included: a purine loop in a bifurcated stem-loop containing the gRNA dimerization initiation site, and the primer binding site (PBS). Despite these sites being present on both unspliced and spliced RNAs, Pr77Gag specifically bound to unspliced RNA, since only that could adopt the native bifurcated stem-loop structure containing looped purines. These results map minimum structural elements required to initiate MMTV gRNA packaging, distinguishing features that are conserved amongst divergent retroviruses from those perhaps unique to MMTV. Unlike purine-rich motifs frequently associated with packaging signals, direct involvement of PBS in gRNA packaging has not been documented in retroviruses. These results enhance our understanding of retroviral gRNA packaging/assembly, making it not only a target for novel therapeutic interventions, but also development of safer gene therapy vectors.


Asunto(s)
Productos del Gen gag/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Empalme del ARN , ARN Viral/metabolismo , Ensamble de Virus/genética , Animales , Sitios de Unión/genética , Cartilla de ADN , Dispersión Dinámica de Luz , Productos del Gen gag/genética , Genoma Viral , Virus del Tumor Mamario del Ratón/genética , Ratones , Conformación de Ácido Nucleico , Purinas , ARN Viral/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
5.
Br J Nutr ; 125(1): 1-9, 2021 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31685042

RESUMEN

The role of adiponectin and leptin signalling pathways has been suggested to play important roles in the protective effects of energy restriction (ER) on mammary tumour (MT) development. To study the effects of ER on the methylation levels in adiponectin receptor 1 (AdipoR1) and leptin receptor overlapping transcript (Leprot) genes using the pyrosequencing method in mammary fat pad tissue, mouse mammary tumour virus-transforming growth factor-α (MMTV-TGF-α) female mice were randomly assigned to ad libitum (AL), chronic ER (CER, 15 % ER) or intermittent ER (3 weeks AL and 1 week 60 % ER in cyclic periods) groups at 10 weeks of age until 82 weeks of age. The methylation levels of AdipoR1 in the CER group were higher than those in the AL group at week 49/50 (P < 0·05), while the levels of methylation for AdipoR1 and Leprot genes were similar among the other groups. Also, the methylation levels at CpG2 and CpG3 regions of the promoter region of the AdipoR1 gene in the CER group were three times higher (P < 0·05), while CpG1 island of Leprot methylation was significantly lower compared with the other groups (P < 0·05). Adiponectin and leptin gene expression levels were consistent with the methylation levels. We also observed a change with ageing in methylation levels of these genes. These results indicate that different types of ER modify methylation levels of AdipoR1 and Leprot in different ways and CER had a more significant effect on methylation levels of both genes. Epigenetic regulation of these genes may play important roles in the preventive effects of ER against MT development and ageing processes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Restricción Calórica/métodos , Ingestión de Energía/genética , Neoplasias Mamarias Experimentales/dietoterapia , Receptores de Adiponectina/metabolismo , Animales , Islas de CpG , Femenino , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/metabolismo , Metilación , Ratones , Transducción de Señal/genética , Factor de Crecimiento Transformador alfa/metabolismo
6.
Front Cell Infect Microbiol ; 11: 807462, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35096654

RESUMEN

Mouse mammary tumor virus (MMTV) is a virus that induces breast cancer in mice. During lactation, MMTV can transmit from mother to offspring through milk, and Peyer's patches (PPs) in mouse intestine are the first and specific target organ. MMTV can be transported into PPs by microfold cells and then activate antigen-presenting cells (APCs) by directly binding with Toll-like receptors (TLRs) whereas infect them through mouse transferrin receptor 1 (mTfR1). After being endocytosed, MMTV is reversely transcribed and the cDNA inserts into the host genome. Superantigen (SAg) expressed by provirus is presented by APCs to cognate CD4+ T cells via MHCII molecules to induce SAg response, which leads to substantial proliferation and recruitment of related immune cells. Both APCs and T cells can be infected by MMTV and these extensively proliferated lymphocytes and recruited dendritic cells act as hotbeds for viral replication and amplification. In this case, intestinal lymphatic tissues can actually become the source of infection for the transmission of MMTV in vivo, which results in mammary gland infection by MMTV and eventually lead to the occurrence of breast cancer.


Asunto(s)
Infecciones por Retroviridae , Infecciones Tumorales por Virus , Animales , Femenino , Intestinos , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Ratones Endogámicos BALB C , Superantígenos/genética , Linfocitos T
7.
Cell Oncol (Dordr) ; 43(6): 1117-1127, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32691367

RESUMEN

PURPOSE: Breast cancer is a heterogeneous disease with several subtypes that currently do not have targeted therapeutic options. Metabolomics has the potential to uncover novel targeted treatment strategies by identifying metabolic pathways required for cancer cells to survive and proliferate. METHODS: The metabolic profiles of two histologically distinct breast cancer subtypes from a MMTV-Myc mouse model, epithelial-mesenchymal-transition (EMT) and papillary, were investigated using mass spectrometry-based metabolomics methods. Based on metabolic profiles, drugs most likely to be effective against each subtype were selected and tested. RESULTS: We found that the EMT and papillary subtypes display different metabolic preferences. Compared to the papillary subtype, the EMT subtype exhibited increased glutathione and TCA cycle metabolism, while the papillary subtype exhibited increased nucleotide biosynthesis compared to the EMT subtype. Targeting these distinct metabolic pathways effectively inhibited cancer cell proliferation in a subtype-specific manner. CONCLUSIONS: Our results demonstrate the feasibility of metabolic profiling to develop novel personalized therapy strategies for different subtypes of breast cancer. Schematic overview of the experimental design for drug selection based on breast cancer subtype-specific metabolism. The epithelial mesenchymal transition (EMT) and papillary tumors are histologically distinct mouse mammary tumor subtypes from the MMTV-Myc mouse model. Cell lines derived from tumors can be used to determine metabolic pathways that can be used to select drug candidates for each subtype.


Asunto(s)
Neoplasias Mamarias Animales/tratamiento farmacológico , Neoplasias Mamarias Animales/metabolismo , Metabolómica , Terapia Molecular Dirigida , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Isótopos de Carbono , Línea Celular Tumoral , Ciclo del Ácido Cítrico/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Neoplasias Mamarias Animales/clasificación , Virus del Tumor Mamario del Ratón/metabolismo , Metaboloma/efectos de los fármacos , Ratones , Nucleótidos/biosíntesis , Proteínas Proto-Oncogénicas c-myc/metabolismo
8.
Nat Commun ; 10(1): 4131, 2019 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-31511510

RESUMEN

Type I interferons (IFN) are being rediscovered as potent anti-tumoral agents. Activation of the STimulator of INterferon Genes (STING) by DMXAA (5,6-dimethylxanthenone-4-acetic acid) can induce strong production of IFNα/ß and rejection of transplanted primary tumors. In the present study, we address whether targeting STING with DMXAA also leads to the regression of spontaneous MMTV-PyMT mammary tumors. We show that these tumors are refractory to DMXAA-induced regression. This is due to a blockade in the phosphorylation of IRF3 and the ensuing IFNα/ß production. Mechanistically, we identify TGFß, which is abundant in spontaneous tumors, as a key molecule limiting this IFN-induced tumor regression by DMXAA. Finally, blocking TGFß restores the production of IFNα by activated MHCII+ tumor-associated macrophages, and enables tumor regression induced by STING activation. On the basis of these findings, we propose that type I IFN-dependent cancer therapies could be greatly improved by combinations including the blockade of TGFß.


Asunto(s)
Interferón-alfa/metabolismo , Interferón beta/metabolismo , Neoplasias Mamarias Animales/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Femenino , Factor 3 Regulador del Interferón/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Fosforilación/efectos de los fármacos , Xantonas/farmacología
9.
J Mol Biol ; 430(21): 4307-4324, 2018 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-30179605

RESUMEN

BACKGROUND: The mouse mammary tumor virus (MMTV) encodes a functional signal peptide, a cleavage product of envelope and Rem proteins. Signal peptide interacts with a 3' cis-acting RNA element, the Rem-responsive element (RmRE), to facilitate expression of both unspliced genomic (gRNA) and spliced mRNAs. An additional RmRE has been proposed at the 5' end of the genome, facilitating nuclear export of the unspliced gRNA, whereas the 3' RmRE could facilitate translation of all other mRNAs, including gRNA. RESULTS: To address this hypothesis, a series of mutations were introduced into a 24-nt region found exclusively in the unspliced gRNA. Mutant clones using MMTV or human cytomegalovirus promoters were tested in both transient and stable transfections to determine their effect on gRNA nuclear export, stability, and translation. Nuclear export of the gRNA was affected only in a small mutant subset in stably transfected Jurkat T cells. Quantitative real-time RT-PCR of actinomycin D-treated cells expressing MMTV revealed that multiple mutants were severely compromised for RNA expression and stability. Both genomic and spliced nuclear RNAs were reduced, leading to abrogation of Gag and Env protein expressed from unspliced and spliced mRNAs, respectively. RT-PCRs with multiple primer pairs indicated failure to elongate genomic MMTV transcripts beyond ~500 nt compared to the wild type in a cell line-dependent manner. CONCLUSIONS: MMTV contains a novel cis-acting element downstream of the major splice donor critical for facilitating MMTV gRNA elongation and stability. Presence of a mirror repeat within the element may represent important viral/host factor binding site(s) within MMTV gRNA.


Asunto(s)
Virus del Tumor Mamario del Ratón/genética , ARN Viral/química , ARN Viral/metabolismo , Transporte Activo de Núcleo Celular , Animales , Citomegalovirus/genética , Dactinomicina , Humanos , Células Jurkat , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Mutación , Regiones Promotoras Genéticas , Empalme del ARN , Estabilidad del ARN
10.
RNA Biol ; 15(8): 1047-1059, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29929424

RESUMEN

Packaging the mouse mammary tumor virus (MMTV) genomic RNA (gRNA) requires the entire 5' untranslated region (UTR) in conjunction with the first 120 nucleotides of the gag gene. This region includes several palindromic (pal) sequence(s) and stable stem loops (SLs). Among these, stem loop 4 (SL4) adopts a bifurcated structure consisting of three stems, two apical loops, and an internal loop. Pal II, located in one of the apical loops, mediates gRNA dimerization, a process intricately linked to packaging. We thus hypothesized that the bifurcated SL4 structure could constitute the major gRNA packaging determinant. To test this hypothesis, the two apical loops and the flanking sequences forming the bifurcated SL4 were individually mutated. These mutations all had deleterious effects on gRNA packaging and propagation. Next, single and compensatory mutants were designed to destabilize then recreate the bifurcated SL4 structure. A structure-function analysis using bioinformatics predictions and RNA chemical probing revealed that mutations that led to the loss of the SL4 bifurcated structure abrogated RNA packaging and propagation, while compensatory mutations that recreated the native SL4 structure restored RNA packaging and propagation to wild type levels. Altogether, our results demonstrate that SL4 constitutes the principal packaging determinant of MMTV gRNA. Our findings further suggest that SL4 acts as a structural switch that can not only differentiate between RNA for translation versus packaging/dimerization, but its location also allows differentiation between spliced and unspliced RNAs during gRNA encapsidation.


Asunto(s)
Dimerización , Virus del Tumor Mamario del Ratón/metabolismo , Biosíntesis de Proteínas , ARN Viral/química , ARN Viral/metabolismo , Ensamble de Virus , Animales , Genómica , Virus del Tumor Mamario del Ratón/química , Virus del Tumor Mamario del Ratón/genética , Ratones , Conformación de Ácido Nucleico , ARN Viral/genética
11.
Mol Oncol ; 11(9): 1172-1188, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28557306

RESUMEN

Oncogenic signalling via Ca2+ -activated K+ channels of intermediate conductance (SK4, also known as KCa 3.1 or IK) has been implicated in different cancer entities including breast cancer. Yet, the role of endogenous SK4 channels for tumorigenesis is unclear. Herein, we generated SK4-negative tumours by crossing SK4-deficient (SK4 KO) mice to the polyoma middle T-antigen (PyMT) and epidermal growth factor receptor 2 (cNeu) breast cancer models in which oncogene expression is driven by the retroviral promoter MMTV. Survival parameters and tumour progression were studied in cancer-prone SK4 KO in comparison with wild-type (WT) mice and in a syngeneic orthotopic mouse model following transplantation of SK4-negative or WT tumour cells. SK4 activity was modulated by genetic or pharmacological means using the SK4 inhibitor TRAM-34 in order to establish the role of breast tumour SK4 for cell growth, electrophysiological signalling, and [Ca2+ ]i oscillations. Ablation of SK4 and TRAM-34 treatment reduced the SK4-generated current fraction, growth factor-dependent Ca2+ entry, cell cycle progression and the proliferation rate of MMTV-PyMT tumour cells. In vivo, PyMT oncogene-driven tumorigenesis was only marginally affected by the global lack of SK4, whereas tumour progression was significantly delayed after orthotopic implantation of MMTV-PyMT SK4 KO breast tumour cells. However, overall survival and progression-free survival time in the MMTV-cNeu mouse model were significantly extended in the absence of SK4. Collectively, our data from murine breast cancer models indicate that SK4 activity is crucial for cell cycle control. Thus, the modulation of this channel should be further investigated towards a potential improvement of existing antitumour strategies in human breast cancer.


Asunto(s)
Señalización del Calcio , Ciclo Celular , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Animales , Calcio/metabolismo , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Femenino , Espacio Intracelular/metabolismo , Estimación de Kaplan-Meier , Masculino , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Factores de Tiempo
12.
Development ; 144(6): 1072-1086, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28100467

RESUMEN

The ubiquitin ligases CBL and CBL-B are negative regulators of tyrosine kinase signaling with established roles in the immune system. However, their physiological roles in epithelial tissues are unknown. Here, we used MMTV-Cre-mediated Cbl gene deletion on a Cbl-b null background, as well as a tamoxifen-inducible mammary stem cell (MaSC)-specific Cbl and Cbl-b double knockout (Cbl/Cbl-b DKO) using Lgr5-EGFP-IRES-CreERT2, to demonstrate a mammary epithelial cell-autonomous requirement of CBL and CBL-B in the maintenance of MaSCs. Using a newly engineered tamoxifen-inducible Cbl and Cbl-b deletion model with a dual fluorescent reporter (Cblflox/flox; Cbl-bflox/flox; Rosa26-CreERT; mT/mG), we show that Cbl/Cbl-b DKO in mammary organoids leads to hyperactivation of AKT-mTOR signaling with depletion of MaSCs. Chemical inhibition of AKT or mTOR rescued MaSCs from Cbl/Cbl-b DKO-induced depletion. Our studies reveal a novel, cell-autonomous requirement of CBL and CBL-B in epithelial stem cell maintenance during organ development and remodeling through modulation of mTOR signaling.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Glándulas Mamarias Animales/citología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Células Madre/citología , Animales , Autorrenovación de las Células/efectos de los fármacos , Separación Celular , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Eliminación de Gen , Integrasas/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Ratones Noqueados , Organoides/citología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Células Madre/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tamoxifeno/farmacología
13.
Virology ; 487: 121-8, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26517399

RESUMEN

The HERV-K(HML-2) family is the most recent addition to the collection of human endogenous retroviruses. It comprises proviruses that encode functional proteins that can assemble into replication defective particles carrying the envelope protein. Using a reconstituted HERV-K113 envelope sequence, we have analyzed its ability to mediate entry into a set of 33 cell lines from 10 species. Of these, 30 were permissive, demonstrating an amphotropism consistent with a broad expression of receptor protein(s). In an initial effort to identify a receptor for HERV-K(HML-2) we investigated whether transferrin receptor 1 and hyaluronidase 2, known cellular receptors of the closely related betaretroviruses mouse mammary tumor virus (MMTV) and Jaagsiekte sheep retrovirus (JSRV), could facilitate HERV-K(HML-2) entry. However, neither of these proteins could serve as a receptor for HERV-K(HML-2). Moreover, during attempts to further characterize the tropism of HERV-K(HML-2), we identified a cellular activity that inhibits infection at a post-entry, pre-integration step.


Asunto(s)
Antígenos CD/metabolismo , Moléculas de Adhesión Celular/metabolismo , Retrovirus Endógenos/metabolismo , Hialuronoglucosaminidasa/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Tropismo Viral/fisiología , Células 3T3 , Animales , Betaretrovirus/metabolismo , Células COS , Gatos , Línea Celular Tumoral , Chlorocebus aethiops , Perros , Proteínas Ligadas a GPI/metabolismo , Células HEK293 , Células HeLa , Humanos , Retrovirus Ovino Jaagsiekte/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Receptores Virales , Células Vero , Internalización del Virus
14.
Oncotarget ; 6(21): 18355-63, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26214095

RESUMEN

Etiology of human breast cancer is unknown, whereas the Mouse Mammary Tumor Virus (MMTV) is recognized as the etiologic agent of mouse mammary carcinoma. Moreover, this experimental model contributed substantially to our understanding of many biological aspects of the human disease. Several data strongly suggest a causative role of MMTV in humans, such as the presence of viral sequences in a high percentage of infiltrating breast carcinoma and in its preinvasive lesions, the production of viral particles in primary cultures of breast cancer, the ability of the virus to infect cells in culture. This paper demonstrates that MMTV is present in human saliva and salivary glands. MMTV presence was investigated by fluorescent PCR, RT-PCR, FISH, immunohistochemistry, and whole transcriptome analysis. Saliva was obtained from newborns, children, adults, and breast cancer patients. The saliva of newborns is MMTV-free, whereas MMTV is present in saliva of children (26.66%), healthy adults (10.60%), and breast cancer patients (57.14% as DNA and 33.9% as RNA). MMTV is also present in 8.10% of salivary glands. RNA-seq analysis performed on saliva of a breast cancer patient demonstrates a high expression of MMTV RNA in comparison to negative controls. The possibility of a contamination by murine DNA was excluded by murine mtDNA and IAP LTR PCR. These findings confirm the presence of MMTV in humans, strongly suggest saliva as route in inter-human infection, and support the hypothesis of a viral origin for human breast carcinoma.


Asunto(s)
Virus del Tumor Mamario del Ratón/fisiología , Infecciones por Retroviridae/virología , Saliva/virología , Infecciones Tumorales por Virus/virología , Adulto , Animales , Neoplasias de la Mama/virología , Femenino , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Lactante , Recién Nacido , Masculino , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Persona de Mediana Edad , Infecciones por Retroviridae/transmisión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Glándulas Salivales/virología , Infecciones Tumorales por Virus/transmisión
15.
Mol Cancer ; 14: 115, 2015 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-26047945

RESUMEN

BACKGROUND: The expression of the chemokine receptor CCR6 has been previously correlated with higher grades and stages of breast cancer and decreased relapse-free survival. Also, its cognate chemokine ligand CCL20 has been reported to induce proliferation of cultured human breast epithelial cells. METHODS: To establish if CCR6 plays a functional role in mammary tumorigenesis, a bigenic MMTV-PyMT CCR6-null mouse was generated and mammary tumor development was assessed. Levels of tumor-infiltrating immune cells within tumor-bearing mammary glands from MMTV-PyMT Ccr6 (WT) and Ccr6 (-/-) mice were also analyzed. RESULTS: Deletion of CCR6 delayed tumor onset, significantly reduced the extent of initial hyperplastic outgrowth, and decreased tumor incidence in PyMT transgenic mice. CCR6 was then shown to promote the recruitment of pro-tumorigenic macrophages to the tumor site, facilitating the onset of neoplasia. CONCLUSIONS: This study delineated for the first time a role for CCR6 in the development of breast cancer, and demonstrated a critical function for this receptor in maintaining the pro-tumorigenic cancer microenvironment.


Asunto(s)
Antígenos Transformadores de Poliomavirus/metabolismo , Macrófagos/metabolismo , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Receptores CCR6/metabolismo , Animales , Carcinogénesis/patología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Eliminación de Gen , Macrófagos/patología , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Microambiente Tumoral
16.
PLoS One ; 10(6): e0131515, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26121257

RESUMEN

The Gag protein of the mouse mammary tumor virus (MMTV) is the chief determinant of subcellular targeting. Electron microscopy studies show that MMTV Gag forms capsids within the cytoplasm and assembles as immature particles with MMTV RNA and the Y box binding protein-1, required for centrosome maturation. Other betaretroviruses, such as Mason-Pfizer monkey retrovirus (M-PMV), assemble adjacent to the pericentriolar region because of a cytoplasmic targeting and retention signal in the Matrix protein. Previous studies suggest that the MMTV Matrix protein may also harbor a similar cytoplasmic targeting and retention signal. Herein, we show that a substantial fraction of MMTV Gag localizes to the pericentriolar region. This was observed in HEK293T, HeLa human cell lines and the mouse derived NMuMG mammary gland cells. Moreover, MMTV capsids were observed adjacent to centrioles when expressed from plasmids encoding either MMTV Gag alone, Gag-Pro-Pol or full-length virus. We found that the cytoplasmic targeting and retention signal in the MMTV Matrix protein was sufficient for pericentriolar targeting, whereas mutation of the glutamine to alanine at position 56 (D56/A) resulted in plasma membrane localization, similar to previous observations from mutational studies of M-PMV Gag. Furthermore, transmission electron microscopy studies showed that MMTV capsids accumulate around centrioles suggesting that, similar to M-PMV, the pericentriolar region may be a site for MMTV assembly. Together, the data imply that MMTV Gag targets the pericentriolar region as a result of the MMTV cytoplasmic targeting and retention signal, possibly aided by the Y box protein-1 required for the assembly of centrosomal microtubules.


Asunto(s)
Centriolos/metabolismo , Productos del Gen gag/metabolismo , Virus del Tumor Mamario del Ratón/metabolismo , Regiones no Traducidas 5'/genética , Secuencia de Aminoácidos , Animales , Cápside/ultraestructura , Productos del Gen gag/química , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Ratones , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Secuencias Reguladoras de Ácidos Nucleicos/genética
17.
PLoS One ; 10(1): e0117239, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25635772

RESUMEN

While most breast cancers are thought to arise from the luminal layer of the breast tissue, it remains unclear which specific cells in the luminal layer are the cells of origin of breast cancer. We have previously reported that WAP-positive luminal epithelial cells are at increased susceptibility to tumor initiation by ErbB2 compared to the bulk population, while the mammary cells with canonical Wnt signaling activity fail to evolve into tumors upon ErbB2 activation. Here, we used retrovirus to introduce ErbB2 into the Krt6a-positive mammary progenitor subset of the luminal epithelium and, for comparison, into the mammary luminal epithelium indiscriminately. Tumors developed from both groups of cells with a similar latency. These data indicate that the Krt6a-positive subset of mammary epithelial cells can be induced to form cancer by ErbB2 but it is not more susceptible to tumorigenesis initiated by ErbB2 than the bulk population of the luminal epithelium.


Asunto(s)
Carcinogénesis/patología , Células Epiteliales/metabolismo , Queratina-6/metabolismo , Glándulas Mamarias Animales/patología , Receptor ErbB-2/metabolismo , Células Madre/metabolismo , Animales , Apoptosis , Carcinogénesis/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/patología , Femenino , Humanos , Virus del Tumor Mamario del Ratón/metabolismo , Ratones Transgénicos , Fosforilación , Retroviridae/metabolismo , Factor de Transcripción STAT5/metabolismo , Células Madre/patología , Virión/metabolismo , Latencia del Virus
18.
PLoS One ; 9(5): e97666, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24831228

RESUMEN

Mammary epithelial (ME) cells cultured under conventional conditions senesce after several passages. Here, we demonstrate that mouse ME cells isolated from normal mammary glands or from mouse mammary tumor virus (MMTV)-Neu-induced mammary tumors, can be cultured indefinitely as conditionally reprogrammed cells (CRCs) on irradiated fibroblasts in the presence of the Rho kinase inhibitor Y-27632. Cell surface progenitor-associated markers are rapidly induced in normal mouse ME-CRCs relative to ME cells. However, the expression of certain mammary progenitor subpopulations, such as CD49f+ ESA+ CD44+, drops significantly in later passages. Nevertheless, mouse ME-CRCs grown in a three-dimensional extracellular matrix gave rise to mammary acinar structures. ME-CRCs isolated from MMTV-Neu transgenic mouse mammary tumors express high levels of HER2/neu, as well as tumor-initiating cell markers, such as CD44+, CD49f+, and ESA+ (EpCam). These patterns of expression are sustained in later CRC passages. Early and late passage ME-CRCs from MMTV-Neu tumors that were implanted in the mammary fat pads of syngeneic or nude mice developed vascular tumors that metastasized within 6 weeks of transplantation. Importantly, the histopathology of these tumors was indistinguishable from that of the parental tumors that develop in the MMTV-Neu mice. Application of the CRC system to mouse mammary epithelial cells provides an attractive model system to study the genetics and phenotype of normal and transformed mouse epithelium in a defined culture environment and in vivo transplant studies.


Asunto(s)
Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/metabolismo , Amidas/química , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Técnicas de Cocultivo , Colágeno/química , Hibridación Genómica Comparativa , Combinación de Medicamentos , Inhibidores Enzimáticos/química , Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/metabolismo , Laminina/química , Ratones , Ratones Transgénicos , Microscopía Confocal , Fenotipo , Proteoglicanos/química , Piridinas/química , Células Madre/citología
19.
PLoS One ; 9(4): e92735, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24718286

RESUMEN

Fibroblast growth factor (FGF) signaling is essential for vertebrate organogenesis, including mammary gland development. The mechanism whereby FGF signaling is regulated in the mammary gland, however, has remained unknown. Using a combination of mouse genetics and 3D ex vivo models, we tested the hypothesis that Spry2 gene, which encodes an inhibitor of signaling via receptor tyrosine kinases (RTKs) in certain contexts, regulates FGF signaling during mammary branching. We found that Spry2 is expressed at various stages of the developing mammary gland. Targeted removal of Spry2 function from mammary epithelium leads to accelerated epithelial invasion. Spry2 is up-regulated by FGF signaling activities and its loss sensitizes mammary epithelium to FGF stimulation, as indicated by increased expression of FGF target genes and epithelia invasion. By contrast, Spry2 gain-of-function in the mammary epithelium results in reduced FGF signaling, epithelial invasion, and stunted branching. Furthermore, reduction of Spry2 expression is correlated with tumor progression in the MMTV-PyMT mouse model. Together, the data show that FGF signaling modulation by Spry2 is essential for epithelial morphogenesis in the mammary gland and it functions to protect the epithelium against tumorigenesis.


Asunto(s)
Epitelio/crecimiento & desarrollo , Epitelio/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Morfogénesis , Transducción de Señal , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Femenino , Heterocigoto , Humanos , Integrasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Virus del Tumor Mamario del Ratón/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas
20.
Glycobiology ; 23(12): 1477-90, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24037315

RESUMEN

Bisected, complex N-glycans on glycoproteins are generated by the glycosyltransferase MGAT3 and cause reduced cell surface binding of galectins. Previously, we showed that MGAT3 reduces growth factor signaling and retards mammary tumor progression driven by the Polyoma middle T antigen (PyMT) expressed in mammary epithelium under the mouse mammary tumor virus (MMTV) promoter. However, the penetrance of the tumor phenotype became variable in mixed FVB/N and C57BL/6 female mice and we therefore investigated a congenic C57BL/6 Mgat3(-/-)/MMTV-PyMT model. In the absence of MGAT3, C57BL/6 Mgat3(-/-)/MMTV-PyMT females exhibited accelerated tumor appearance and increased tumor burden, glucose uptake in tumors and lung metastasis. Nevertheless, activation of extracellular signal-regulated kinase (ERK)1/2 or protein kinase B (AKT) was reduced in ∼20-week C57BL/6 MMTV-PyMT tumors lacking MGAT3. Activation of focal adhesion kinase (FAK), protein tyrosine kinase Src, and p38 mitogen-activated protein kinase were similar to that of controls. All the eight mouse galectin genes were expressed in mammary tumors and tumor epithelial cells (TECs), but galectin-2 and -12 were not detected by western analysis in tumors, and galectin-7 was not detected in 60% of the TEC lines. From microarray data reported for human breast cancers, at least 10 galectin and 7 N-glycan N-acetylglucosaminyl (GlcNAc)-transferase (MGAT) genes are expressed in tumor tissue, and expression often varies significantly between different breast cancer subtypes. Thus, in summary, while MGAT3 and bisected complex N-glycans retard mouse mammary tumor progression, genetic background may modify this effect; identification of key galectins that promote mammary tumor progression in mice is not straightforward because all the eight galectin genes are expressed; and high levels of MGAT3, galectin-4, -8, -10, -13 and -14 transcripts correlate with better relapse-free survival in human breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Galectinas/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Polisacáridos/metabolismo , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Neoplasias de la Mama/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/metabolismo , Ratones , Ratones Endogámicos , N-Acetilglucosaminiltransferasas/deficiencia , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA