Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
1.
Oncogene ; 40(1): 12-27, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33046799

RESUMO

Despite major progress in breast cancer research, the functional contribution of distinct cancer cell clones to malignant tumor progression and metastasis remains largely elusive. We have assessed clonal heterogeneity within individual primary tumors and metastases and also during the distinct stages of malignant tumor progression using clonal tracking of cancer cells in the MMTV-PyMT mouse model of metastatic breast cancer. Comparative gene expression analysis of clonal subpopulations reveals a substantial level of heterogeneity across and also within the various stages of breast carcinogenesis. The intra-stage heterogeneity is primarily manifested by differences in cell proliferation, also found within invasive carcinomas of luminal A-, luminal B-, and HER2-enriched human breast cancer. Surprisingly, in the mouse model of clonal tracing of cancer cells, chemotherapy mainly targets the slow-proliferative clonal populations and fails to efficiently repress the fast-proliferative populations. These insights may have considerable impact on therapy selection and response in breast cancer patients.


Assuntos
Neoplasias da Mama/patologia , Rastreamento de Células/métodos , Perfilação da Expressão Gênica/métodos , Neoplasias Mamárias Experimentais/patologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Receptor ErbB-2/genética , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células , Evolução Clonal , Progressão da Doença , Feminino , Redes Reguladoras de Genes , Humanos , Microdissecção e Captura a Laser , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Análise de Sequência de RNA
2.
Oxid Med Cell Longev ; 2019: 6016278, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31885806

RESUMO

HIV-induced immune suppression results in the high prevalence of HIV/AIDS-associated malignancies including Kaposi sarcoma, non-Hodgkin lymphoma, and cervical cancer. HIV-infected people are also at an increased risk of "non-AIDS-defining" malignancies not directly linked to immune suppression but associated with viral infections. Their incidence is increasing despite successful antiretroviral therapy. The mechanism behind this phenomenon remains unclear. Here, we obtained daughter clones of murine mammary gland adenocarcinoma 4T1luc2 cells expressing consensus reverse transcriptase of HIV-1 subtype A FSU_A strain (RT_A) with and without primary mutations of drug resistance. In in vitro tests, mutations of resistance to nucleoside inhibitors K65R/M184V reduced the polymerase, and to nonnucleoside inhibitors K103N/G190S, the RNase H activities of RT_A. Expression of these RT_A variants in 4T1luc2 cells led to increased production of the reactive oxygen species (ROS), lipid peroxidation, enhanced cell motility in the wound healing assay, and upregulation of expression of Vimentin and Twist. These properties, particularly, the expression of Twist, correlated with the levels of expression RT_A and/or the production of ROS. When implanted into syngeneic BALB/C mice, 4T1luc2 cells expressing nonmutated RT_A demonstrated enhanced rate of tumor growth and increased metastatic activity, dependent on the level of expression of RT_A and Twist. No enhancement was observed for the clones expressing mutated RT_A variants. Plausible mechanisms are discussed involving differential interactions of mutated and nonmutated RTs with its cellular partners involved in the regulation of ROS. This study establishes links between the expression of HIV-1 RT, production of ROS, induction of EMT, and enhanced propagation of RT-expressing tumor cells. Such scenario can be proposed as one of the mechanisms of HIV-induced/enhanced carcinogenesis not associated with immune suppression.


Assuntos
Adenocarcinoma/virologia , Neoplasias da Mama/virologia , Infecções por HIV/metabolismo , Transcriptase Reversa do HIV/metabolismo , HIV-1/metabolismo , Neoplasias Mamárias Experimentais/virologia , Proteína 1 Relacionada a Twist/metabolismo , Animais , Carcinogênese , Processos de Crescimento Celular , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Infecções por HIV/patologia , Transcriptase Reversa do HIV/genética , HIV-1/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Mutação/genética , Metástase Neoplásica , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 Relacionada a Twist/genética , Regulação para Cima
3.
mBio ; 10(4)2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31409681

RESUMO

Complex human-pathogenic retroviruses cause high morbidity and mortality worldwide, but resist antiviral drugs and vaccine development due to evasion of the immune response. A complex retrovirus, mouse mammary tumor virus (MMTV), requires replication in B and T lymphocytes for mammary gland transmission and is antagonized by the innate immune restriction factor murine Apobec3 (mA3). To determine whether the regulatory/accessory protein Rem affects innate responses to MMTV, a splice-donor mutant (MMTV-SD) lacking Rem expression was injected into BALB/c mice. Mammary tumors induced by MMTV-SD had a lower proviral load, lower incidence, and longer latency than mammary tumors induced by wild-type MMTV (MMTV-WT). MMTV-SD proviruses had many G-to-A mutations on the proviral plus strand, but also C-to-T transitions within WRC motifs. Similarly, a lymphomagenic MMTV variant lacking Rem expression showed decreased proviral loads and increased WRC motif mutations relative to those in wild-type-virus-induced tumors, consistent with activation-induced cytidine deaminase (AID) mutagenesis in lymphoid cells. These mutations are typical of the Apobec family member AID, a B-cell-specific mutagenic protein involved in antibody variable region hypermutation. In contrast, mutations in WRC motifs and proviral loads were similar in MMTV-WT and MMTV-SD proviruses from tumors in AID-insufficient mice. AID was not packaged in MMTV virions. Rem coexpression in transfection experiments led to AID proteasomal degradation. Our data suggest that rem specifies a human-pathogenic immunodeficiency virus type 1 (HIV-1) Vif-like protein that inhibits AID and antagonizes innate immunity during MMTV replication in lymphocytes.IMPORTANCE Complex retroviruses, such as human-pathogenic immunodeficiency virus type 1 (HIV-1), cause many human deaths. These retroviruses produce lifelong infections through viral proteins that interfere with host immunity. The complex retrovirus mouse mammary tumor virus (MMTV) allows for studies of host-pathogen interactions not possible in humans. A mutation preventing expression of the MMTV Rem protein in two different MMTV strains decreased proviral loads in tumors and increased viral genome mutations typical of an evolutionarily ancient enzyme, AID. Although the presence of AID generally improves antibody-based immunity, it may contribute to human cancer progression. We observed that coexpression of MMTV Rem and AID led to AID destruction. Our results suggest that Rem is the first known protein inhibitor of AID and that further experiments could lead to new disease treatments.


Assuntos
Citidina Desaminase/antagonistas & inibidores , Vírus do Tumor Mamário do Camundongo/genética , Provírus/genética , Proteínas Virais Reguladoras e Acessórias/genética , Animais , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Feminino , Imunidade Inata , Masculino , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutação , Provírus/fisiologia , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia , Carga Viral/genética , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral
4.
Breast Cancer Res ; 20(1): 138, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30458886

RESUMO

BACKGROUND: Early analyses of human breast cancer identified high expression of the insulin-like growth factor type 1 receptor (IGF-1R) correlated with hormone receptor positive breast cancer and associated with a favorable prognosis, whereas low expression of IGF-1R correlated with triple negative breast cancer (TNBC). We previously demonstrated that the IGF-1R acts as a tumor and metastasis suppressor in the Wnt1 mouse model of TNBC. The mechanisms for how reduced IGF-1R contributes to TNBC phenotypes is unknown. METHODS: We analyzed the METABRIC dataset to further stratify IGF-1R expression with patient survival and specific parameters of TNBC. To investigate molecular events associated with the loss of IGF-1R function in breast tumor cells, we inhibited IGF-1R in human cell lines using an IGF-1R blocking antibody and analyzed MMTV-Wnt1-mediated mouse tumors with reduced IGF-1R function through expression of a dominant-negative transgene. RESULTS: Our analysis of the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset revealed association between low IGF-1R and reduced overall patient survival. IGF-1R expression was inversely correlated with patient survival even within hormone receptor-positive breast cancers, indicating reduced overall patient survival with low IGF-1R was not due simply to low IGF-1R expression within TNBCs. Inhibiting IGF-1R in either mouse or human tumor epithelial cells increased reactive oxygen species (ROS) production and activation of the endoplasmic reticulum stress response. IGF-1R inhibition in tumor epithelial cells elevated interleukin (IL)-6 and C-C motif chemokine ligand 2 (CCL2) expression, which was reversed by ROS scavenging. Moreover, the Wnt1/dnIGF-1R primary tumors displayed a tumor-promoting immune phenotype. The increased CCL2 promoted an influx of CD11b+ monocytes into the primary tumor that also had increased matrix metalloproteinase (MMP)-2, MMP-3, and MMP-9 expression. Increased MMP activity in the tumor stroma was associated with enhanced matrix remodeling and collagen deposition. Further analysis of the METABRIC dataset revealed an increase in IL-6, CCL2, and MMP-9 expression in patients with low IGF-1R, consistent with our mouse tumor model and data in human breast cancer cell lines. CONCLUSIONS: Our data support the hypothesis that reduction of IGF-1R function increases cellular stress and cytokine production to promote an aggressive tumor microenvironment through infiltration of immune cells and matrix remodeling.


Assuntos
Citocinas/metabolismo , Estresse do Retículo Endoplasmático , Neoplasias Mamárias Experimentais/patologia , Receptores de Somatomedina/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Animais , Linhagem Celular Tumoral , Conjuntos de Dados como Assunto , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Transgênicos , Receptor IGF Tipo 1 , Transdução de Sinais , Microambiente Tumoral , Proteína Wnt1/genética
5.
Breast Cancer Res ; 20(1): 131, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30367629

RESUMO

BACKGROUND: Amphiregulin (AREG), a ligand of the epidermal growth factor receptor, is not only essential for proper mammary ductal development, but also associated with breast cancer proliferation and growth. In the absence of AREG, mammary ductal growth is stunted and fails to expand. Furthermore, suppression of AREG expression in estrogen receptor-positive breast tumor cells inhibits in-vitro and in-vivo growth. METHODS: We crossed AREG-null (AREG-/-) mice with the murine luminal B breast cancer model, MMTV-PyMT (PyMT), to generate spontaneous breast tumors that lack AREG (AREG-/- PyMT). We evaluated tumor growth, cytokeratin-8 (K8)-positive luminal cells, cytokeratin-14 (K14)-positive myoepithelial cells, and expression of AREG, Ki67, and PyMT. Primary myoepithelial cells from nontumor-bearing AREG+/+ mice underwent fluorescence-activated cell sorting and were adapted to culture for in-vitro coculture studies with AT-3 cells, a cell line derived from C57Bl/6 PyMT mammary tumors. RESULTS: Intriguingly, PyMT-induced lesions progress more rapidly in AREG-/- mice than in AREG+/+ mice. Quantification of K8+ luminal and K14+ myoepithelial cells in non-PyMT AREG-/- mammary glands showed fewer K14+ cells and a thinner myoepithelial layer. Study of AT-3 cells indicated that coculture with myoepithelial cells or exposure to AREG, epidermal growth factor, or basic fibroblast growth factor can suppress PyMT expression. Late-stage AREG-/- PyMT tumors are significantly less solid in structure, with more areas of papillary and cystic growth. Papillary areas appear to be both less proliferative and less necrotic. In The Cancer Genome Atlas database, luminal-B invasive papillary carcinomas have lower AREG expression than luminal B invasive ductal carcinomas. CONCLUSIONS: Our study has revealed a previously unknown role of AREG in myoepithelial cell development and PyMT expression. AREG expression is essential for proper myoepithelial coverage of mammary ducts. Both AREG and myoepithelial cells can suppress PyMT expression. We find that lower AREG expression is associated with invasive papillary breast cancer in both the MMTV-PyMT model and human breast cancer.


Assuntos
Anfirregulina/metabolismo , Células Epiteliais/patologia , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Anfirregulina/genética , Animais , Antígenos Transformantes de Poliomavirus/genética , Antígenos Transformantes de Poliomavirus/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Células Epiteliais/virologia , Feminino , Humanos , Glândulas Mamárias Animais/citologia , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Invasividade Neoplásica/patologia , Polyomavirus/genética , Polyomavirus/imunologia
6.
Nat Commun ; 9(1): 1368, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29636461

RESUMO

Formate overflow coupled to mitochondrial oxidative metabolism\ has been observed in cancer cell lines, but whether that takes place in the tumor microenvironment is not known. Here we report the observation of serine catabolism to formate in normal murine tissues, with a relative rate correlating with serine levels and the tissue oxidative state. Yet, serine catabolism to formate is increased in the transformed tissue of in vivo models of intestinal adenomas and mammary carcinomas. The increased serine catabolism to formate is associated with increased serum formate levels. Finally, we show that inhibition of formate production by genetic interference reduces cancer cell invasion and this phenotype can be rescued by exogenous formate. We conclude that increased formate overflow is a hallmark of oxidative cancers and that high formate levels promote invasion via a yet unknown mechanism.


Assuntos
Adenoma/metabolismo , Formiatos/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Intestinais/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Serina/metabolismo , Adenoma/genética , Adenoma/patologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Formiatos/farmacologia , Glicina Hidroximetiltransferase/genética , Glicina Hidroximetiltransferase/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/genética , Neoplasias Intestinais/patologia , Intestinos/patologia , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Glândulas Mamárias Animais/metabolismo , Glândulas Mamárias Animais/patologia , Glândulas Mamárias Animais/virologia , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Metotrexato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Oxirredução , Microambiente Tumoral/efeitos dos fármacos
7.
Cancer Res ; 78(6): 1484-1496, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29330144

RESUMO

In breast cancer, increased expression of the cytoskeletal adaptor protein Kindlin-1 has been linked to increased risks of lung metastasis, but the functional basis is unknown. Here, we show that in a mouse model of polyomavirus middle T antigen-induced mammary tumorigenesis, loss of Kindlin-1 reduced early pulmonary arrest and later development of lung metastasis. This phenotype relied on the ability of Kindlin-1 to bind and activate ß integrin heterodimers. Kindlin-1 loss reduced α4 integrin-mediated adhesion of mammary tumor cells to the adhesion molecule VCAM-1 on endothelial cells. Treating mice with an anti-VCAM-1 blocking antibody prevented early pulmonary arrest. Kindlin-1 loss also resulted in reduced secretion of several factors linked to metastatic spread, including the lung metastasis regulator tenascin-C, showing that Kindlin-1 regulated metastatic dissemination by an additional mechanism in the tumor microenvironment. Overall, our results show that Kindlin-1 contributes functionally to early pulmonary metastasis of breast cancer.Significance: These findings provide a mechanistic proof in mice that Kindin-1, an integrin-binding adaptor protein, is a critical mediator of early lung metastasis of breast cancer. Cancer Res; 78(6); 1484-96. ©2018 AACR.


Assuntos
Neoplasias da Mama/patologia , Proteínas de Transporte/metabolismo , Neoplasias Pulmonares/secundário , Animais , Anticorpos Monoclonais/farmacologia , Antígenos Transformantes de Poliomavirus/toxicidade , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Transporte/genética , Adesão Celular/efeitos dos fármacos , Células Endoteliais/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Integrinas/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/virologia , Camundongos Transgênicos , Molécula 1 de Adesão de Célula Vascular/imunologia
8.
Virology ; 513: 180-187, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29096160

RESUMO

Mouse mammary tumor virus (MMTV) induces breast cancer in mice in the absence of known virally-encoded oncogenes. Tumorigenesis by MMTV is thought to occur primarily through insertional mutagenesis, leading to the activation of cellular proto-oncogenes and outgrowth of selected cells. Here we investigated whether MMTV encodes microRNAs (miRNAs) and/or modulates host miRNAs that could contribute to tumorigenesis. High throughput small RNA sequencing analysis of MMTV-infected cells and MMTV-induced mammary tumors demonstrates that MMTV does not encode miRNAs. However, infected tissues have altered levels of several host miRNAs, including increased expression of members of the oncogenic miRNA cluster, miR-17-92. Notably, similar changes in miRNA levels have been previously reported in human breast cancers. Combined, our results demonstrate that virally encoded miRNAs do not contribute to MMTV-mediated tumorigenesis, but that changes in specific host miRNAs in infected cells may contribute to virus replication and tumor biology.


Assuntos
Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/fisiologia , MicroRNAs/análise , Animais , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos
9.
J Virol Methods ; 245: 28-34, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28315379

RESUMO

Semliki Forest virus (SFV) is a potential cancer gene therapy vector capable of providing high and transient expression of heterologous proteins in mammalian cells. However, SFV has shown suboptimal transduction levels in several cancer cell types as well as wide biodistribution of SFV has been observed after in vivo applications. Magnetic nanoparticles (MNPs) have been shown to increase cell transduction with several viral vectors in vitro under an external magnetic field and enhance magnetically guided viral vector delivery. Here, we examined a panel of MNPs for enhanced cancer cell transduction with SFV vector. Magneto-transduction using positively charged MNPs increased Semliki Forest virus transduction in TS/A mouse mammary carcinoma cells in vitro in the presence of fetal bovine serum. Positively charged MNPs efficiently captured SFV particles independently of capturing medium, and MNPs-SFV complexes were successfully separated from suspension by magnetic precipitation. These results reveal the potential application of MNPs for enhanced gene delivery by SFV vector as well as proposes magnetic precipitation for efficient concentration of SFV particles from different media.


Assuntos
Nanopartículas de Magnetita , Vírus da Floresta de Semliki/genética , Transdução Genética/métodos , Animais , Bovinos , Linhagem Celular Tumoral , Feminino , Técnicas de Transferência de Genes , Terapia Genética/métodos , Vetores Genéticos , Neoplasias Mamárias Experimentais/virologia , Camundongos
10.
Mol Cancer ; 16(1): 19, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28137279

RESUMO

BACKGROUND: Breast cancer is the major cause of cancer-related mortality in women. It is thought that quiescent stem-like cells within solid tumors are responsible for cancer maintenance, progression and eventual metastasis. We recently reported that the chemokine receptor CCR7, a multi-functional regulator of breast cancer, maintains the stem-like cell population. METHODS: This study used a combination of molecular and cellular assays on primary mammary tumor cells from the MMTV-PyMT transgenic mouse with or without CCR7 to examine the signaling crosstalk between CCR7 and Notch pathways. RESULTS: We show for the first time that CCR7 functionally intersects with the Notch signaling pathway to regulate mammary cancer stem-like cells. In this cell subpopulation, CCR7 stimulation activated the Notch signaling pathway, and deletion of CCR7 significantly reduced the levels of activated cleaved Notch1. Moreover, blocking Notch activity prevented specific ligand-induced signaling of CCR7 and augmentation of mammary cancer stem-like cell function. CONCLUSION: Crosstalk between CCR7 and Notch1 promotes stemness in mammary cancer cells and may ultimately potentiate mammary tumor progression. Therefore, dual targeting of both the CCR7 receptor and Notch1 signaling axes may be a potential therapeutic avenue to specifically inhibit the functions of breast cancer stem cells.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Células-Tronco Neoplásicas/metabolismo , Receptor Notch1/metabolismo , Receptores CCR7/genética , Animais , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Camundongos , Camundongos Transgênicos , Receptor Notch1/genética , Receptores CCR7/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
11.
Med Oncol ; 34(3): 43, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28185165

RESUMO

Breast cancer (BC) is the most common type of cancer among women and is the second most common cause of cancer-related deaths, following lung cancer. Severe toxicity associated with a long-term use of BC chemo- and radiotherapy makes it essential to look for newer therapeutics. Additionally, molecular heterogeneity at both intratumoral and intertumoral levels among BC subtypes is known to result in a differential response to standard therapeutics. Oncolytic viruses (OVs) have emerged as one of the most promising treatment options for BC. Many preclinical and clinical studies have shown that OVs are effective in treating BC, both as a single therapeutic agent and as a part of combination therapies. Combination therapies involving multimodal therapeutics including OVs are becoming popular as they allow to achieve the synergistic therapeutic effects, while minimizing the associated toxicities. Here, we review the OVs for BC therapy in preclinical studies and in clinical trials, both as a monotherapy and as part of a combination therapy. We also briefly discuss the potential therapeutic targets for BC, as these are likely to be critical for the development of new OVs.


Assuntos
Neoplasias da Mama/terapia , Neoplasias da Mama/virologia , Terapia Viral Oncolítica/métodos , Animais , Feminino , Humanos , Neoplasias Mamárias Experimentais/terapia , Neoplasias Mamárias Experimentais/virologia
12.
BMC Genomics ; 18(1): 185, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28212608

RESUMO

BACKGROUND: Malignant breast cancer with complex molecular mechanisms of progression and metastasis remains a leading cause of death in women. To improve diagnosis and drug development, it is critical to identify panels of genes and molecular pathways involved in tumor progression and malignant transition. Using the PyMT mouse, a genetically engineered mouse model that has been widely used to study human breast cancer, we profiled and analyzed gene expression from four distinct stages of tumor progression (hyperplasia, adenoma/MIN, early carcinoma and late carcinoma) during which malignant transition occurs. RESULTS: We found remarkable expression similarity among the four stages, meaning genes altered in the later stages showed trace in the beginning of tumor progression. We identified a large number of differentially expressed genes in PyMT samples of all stages compared with normal mammary glands, enriched in cancer-related pathways. Using co-expression networks, we found panels of genes as signature modules with some hub genes that predict metastatic risk. Time-course analysis revealed genes with expression transition when shifting to malignant stages. These may provide additional insight into the molecular mechanisms beyond pathways. CONCLUSIONS: Thus, in this study, our various analyses with the PyMT mouse model shed new light on transcriptomic dynamics during breast cancer malignant progression.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Progressão da Doença , Perfilação da Expressão Gênica , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Vírus do Tumor Mamário do Camundongo/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Expressão Gênica , Redes Reguladoras de Genes , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias
13.
J Natl Cancer Inst ; 109(6)2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28040797

RESUMO

Background: A nontoxic chemopreventive intervention efficacious against different subtypes of breast cancer is still a clinically unmet need. The present study was undertaken to determine the efficacy of an Ayurvedic medicine phytochemical (Withaferin A, [WA]) for chemoprevention of breast cancer and to elucidate its mode of action. Methods: Chemopreventive efficacy of WA (4 and 8 mg/kg body weight) was determined using a rat model of breast cancer induced by N-methyl-N-nitrosourea (MNU; n = 14 for control group, n = 15 for 4 mg/kg group, and n = 18 for 8 mg/kg group). The mechanisms underlying breast cancer chemoprevention by WA were elucidated by immunoblotting, biochemical assays, immunohistochemistry, and cytokine profiling using plasma and tumors from the MNU-rat (n = 8-12 for control group, n = 7-11 for 4 mg/kg group, and n = 8-12 for 8 mg/kg group) and/or mouse mammary tumor virus-neu (MMTV-neu) models (n = 4-11 for control group and n = 4-21 for 4 mg/kg group). Inhibitory effect of WA on exit from mitosis and leptin-induced oncogenic signaling was determined using MCF-7 and/or MDA-MB-231 cells. All statistical tests were two-sided. Results: Incidence, multiplicity, and burden of breast cancer in rats were decreased by WA administration. For example, the tumor weight in the 8 mg/kg group was lower by about 68% compared with controls (8 mg/kg vs control, mean = 2.76 vs 8.59, difference = -5.83, 95% confidence interval of difference = -9.89 to -1.76, P = .004). Mitotic arrest and apoptosis induction were some common determinants of breast cancer chemoprevention by WA in the MNU-rat and MMTV-neu models. Cytokine profiling showed suppression of plasma leptin levels by WA in rats. WA inhibited leptin-induced oncogenic signaling in cultured breast cancer cells. Conclusions: WA is a promising chemopreventative phytochemical with the ability to inhibit at least two different subtypes of breast cancer.


Assuntos
Neoplasias da Mama/prevenção & controle , Neoplasias Mamárias Experimentais/prevenção & controle , Vírus do Tumor Mamário do Camundongo , Infecções por Retroviridae/complicações , Infecções Tumorais por Vírus/complicações , Vitanolídeos/uso terapêutico , 8-Hidroxi-2'-Desoxiguanosina , Acetilcoenzima A/sangue , Família Aldeído Desidrogenase 1 , Animais , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/análise , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/química , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Citocinas/sangue , Desoxiguanosina/análogos & derivados , Desoxiguanosina/análise , Complexo III da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Fatores de Transcrição Forkhead/análise , Humanos , Antígeno Ki-67/análise , Ácido Láctico/sangue , Leptina/sangue , Células MCF-7 , Malatos/sangue , Neoplasias Mamárias Experimentais/química , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/virologia , Metilnitrosoureia , Camundongos , Mitose/efeitos dos fármacos , Índice Mitótico , Ratos , Receptores de Estrogênio/análise , Retinal Desidrogenase/análise , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral , Vitanolídeos/análise , Vitanolídeos/farmacologia
14.
PLoS One ; 11(3): e0151468, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26978528

RESUMO

CD24 is a small, heavily glycosylated, GPI-linked membrane protein, whose expression has been associated with the tumorigenesis and progression of several types of cancer. Here, we studied the expression of CD24 in tumors of MMTV-PyMT, Apc1572/T+ and TRAMP genetic mouse models that spontaneously develop mammary or prostate carcinoma, respectively. We found that CD24 is expressed during tumor development in all three models. In MMTV-PyMT and Apc1572T/+ breast tumors, CD24 was strongly but heterogeneously expressed during early tumorigenesis, but decreased in more advanced stages, and accordingly was increased in poorly differentiated lesions compared with well differentiated lesions. In prostate tumors developing in TRAMP mice, CD24 expression was strong within hyperplastic lesions in comparison with non-hyperplastic regions, and heterogeneous CD24 expression was maintained in advanced prostate carcinomas. To investigate whether CD24 plays a functional role in tumorigenesis in these models, we crossed CD24 deficient mice with MMTV-PyMT, Apc1572T/+ and TRAMP mice, and assessed the influence of CD24 deficiency on tumor onset and tumor burden. We found that mice negative or positive for CD24 did not significantly differ in terms of tumor initiation and burden in the genetic tumor models tested, with the exception of Apc1572T/+ mice, in which lack of CD24 reduced the mammary tumor burden slightly but significantly. Together, our data suggest that while CD24 is distinctively expressed during the early development of murine mammary and prostate tumors, it is not essential for the formation of tumors developing in MMTV-PyMT, Apc1572T/+ and TRAMP mice.


Assuntos
Antígeno CD24/fisiologia , Transformação Celular Neoplásica/genética , Neoplasias Mamárias Experimentais/genética , Síndromes Neoplásicas Hereditárias/genética , Neoplasias da Próstata/genética , Animais , Antígeno CD24/genética , Diferenciação Celular , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Regulação Neoplásica da Expressão Gênica , Genes APC , Masculino , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Síndromes Neoplásicas Hereditárias/etiologia , Próstata/patologia , Infecções por Retroviridae/genética , Glândulas Seminais/patologia , Infecções Tumorais por Vírus/genética
15.
Breast Cancer Res ; 17(1): 132, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26429062

RESUMO

INTRODUCTION: Breast cancer exhibits significant molecular, histological, and pathological diversity. Factors that impact this heterogeneity are poorly understood; however, transformation of distinct normal cell populations of the breast may generate different tumor phenotypes. Our previous study demonstrated that the polyomavirus middle T antigen (PyMT) oncogene can establish diverse tumor subtypes when broadly expressed within mouse mammary epithelial cells. In the present study, we assessed the molecular, histological, and metastatic outcomes in distinct mammary cell populations transformed with the PyMT gene. METHODS: Isolated mouse mammary epithelial cells were transduced with a lentivirus encoding PyMT during an overnight infection and then sorted into hormone receptor-positive luminal (CD133+), hormone receptor-negative luminal (CD133-), basal, and stem cell populations using the cell surface markers CD24, CD49f, and CD133. Each population was subsequently transplanted into syngeneic cleared mouse mammary fat pads to generate tumors. Tumors were classified by histology, estrogen receptor status, molecular subtype, and metastatic potential to investigate whether transformation of different enriched populations affects tumor phenotype. RESULTS: Although enriched mammary epithelial cell populations showed no difference in either the ability to form tumors or tumor latency, differences in prevalence of solid adenocarcinomas and squamous, papillary, and sebaceous-like tumors were observed. In particular, squamous metaplasia was observed more frequently in tumors derived from basal and stem cells than in luminal cells. Interestingly, both molecularly basal and luminal tumors developed from luminal CD133+, basal, and stem cell populations; however, luminal CD133- cells gave rise exclusively to molecularly basal tumors. Tumors arising from the luminal CD133-, basal, and stem cell populations were highly metastatic; however, luminal CD133+ cells generated tumors that were significantly less metastatic, possibly due to an inability of these tumor cells to escape the primary tumor site. CONCLUSIONS: Expression of PyMT within different mammary cell populations influences tumor histology, molecular subtype, and metastatic potential. The data demonstrate that luminal CD133+ cells give rise to less metastatic tumors, luminal CD133- cells preferentially establish basal tumors, and the cell of origin for squamous metaplasia likely resides in the basal and stem cell populations.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Neoplasias Pulmonares/virologia , Neoplasias Mamárias Experimentais/virologia , Neoplasia de Células Basais/virologia , Antígeno AC133 , Animais , Antígenos CD/metabolismo , Transformação Celular Viral , Células Cultivadas , Células Epiteliais/virologia , Feminino , Glicoproteínas/metabolismo , Neoplasias Pulmonares/secundário , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Transplante de Neoplasias , Neoplasia de Células Basais/secundário , Peptídeos/metabolismo , Polyomavirus/genética
16.
Int J Mol Sci ; 16(4): 7655-71, 2015 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-25853264

RESUMO

There is increasing evidence that prenatal exposure to environmental factors may modify breast cancer risk later in life. This study aimed to investigate the effects of in utero exposure to low-dose alcohol on mammary development and tumor risk. Pregnant MMTV-erbB-2 mice were exposed to alcohol (6 g/kg/day) between day 13 and day 19 of gestation, and the female offspring were examined for tumor risk. Whole mount analysis indicated that in utero exposure to low-dose alcohol induced significant increases in ductal extension at 10 weeks of age. Molecular analysis showed that in utero alcohol exposure induced upregulation of ERα signaling and activation of Akt and Erk1/2 in pubertal mammary glands. However, enhanced signaling in the EGFR/erbB-2 pathway appeared to be more prominent in 10-week-old glands than did signaling in the other pathways. Interestingly, tumor development in mice with in utero exposure to low-dose alcohol was slightly delayed compared to control mice, but tumor multiplicity was increased. The results indicate that in utero exposure to low-dose alcohol induces the reprogramming of mammary development by mechanisms that include altered signaling in the estrogen receptor (ER) and erbB-2 pathways. The intriguing tumor development pattern might be related to alcohol dose and exposure conditions, and warrants further investigation.


Assuntos
Etanol/toxicidade , Glândulas Mamárias Animais/efeitos dos fármacos , Neoplasias Mamárias Experimentais/patologia , Efeitos Tardios da Exposição Pré-Natal/patologia , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Transformação Celular Viral/genética , Receptor alfa de Estrogênio/metabolismo , Etanol/farmacologia , Feminino , Feto/efeitos dos fármacos , Glândulas Mamárias Animais/crescimento & desenvolvimento , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/patogenicidade , Camundongos , Camundongos Transgênicos , Gravidez , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Receptor ErbB-2/genética , Regulação para Cima
17.
Breast Cancer Res Treat ; 149(1): 99-108, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25519042

RESUMO

The urokinase receptor (uPAR) plays a critical role in breast cancer (BC) progression and metastases and is a validated target for novel therapies. The current study investigates the effects of MV-uPA, an oncolytic measles virus fully retargeted against uPAR in syngeneic and xenograft BC metastases models. In vitro replication and cytotoxicity of MVs retargeted against human (MV-h-uPA) or mouse (MV-m-uPA) uPAR were assessed in human and murine cancer and non-cancer mammary epithelial cells. The in vivo effects of species-specific uPAR retargeted MVs were assessed in syngeneic and xenograft models of experimental metastases, established by intravenous administration of luciferase expressing 4T1 or MDA-MD-231 cells. Metastases progression was assessed by in vivo bioluminescence imaging. Tumor targeting was evaluated by qRT-PCR of MV-N, rescue of viable viral particles, and immunostaining of MV particles in lungs from tumor bearing mice. In vitro, MV-h-uPA and MV-m-uPA selectively infected, replicated, and induced cytotoxicity in cancer compared to non-cancer cells in a species-specific manner. In vivo, MV-m-uPA delayed 4T1 lung metastases progression and prolonged survival. These effects were associated with identification of viable viral particles, viral RNA, and detection of MV-N by immunostaining from lung tissues in treated mice. In the human MDA-MB-231 metastases model, intravenous administration of MV-h-uPA markedly inhibited metastases progression and significantly improved survival, compared to controls. No significant treatment-related toxicity was observed in treated mice. The above preclinical findings strongly suggest that uPAR retargeted measles virotherapy is a novel and feasible systemic therapy strategy against metastatic breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias Mamárias Experimentais/genética , Vírus do Sarampo/genética , Terapia Viral Oncolítica , Receptores de Ativador de Plasminogênio Tipo Uroquinase/biossíntese , Administração Intravenosa , Animais , Neoplasias da Mama/terapia , Neoplasias da Mama/virologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Mamárias Experimentais/terapia , Neoplasias Mamárias Experimentais/virologia , Camundongos , Metástase Neoplásica , Vírus Oncolíticos/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
18.
J Vis Exp ; (93): e52232, 2014 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-25490047

RESUMO

Oncolytic viruses are a novel anticancer therapy with the ability to target tumor cells, while leaving healthy cells intact. For this strategy to be successful, recent studies have shown that involvement of the host immune system is essential. Therefore, oncolytic virotherapy should be evaluated within the context of an immunocompetent model. Furthermore, the study of antitumor therapies in tolerized animal models may better recapitulate results seen in clinical trials. Cotton rats, commonly used to study respiratory viruses, are an attractive model to study oncolytic virotherapy as syngeneic models of mammary carcinoma and osteosarcoma are well established. However, there is a lack of published information on the proper handling procedure for these highly excitable rodents. The handling and capture approach outlined minimizes animal stress to facilitate experimentation. This technique hinges upon the ability of the researcher to keep calm during handling and perform procedures in a timely fashion. Finally, we describe how to prepare cotton rat mammary tumor cells for consistent subcutaneous tumor formation, and how to perform intratumoral and intraperitoneal injections. These methods can be applied to a wide range of studies furthering the development of the cotton rat as a relevant pre-clinical model to study antitumor therapy.


Assuntos
Neoplasias Experimentais/terapia , Neoplasias Experimentais/virologia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/fisiologia , Animais , Neoplasias Ósseas/terapia , Neoplasias Ósseas/virologia , Modelos Animais de Doenças , Feminino , Neoplasias Mamárias Experimentais/terapia , Neoplasias Mamárias Experimentais/virologia , Osteossarcoma/terapia , Osteossarcoma/virologia , Ratos , Sigmodontinae , Estresse Fisiológico
19.
J Vis Exp ; (85)2014 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-24748051

RESUMO

Breast cancer is a heterogeneous disease involving complex cellular interactions between the developing tumor and immune system, eventually resulting in exponential tumor growth and metastasis to distal tissues and the collapse of anti-tumor immunity. Many useful animal models exist to study breast cancer, but none completely recapitulate the disease progression that occurs in humans. In order to gain a better understanding of the cellular interactions that result in the formation of latent metastasis and decreased survival, we have generated an inducible transgenic mouse model of YFP-expressing ductal carcinoma that develops after sexual maturity in immune-competent mice and is driven by consistent, endocrine-independent oncogene expression. Activation of YFP, ablation of p53, and expression of an oncogenic form of K-ras was achieved by the delivery of an adenovirus expressing Cre-recombinase into the mammary duct of sexually mature, virgin female mice. Tumors begin to appear 6 weeks after the initiation of oncogenic events. After tumors become apparent, they progress slowly for approximately two weeks before they begin to grow exponentially. After 7-8 weeks post-adenovirus injection, vasculature is observed connecting the tumor mass to distal lymph nodes, with eventual lymphovascular invasion of YFP+ tumor cells to the distal axillary lymph nodes. Infiltrating leukocyte populations are similar to those found in human breast carcinomas, including the presence of αß and γδ T cells, macrophages and MDSCs. This unique model will facilitate the study of cellular and immunological mechanisms involved in latent metastasis and dormancy in addition to being useful for designing novel immunotherapeutic interventions to treat invasive breast cancer.


Assuntos
Modelos Animais de Doenças , Neoplasias Mamárias Experimentais/patologia , Adenoviridae/genética , Alelos , Animais , Feminino , Genes p53 , Genes ras , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
20.
J Cancer Res Clin Oncol ; 140(1): 99-107, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24194353

RESUMO

PURPOSE: JF1/Ms mice, an inbred strain derived from Japanese wild mice, carry a germline hypomorphic mutation in the endothelin receptor type B gene (Ednrb). We observed that the JF1/Ms mice develop various spontaneous tumors at a high incidence late in life. The aim of this study was to elucidate the mechanism responsible for spontaneous tumors in these mice. Possible relevance of milk-borne mammary tumor virus and gene alterations in Ednrb to tumorigenesis was explored. METHODS: Expression and methylation status of Ednrb were quantitatively analyzed in normal and cancer tissues of mammary gland, liver, submandibular gland as well as in a cultured cell line, MW1, established from a submandibular gland adenocarcinoma. The biological effects of EDNRB were examined in the MW1 cells transfected with wild-type Ednrb. RESULTS: Transcripts of Ednrb were barely detectable, and the promoter region of Ednrb was hypermethylated in tumor tissues and the MW1 cells. In contrast, normal counterpart tissues showed positive expression of Ednrb transcripts and had unmethylated promoter regions. Treatment of the MW1 cells with 5-Aza-dC restored transcription of Ednrb to normal levels. Transfection of the MW1 cells with Ednrb1 (MW1-Ednrb1) resulted in lower growth rates and morphological changes compared with the mock-transfected MW1 cells (MW1-mock1). Furthermore, the MW1-Ednrb1 cells transplanted in syngeneic mice showed a lower proliferation rate than the MW1-mock1 cells. CONCLUSIONS: Germline mutation and subsequent promoter methylation of Ednrb may be relevant to cancer susceptibility in the JF1/Ms mice. These data indicate that Ednrb acts as a tumor suppressor, as reported in human prostate, bladder, and clear cell renal carcinomas.


Assuntos
Metilação de DNA , Mutação em Linhagem Germinativa , Camundongos Endogâmicos/genética , Neoplasias Experimentais/genética , Receptor de Endotelina B/genética , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Feminino , Genes Supressores de Tumor , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/patologia , Neoplasias Mamárias Experimentais/virologia , Vírus do Tumor Mamário do Camundongo/isolamento & purificação , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/patologia , Neoplasias Experimentais/virologia , Regiões Promotoras Genéticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA