Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 147
Filter
1.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768912

ABSTRACT

Metastasis reflects both the inherent properties of tumor cells and the response of the stroma to the presence of the tumor. Vascular barrier properties, either due to endothelial cell (EC) or pericyte function, play an important role in metastasis in addition to the contribution of the immune system. The Shb gene encodes the Src homology-2 domain protein B that operates downstream of tyrosine kinases in both vascular and immune cells. We have investigated E0771.lmb breast carcinoma metastasis in mice with conditional deletion of the Shb gene using the Cdh5-CreERt2 transgene, resulting in inactivation of the Shb-gene in EC and some hematopoietic cell populations. Lung metastasis from orthotopic tumors, tumor vascular and immune cell characteristics, and immune cell gene expression profiles were determined. We found no increase in vascular leakage that could explain the observed increase in metastasis upon the loss of Shb expression. Instead, Shb deficiency in EC promoted the recruitment of monocytic/macrophagic myeloid-derived suppressor cells (mMDSC), an immune cell type that confers a suppressive immune response, thus enhancing lung metastasis. An MDSC-promoting cytokine/chemokine profile was simultaneously observed in tumors grown in mice with EC-specific Shb deficiency, providing an explanation for the expanded mMDSC population. The results demonstrate an intricate interplay between tumor EC and immune cells that pivots between pro-tumoral and anti-tumoral properties, depending on relevant genetic and/or environmental factors operating in the microenvironment.


Subject(s)
Endothelial Cells/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , Myeloid-Derived Suppressor Cells/pathology , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins/physiology , Tumor Microenvironment , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Endothelial Cells/metabolism , Female , Lung Neoplasms/etiology , Lung Neoplasms/metabolism , Macrophages/metabolism , Macrophages/pathology , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myeloid-Derived Suppressor Cells/metabolism , Neovascularization, Pathologic/metabolism
2.
Pesqui. vet. bras ; 40(6): 466-473, June 2020. tab, graf, ilus
Article in English | VETINDEX, LILACS | ID: biblio-1135647

ABSTRACT

Mammary tumors in female dogs are the most frequent and corresponds to half of the canine tumors. The objectives of this study were to determine the risk factors associated with the occurrence of mammary tumors in female dogs and to evaluate the macroscopic characteristics of these neoformations, using 386 dogs from the "Outubro Rosa Pets" events done within the cities of Uberlândia and Patos de Minas, Minas Gerais State, Brazil, in 2015 (n=194), 2016 (n=105) and 2017 (n=87). For the determination of risk factors, the binary logistic regression test (P<0.05) was performed. The occurrence of mammary tumors was 23.6% (91/386). The significant risk factors identified were increased age (P<0.001), overweight (P=0.048) and non-castration (P<0.001) with a chance of, respectively, 1.6, 2.3 and 9.3 times for the development of mammary tumors. In dogs with mammary tumors (n=91), 153 lesions were present, of which 39 female dogs had two or more lesions (42.8%). Most of the lesions were at the caudal abdominal (M4) and inguinal (M5) mammary glands (60.13%, 92/153). Relative to the size of the lesions, it was observed that in 78% of the female dogs the lesions were determined asT1 (<3cm), 16.5% were T2 (3-5cm) and 5.5% T3 (>5cm). At least 15.4% (14/91) of the dogs had one of the regional lymph nodes increased. In conclusion, the occurrence of mammary tumors in the evaluated population was 23.6% and that age, overweight and non-realization of ovariohysterectomy are risk factors associated with the development of mammary tumors.(AU)


Em cadelas os tumores mamários são os mais frequentes e correspondem a aproximadamente metade dos tumores em cães. Este estudo teve os objetivos de determinar os fatores de risco envolvidos na ocorrência de tumores mamários em cadelas e avaliar as características macroscópicas destas neoformações, utilizando 386 cadelas do evento "Outubro Rosa Pets" nos municípios de Uberlândia e Patos de Minas, Minas Gerais, Brasil, em 2015 (n=194), 2016 (n=105) e 2017 (n=87). Para a determinação dos fatores de risco utilizou-se o teste de Regressão logística binária (P<0,05). A ocorrência de tumores mamários foi de 23,6% (91/386). Os fatores de risco significativos identificados foram aumento da idade (P<0,001), sobrepeso (P=0,048) e não-castração (P<0,001) com a chance de, respectivamente, 1,6, 2,3 e 9,3 vezes de desenvolvimento de tumores mamários. Nas cadelas com tumores mamários (n=91), constatou-se a presença de 153 lesões, sendo que 39 cadelas apresentaram duas ou mais lesões (42,8%). A maioria das lesões localizaram-se nas mamas abdominais caudais (M4) e inguinais (M5) (60,13%; 92/153). Em relação ao tamanho das lesões, observou-se que 78% das cadelas eram T1 (<3cm), 16,5% T2 (3-5cm) e 5,5% T3 (>5cm). Pelo menos 15,4% (14/91) das cadelas apresentaram um dos linfonodos regionais aumentados. Conclui-se que a ocorrência dos tumores mamários na população avaliada foi de 23,6% e que a idade, sobrepeso e não ovariohisterectomia são fatores de risco para o desenvolvimento de tumores mamários.(AU)


Subject(s)
Animals , Female , Dogs , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/epidemiology , Dog Diseases , Mammary Neoplasms, Animal/prevention & control , Risk Factors
3.
Sci Rep ; 9(1): 13485, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31530877

ABSTRACT

We report detection of Baculoviral inhibitor of apoptosis repeat containing-5 (BIRC5) protein biomarker in dog serum by label-free surface plasmon resonance (SPR) immunosensor. Initially, overexpression of BIRC5 in canine mammary tumour (CMT) tissues was confirmed by real-time PCR. Recombinant BIRC5 was produced and protein specific antibodies developed in guinea pig specifically reacted with native protein in immunohistochemistry and immunocytochemistry. SPR immunosensor was developed by fabricating anti-BIRC5 antibodies on gold sensor disc. The equilibrium dissociation constant, (KD = kd/ka) was 12.1 × 10-12 M; which indicates that antibodies are of high affinity with sensitivity in picomolar range. The SPR assay could detect as low as 6.25 pg/ml of BIRC5 protein in a calibration experiment (r2 = 0.9964). On testing real clinical samples, 95% specificity and 73.33% sensitivity were recorded. The average amount of serum BIRC5 in dogs with CMT was 110.02 ± 9.77 pg/ml; whereas, in non-cancerous disease conditions, 44.79 ± 4.28 pg/ml and in healthy dog sera 30.28 ± 2.99 pg/ml protein was detected. The SPR immunosensor for detection of BIRC5 in dog sera is reported for the first time and this may find prognostic and diagnostic applications in management of CMT. In future, 'on-site' sensors can be developed using this technique for near-patient testing.


Subject(s)
Biomarkers, Tumor , Dog Diseases/diagnosis , Dog Diseases/metabolism , Mammary Neoplasms, Animal/diagnosis , Mammary Neoplasms, Animal/metabolism , Surface Plasmon Resonance , Survivin/metabolism , Animals , Biosensing Techniques , Dog Diseases/etiology , Dogs , Immunoassay , Immunohistochemistry , Limit of Detection , Mammary Neoplasms, Animal/etiology , Polymerase Chain Reaction , ROC Curve , Recombinant Proteins , Surface Plasmon Resonance/methods
4.
Cancer Prev Res (Phila) ; 12(4): 201-210, 2019 04.
Article in English | MEDLINE | ID: mdl-30885926

ABSTRACT

Deep-frying is a popular form of food preparation used globally and throughout in the United States. Each time dietary oils are heated to deep-frying temperatures, they undergo chemical alterations that result in a new matrix of lipid structures. These lipid products include triglyceride dimers, polymers, oxidized triglycerides, and cyclic monomers, which raises nutritional concerns about associations between these lipid products and heightened health risks. Reports of associations between thermally abused frying oil and deleterious health outcomes currently exist, yet there is little information concerning the effects of thermally abused frying oil consumption and the progression of breast cancer. This study used a late-stage breast cancer murine model and in vivo bioluminescent imaging to monitor progression of metastasis of 4T1 tumor cells in animals consuming fresh soybean oil (SBO) and a thermally abused frying oil (TAFO). Bioluminescent and histologic examinations demonstrated that TAFO consumption resulted in a marked increase of metastatic lung tumor formation compared to SBO consumption. Further, in animals consuming the TAFO treatment diet, metastatic tumors in the lung displayed a 1.4-fold increase in the Ki-67 marker of cellular proliferation and RNA-sequencing analysis of the hepatic tissue revealed a dietary-induced modulation of gene expression in the liver.


Subject(s)
Cell Proliferation , Cooking/methods , Dietary Fats, Unsaturated/toxicity , Disease Models, Animal , Hot Temperature/adverse effects , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/pathology , Animals , Apoptosis , Female , Lung Neoplasms/etiology , Mammary Neoplasms, Animal/etiology , Mice , Mice, Inbred BALB C , Tumor Cells, Cultured
5.
Nutr Res ; 61: 41-52, 2019 01.
Article in English | MEDLINE | ID: mdl-30683438

ABSTRACT

Previous studies have shown that early life intake of high-fat diet or western-style diet (WD) enhances the development of mammary tumors in adult female rats. Thus, we hypothesized that maternal WD throughout pregnancy and the lactation period could speed up the development of MNU-induced mammary tumors and alter their gene expression. For this, the present study investigated the gene expression profile of chemically-induced mammary tumors in female rat offspring from dams fed a WD or a control diet. Pregnant female Sprague-Dawley rats received a WD (high-fat, low-fiber and oligoelements) or a control diet from gestational day 12 until post-natal day (PND) 21. At PND 21, female offspring received a single dose of N-Methyl-N-Nitrosourea (MNU, 50 mg/kg body weight) and were fed a control diet for 13 weeks. Tumor incidence, multiplicity, and latency were recorded and mammary gland samples were collected for histopathology and gene expression analysis. Tumor multiplicity and histological grade were significantly higher and tumor latency was lower in WD offspring compared to control offspring. Transcriptome profiling identified 57 differentially expressed genes in tumors from WD offspring as compared to control offspring. There was also an increase in mRNA expression of genes such as Emp3, Ccl7, Ets1, Abcc5, and Cyr61, indicative of more aggressive disease detected in tumors from WD offspring. Thus, maternal WD diet increased MNU-induced mammary carcinogenesis in adult female offspring through transcriptome changes that resulted in a more aggressive disease.


Subject(s)
Diet, High-Fat , Diet, Western , Mammary Neoplasms, Animal/etiology , Maternal Nutritional Physiological Phenomena , Pregnancy Complications , Prenatal Exposure Delayed Effects/genetics , Transcriptome , Animals , Female , Gene Expression Profiling , Genes, Neoplasm , Lactation , Mammary Neoplasms, Animal/chemically induced , Mammary Neoplasms, Animal/pathology , Methylnitrosourea , Mothers , Neoplasm Grading , Pregnancy , RNA, Messenger/metabolism , Rats, Sprague-Dawley
6.
Sci Rep ; 9(1): 20271, 2019 12 30.
Article in English | MEDLINE | ID: mdl-31889127

ABSTRACT

Maternal or paternal high fat (HF) diet can modify the epigenome in germ cells and fetal somatic cells leading to an increased susceptibility among female offspring of multiple generations to develop breast cancer. We determined if combined treatment with broad spectrum DNA methyltransferase (DNMT) inhibitor hydralazine and histone deacetylase (HDAC) inhibitor valproic acid (VPA) will reverse this increased risk. C57BL/6 mouse dams were fed either a corn oil-based HF or control diet during pregnancy. Starting at age 7 weeks, female offspring were administered 3 doses of 7,12-dimethylbenz[a]anthracene (DMBA) to initiate mammary cancer. After last dose, offspring started receiving VPA/hydralazine administered via drinking water: no adverse health effects were detected. VPA/hydralazine reduced mammary tumor multiplicity and lengthened tumor latency in HF offspring when compared with non-treated HF offspring. The drug combination inhibited DNMT3a protein levels and increased expression of the tumor suppressor gene Cdkn2a/p16 in mammary tumors of HF offspring. In control mice not exposed to HF diet in utero, VPA/hydralazine increased mammary tumor incidence and burden, and elevated expression of the unfolded protein response and autophagy genes, including HIF-1α, NFkB, PERK, and SQSTM1/p62. Expression of these genes was already upregulated in HF offspring prior to VPA/hydralazine treatment. These findings suggest that breast cancer prevention strategies with HDAC/DNMT inhibitors need to be individually tailored.


Subject(s)
Cell Transformation, Neoplastic , Diet, High-Fat , Hydralazine/metabolism , Mammary Neoplasms, Animal/etiology , Maternal Exposure , Prenatal Exposure Delayed Effects , Valproic Acid/metabolism , Animals , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , CpG Islands , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation , Diet, High-Fat/adverse effects , Disease Susceptibility , Female , Hydralazine/administration & dosage , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental , Mice , Pregnancy , Tumor Burden , Valproic Acid/administration & dosage
7.
Mol Nutr Food Res ; 63(6): e1801150, 2019 03.
Article in English | MEDLINE | ID: mdl-30569599

ABSTRACT

SCOPE: The present study investigates the precise mechanism by which maternal n-3 PUFAs decrease mammary cancer risk of offspring in terms of epigenetics. METHODS AND RESULTS: Transgenic fat-1 and wild-type C57BL/6J littermates are fed an n-6 PUFAs diet during pregnancy. Wild-genotype offspring of fat-1 mothers (fat-1 group) are compared with wild-genotype offspring of C57BL/6J mothers (control group) in breast cancer risk. Fat-1 group shows a significantly lower tumor incidence and smaller tumor volume compared with control group. n-3 PUFAs in fat-1 mothers change the expression of long noncoding RNA (lncRNA, 53 upregulated and 45 downregulated) in mammary glands of offspring. The lncRNA changes are associated with the changes of mRNA in multiple oncogenic signaling pathways, especially NF-κB, Jak-STAT, and MAPK pathways. Expression of key protein in these pathways, namely p65, p60, STAT3, Jak1, and p38, are significantly inhibited in fat-1 group. In line with these results, reduced proliferation and increased apoptosis are also observed in mammary epithelial of fat-1 group than control group. CONCLUSION: The anticancer effect of maternal n-3 PUFAs is related to the regulation of lncRNA expression, which can further regulate the susceptibility of offspring to breast cancer.


Subject(s)
Fatty Acids, Omega-3/metabolism , Mammary Glands, Animal/cytology , Mammary Neoplasms, Animal/etiology , RNA, Long Noncoding/genetics , Animals , Apoptosis , Caenorhabditis elegans Proteins/genetics , Cell Proliferation , Estradiol/blood , Fatty Acid Desaturases/genetics , Fatty Acids/analysis , Female , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/genetics , Mice, Transgenic , Pregnancy , Risk Factors
8.
BMC Cancer ; 18(1): 741, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30012106

ABSTRACT

BACKGROUND: The receptor CXCR4 and its ligand CXCL12 play crucial roles in breast cancer. Despite the fact that the spontaneous feline mammary carcinoma (FMC) is considered a suitable model for breast cancer studies, the importance of the CXCR4/CXCL12 axis in FMC is completely unknown. Therefore, this work aims to elucidate the role of CXCR4 and its ligand in the progression of FMC and metastatic disease. METHODS: CXCR4 and CXCL12 expression was analyzed by immunohistochemistry and immunofluorescence on primary tumors (PT), regional and distant metastases of female cats with mammary carcinoma and correlated with serum CXCL12 levels, tumor molecular subtypes and clinicopathological features. RESULTS: CXCR4 was more expressed in PT than in metastases (p = 0.0067), whereas CXCL12 was highly expressed in metastatic lesions located in liver and lung (p < 0.0001), as reported for human breast cancer. Moreover, cats with CXCR4 positive PT exhibited significantly lower serum CXCL12 levels than cats with CXCR4 negative mammary carcinomas (p = 0.0324). At metastatic lesions, HER2-overexpressing tumors presented higher CXCR4 expression than the other molecular tumor subtypes (p = 0.012) and significant differences in overall (p = 0.0147) and disease-free survival (p = 0.0279) curves between the cats with CXCL12 positive and CXCL12 negative tumors were found. Indeed, CXCL12 negative PT were associated with unfavorable prognosis in cats with HER2-overexpressing tumors. CONCLUSIONS: This work exposes part of the complex interaction between CXCR4 and CXCL12 in PT, but also in metastases of a breast cancer model. These findings could uncover novel therapeutic tools to be used in cats and humans.


Subject(s)
Cat Diseases/etiology , Chemokine CXCL12/physiology , Mammary Neoplasms, Animal/etiology , Receptor, ErbB-2/analysis , Receptors, CXCR4/physiology , Animals , Cat Diseases/immunology , Cat Diseases/pathology , Cats , Chemokine CXCL12/analysis , Female , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Neoplasm Metastasis , Prognosis , Receptors, CXCR4/analysis
9.
Cytometry A ; 93(1): 125-136, 2018 01.
Article in English | MEDLINE | ID: mdl-28834173

ABSTRACT

Postnatal mammary gland development requires the presence of mammary stem and progenitor cells (MaSC), which give rise to functional milk-secreting cells and regenerate the mammary epithelium with each cycle of lactation. These long-lived, tissue-resident MaSC are also targets for malignant transformation and may be cancer cells-of-origin. Consequently, MaSC are extensively researched in relation to their role and function in development, tissue regeneration, lactation, and breast cancer. The basic structure and function of the mammary gland are conserved among all mammalian species, from the most primitive to the most evolved. However, species vary greatly in their lactation strategies and mammary cancer incidence, making MaSC an interesting focus for comparative research. MaSC have been characterized in mice, to a lesser degree in humans, and to an even lesser degree in few additional mammals. They remain uncharacterized in most mammalian species, including "ancient" monotremes, marsupials, wild, and rare species, as well as in common and domestic species such as cats. Identification and comparison of MaSC across a large variety of species, particularly those with extreme lactational adaptations or low mammary cancer incidence, is expected to deepen our understanding of development and malignancy in the mammary gland. Here, we review the current status of MaSC characterization across species, and underline species variations in lactation and mammary cancer through which we may learn about the role of MaSC in these processes. © 2017 International Society for Advancement of Cytometry.


Subject(s)
Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Stem Cells/cytology , Animals , Animals, Domestic , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Female , Humans , Lactation/physiology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/physiology , Mammary Glands, Human/pathology , Mammary Glands, Human/physiology , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/pathology , Mice , Neoplastic Stem Cells/pathology , Primates , Rodentia , Ruminants , Species Specificity , Stem Cells/pathology , Stem Cells/physiology
10.
Cancer Prev Res (Phila) ; 10(10): 553-562, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28904060

ABSTRACT

Obesity and alterations in metabolic programming from early diet exposures can affect the propensity to disease in later life. Through dietary manipulation, developing mouse pups were exposed to a hyperinsulinemic, hyperglycemic milieu during three developmental phases: gestation, lactation, and postweaning. Analyses showed that a postweaning high fat/high sugar (HF/HS) diet had the main negative effect on adult body weight, glucose tolerance, and insulin resistance. However, dimethylbenz[a]anthracene (DMBA)-induced carcinogenesis revealed that animals born to a mother fed a HF/HS gestation diet, nursed by a mother on a mildly diet-restricted, low fat/low sugar diet (DR) and weaned onto a HF/HS diet (HF/DR/HF) had the highest mammary tumor incidence, while HF/HF/DR had the lowest tumor incidence. Cox proportional hazards analysis showed that a HF/HS postweaning diet doubled mammary cancer risk, and a HF/HS diet during gestation and postweaning increased risk 5.5 times. Exposure to a HF/HS diet during gestation, when combined with a postweaning DR diet, had a protective effect, reducing mammary tumor risk by 86% (HR = 0.142). Serum adipocytokine analysis revealed significant diet-dependent differences in leptin/adiponectin ratio and IGF-1. Flow cytometry analysis of cells isolated from mammary glands from a high tumor incidence group, DR/HF/HF, showed a significant increase in the size of the mammary stem cell compartment compared with a low tumor group, HF/HF/DR. These results indicate that dietary reprogramming induces an expansion of the mammary stem cell compartment during mammary development, increasing likely carcinogen targets and mammary cancer risk. Cancer Prev Res; 10(10); 553-62. ©2017 AACRSee related editorial by Freedland, p. 551-2.


Subject(s)
Carcinogenesis/metabolism , Diet, High-Fat/adverse effects , Dietary Sugars/adverse effects , Mammary Glands, Animal/embryology , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Experimental/metabolism , Stem Cells/pathology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Adiponectin/blood , Animals , Body Weight/physiology , Diet, Fat-Restricted , Feeding Behavior , Female , Insulin Resistance/physiology , Insulin-Like Growth Factor I/analysis , Lactation/metabolism , Leptin/blood , Mammary Glands, Animal/cytology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/blood , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/prevention & control , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/prevention & control , Maternal Exposure/adverse effects , Mice , Mice, Inbred SENCAR , Obesity/etiology , Obesity/metabolism , Risk Factors , Time Factors
11.
Sci Rep ; 7(1): 11306, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28900173

ABSTRACT

The fibroblast growth factor receptor (FGFR) family of receptor tyrosine kinases (RTKs) regulates signaling pathways involved in cell proliferation and differentiation. Currently, the anti-tumor properties of FGFR inhibitors are being tested in preclinical and clinical studies. Nevertheless, reports on FGFR inhibitor-mediated breast cancer prevention are sparse. In this study, we investigated the anti-cancer benefits of AZD4547, an FGFR1-3 inhibitor, in ErbB2-overexpressing breast cancer models. AZD4547 (1-5 µM) demonstrated potent anti-proliferative effects, inhibition of stemness, and suppression of FGFR/RTK signaling in ErbB2-overexpressing human breast cancer cells. To study the in vivo effects of AZD4547 on mammary development, mammary epithelial cell (MEC) populations, and oncogenic signaling, MMTV-ErbB2 transgenic mice were administered AZD4547 (2-6 mg/kg/day) for 10 weeks during the 'risk window' for mammary tumor development. AZD4547 significantly inhibited ductal branching and MEC proliferation in vivo, which corroborated the in vitro anti-proliferative properties. AZD4547 also depleted CD24/CD49f-sorted MEC populations, as well as the CD61highCD49fhigh tumor-initiating cell-enriched population. Importantly, AZD4547 impaired stem cell-like characteristics in primary MECs and spontaneous tumor cells. Moreover, AZD4547 downregulated RTK, mTOR, and Wnt/ß-catenin signaling pathways in premalignant mammary tissues. Collectively, our data provide critical preclinical evidence for AZD4547 as a potential breast cancer preventative and therapeutic agent.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/pathology , Piperazines/pharmacology , Precancerous Conditions , Pyrazoles/pharmacology , Receptor, ErbB-2/genetics , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Self Renewal/genetics , Cell Survival/drug effects , Female , Gene Expression , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Experimental , Mice , Mice, Transgenic , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Signal Transduction , Stem Cells/metabolism
12.
Cancer Res ; 77(9): 2500-2511, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28373182

ABSTRACT

The association between obesity and breast cancer risk and prognosis is well established in estrogen receptor (ER)-positive disease but less clear in HER2-positive disease. Here, we report preclinical evidence suggesting weight maintenance through calorie restriction (CR) may limit risk of HER2-positive breast cancer. In female MMTV-HER2/neu transgenic mice, we found that ERα and ERß expression, mammary tumorigenesis, and survival are energy balance dependent in association with epigenetic reprogramming. Mice were randomized to receive a CR, overweight-inducing, or diet-induced obesity regimen (n = 27/group). Subsets of mice (n = 4/group/time point) were euthanized after 1, 3, and 5 months to characterize diet-dependent metabolic, transcriptional, and epigenetic perturbations. Remaining mice were followed up to 22 months. Relative to the overweight and diet-induced obesity regimens, CR decreased body weight, adiposity, and serum metabolic hormones as expected and also elicited an increase in mammary ERα and ERß expression. Increased DNA methylation accompanied this pattern, particularly at CpG dinucleotides located within binding or flanking regions for the transcriptional regulator CCCTC-binding factor of ESR1 and ESR2, consistent with sustained transcriptional activation of ERα and ERß. Mammary expression of the DNA methylation enzyme DNMT1 was stable in CR mice but increased over time in overweight and diet-induced obesity mice, suggesting CR obviates epigenetic alterations concurrent with chronic excess energy intake. In the survival study, CR elicited a significant suppression in spontaneous mammary tumorigenesis. Overall, our findings suggest a mechanistic rationale to prevent or reverse excess body weight as a strategy to reduce HER2-positive breast cancer risk. Cancer Res; 77(9); 2500-11. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/genetics , Estrogen Receptor beta/genetics , Mammary Neoplasms, Animal/genetics , Obesity/genetics , Animals , Breast Neoplasms/physiopathology , Caloric Restriction , Carcinogenesis/genetics , DNA Methylation/genetics , Energy Metabolism/genetics , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Animal/etiology , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Obesity/complications , Obesity/physiopathology , Receptor, ErbB-2/genetics , Risk Factors
13.
J Vet Sci ; 18(1): 101-104, 2017 Mar 30.
Article in English | MEDLINE | ID: mdl-27456773

ABSTRACT

The expression of stage-specific embryonic antigens (SSEAs) was determined in several types of canine cancer cells. Flow cytometry showed SSEA-1 expression in glioblastoma, melanoma, and mammary cancer cells, although none expressed SSEA-3 or SSEA-4. Expression of SSEA-1 was not detected in lymphoma, osteosarcoma, or hemangiosarcoma cell lines. Relatively stable SSEA-1 expression was observed between 24 and 72 h of culture. After 8 days in culture, sorted SSEA-1- and SSEA-1+ cells re-established SSEA-1 expression to levels comparable to those observed in unsorted cells. Our results document, for the first time, the expression of SSEA-1 in several canine cancer cell lines.


Subject(s)
Glioblastoma/veterinary , Mammary Neoplasms, Animal/metabolism , Melanoma/veterinary , Stage-Specific Embryonic Antigens/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Dog Diseases , Dogs , Female , Flow Cytometry/veterinary , Glioblastoma/etiology , Glioblastoma/metabolism , Mammary Neoplasms, Animal/etiology , Melanoma/etiology , Melanoma/metabolism
14.
Proc Natl Acad Sci U S A ; 113(33): E4820-7, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27496330

ABSTRACT

Branching morphogenesis in the mammary gland is achieved by the migration of epithelial cells through a microenvironment consisting of stromal cells and extracellular matrix (ECM). Here we show that galectin-1 (Gal-1), an endogenous lectin that recognizes glycans bearing N-acetyllactosamine (LacNAc) epitopes, induces branching migration of mammary epithelia in vivo, ex vivo, and in 3D organotypic cultures. Surprisingly, Gal-1's effects on mammary patterning were independent of its glycan-binding ability and instead required localization within the nuclei of mammary epithelia. Nuclear translocation of Gal-1, in turn, was regulated by discrete cell-surface glycans restricted to the front of the mammary end buds. Specifically, α2,6-sialylation of terminal LacNAc residues in the end buds masked Gal-1 ligands, thereby liberating the protein for nuclear translocation. Within mammary epithelia, Gal-1 localized within nuclear Gemini bodies and drove epithelial invasiveness. Conversely, unsialylated LacNAc glycans, enriched in the epithelial ducts, sequestered Gal-1 in the extracellular environment, ultimately attenuating invasive potential. We also found that malignant breast cells possess higher levels of nuclear Gal-1 and α2,6-SA and lower levels of LacNAc than nonmalignant cells in culture and in vivo and that nuclear localization of Gal-1 promotes a transformed phenotype. Our findings suggest that differential glycosylation at the level of tissue microanatomy regulates the nuclear function of Gal-1 in the context of mammary gland morphogenesis and in cancer progression.


Subject(s)
Cell Nucleus/metabolism , Galectin 1/physiology , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Animal/etiology , Morphogenesis , Polysaccharides/physiology , Animals , Female , Glycosylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
15.
Vet Radiol Ultrasound ; 57(5): 475-81, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27412486

ABSTRACT

This randomized controlled trial study aimed to identify the optimal positive pressure (PP) level that can clear atelectasis while avoiding pulmonary hyperinflation during the breath-hold technique in dogs undergoing thoracic computed tomography (CT). Sixty dogs affected by mammary tumors undergoing thoracic CT for the screening of pulmonary metastases were randomly assigned to six groups with different levels of PP during the breath-hold technique: 0 (control), 5 (PP5), 8 (PP8), 10 (PP10), 12 (PP12), and 15 (PP15) cmH2 O. The percentage of atelectatic lung region was lower in the PP10 (3.7 ± 1.1%; P = 0.002), PP12 (3.4 ± 1.3%; P = 0.0001), and PP15 (2.8 ± 0.9%; P = 0.006) groups than in the control group (5.0 ± 2.3%), and the percentage of poorly aerated lung region was lower in the PP8 (15.1 ± 2.6%; P = 0.0009), PP10 (13.0 ± 2.0 %; P = 0.002), PP12 (13.0 ± 2.2 %; P = 0.0002), and PP15 (11.1 ± 1.9%; P = 0.0002) groups than in the control group (19.8 ± 5.0). The percentage of normally aerated lung region, however, was higher in the PP10 (79.7 ± 4.1%; P = 0.005), PP12 (79.8 ± 5.1%; P = 0.0002), and PP15 (80.2 ± 4.9%; P = 0.002) groups than in the control group (73.4 ± 6.6%). A PP of 10-12 cmH2 O during the breath-hold technique should be considered to improve lung aeration during a breath-hold technique in dogs undergoing thoracic CT.


Subject(s)
Dog Diseases/diagnostic imaging , Mammary Neoplasms, Animal/diagnostic imaging , Pulmonary Atelectasis/veterinary , Tomography, X-Ray Computed/veterinary , Animals , Dogs , Female , Mammary Neoplasms, Animal/etiology , Pulmonary Atelectasis/diagnostic imaging , Pulmonary Atelectasis/prevention & control , Tomography, X-Ray Computed/methods
16.
Sci Rep ; 6: 28602, 2016 06 24.
Article in English | MEDLINE | ID: mdl-27339599

ABSTRACT

While many studies have shown that maternal weight and nutrition in pregnancy affects offspring's breast cancer risk, no studies have investigated the impact of paternal body weight on daughters' risk of this disease. Here, we show that diet-induced paternal overweight around the time of conception can epigenetically reprogram father's germ-line and modulate their daughters' birth weight and likelihood of developing breast cancer, using a mouse model. Increased body weight was associated with changes in the miRNA expression profile in paternal sperm. Daughters of overweight fathers had higher rates of carcinogen-induced mammary tumors which were associated with delayed mammary gland development and alterations in mammary miRNA expression. The hypoxia signaling pathway, targeted by miRNAs down-regulated in daughters of overweight fathers, was activated in their mammary tissues and tumors. This study provides evidence that paternal peri-conceptional body weight may affect daughters' mammary development and breast cancer risk and warrants further studies in other animal models and humans.


Subject(s)
Breast Neoplasms/etiology , Mammary Neoplasms, Animal/etiology , Overweight/complications , Animals , Birth Weight/genetics , Body Mass Index , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Diet/methods , Disease Models, Animal , Down-Regulation/genetics , Fathers , Female , Male , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Nuclear Family , Overweight/pathology , Parent-Child Relations , Pregnancy , Risk
17.
Int J Cancer ; 138(3): 642-51, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26296387

ABSTRACT

The tree shrew is becoming an attractive experimental animal model for human breast cancer owing to a closer relationship to primates/humans than rodents. Tree shrews are superior to classical primates because tree shrew are easier to manipulate, maintain and propagate. It is required to establish a high-efficiency tree shrew breast cancer model for etiological research and drug assessment. Our previous studies suggest that 7,12-dimethylbenz(a)anthracene (DMBA) and medroxyprogesterone acetate (MPA) induce breast tumors in tree shrews with a low frequency (<50%) and long latency (∼ 7-month), making these methods less than ideal. We induced mammary tumors in tree shrew (Tupaia belangeri chinensis) by injection of lentivirus expressing the PyMT oncogene into mammary ducts of 22 animals. Most tree shrews developed mammary tumors with a latency of about three weeks, and by 7 weeks all injected tree shrews had developed mammary tumors. Among these, papillary carcinoma is the predominant tumor type. One case showed lymph node and lung metastasis. Interestingly, the expression levels of phosphorylated AKT, ERK and STAT3 were elevated in 41-68% of PyMT-induced mammary tumors, but not all tumors. Finally, we observed that the growth of PyMT-induced tree shrew mammary tumors was significantly inhibited by Cisplatin and Epidoxorubicin. PyMT-induced tree shrew mammary tumor model may be suitable for further breast cancer research and drug development, due to its high efficiency and short latency.


Subject(s)
Antigens, Viral, Tumor/genetics , Disease Models, Animal , Mammary Neoplasms, Animal/etiology , Polyomavirus/immunology , Tupaiidae , Animals , Carcinoma, Papillary/etiology , Epithelial Cells/pathology , Estrogen Receptor alpha/analysis , Female , Lentivirus/genetics , Mammary Neoplasms, Animal/chemistry , Mammary Neoplasms, Animal/drug therapy , STAT3 Transcription Factor/metabolism
18.
In Vitro Cell Dev Biol Anim ; 51(8): 776-86, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25898824

ABSTRACT

Overexpression of the oncoprotein erbB2/HER2 is present in 20-30% of breast cancer patients and inversely correlates with patient survival. Reports have demonstrated the deterministic power of the mammary microenvironment where the normal mammary microenvironment redirects cells of non-mammary origin or tumor-derived cells to adopt a mammary phenotype in an in vivo model. This phenomenon is termed tumor cell redirection. Tumor-derived cells that overexpress the erbB2 oncoprotein lose their tumor-forming capacity in this model. In this model, phosphorylation of erbB2 is attenuated thus reducing the tumor cell's tumor-forming potential. In this report, we describe our results using an in vitro model based on the in vivo model mentioned previously. Tumor-derived cells are mixed in predetermined ratios with normal mammary epithelial cells prior to seeding in vitro. In this in vitro model, the tumor-derived cells are redirected as determined by attenuated phosphorylation of the receptor and reduced sphere and colony formation. These results match those observed in the in vivo model. This in vitro model will allow expanded experimental options in the future to determine additional aspects of tumor cell redirection that can be translated to other types of cancer.


Subject(s)
Carcinogenesis/genetics , Mammary Neoplasms, Animal/etiology , Receptor, ErbB-2/physiology , Animals , Blotting, Western , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Mice , Mice, Transgenic , Models, Biological , Receptors, Virus/physiology , Signal Transduction/physiology
19.
Res Vet Sci ; 100: 232-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25843897

ABSTRACT

Local anesthetics have been reported to induce apoptosis in various cell lines. In this study, we showed that bupivacaine also induced apoptosis in DTK-SME cells, a vimentin(+)/AE1(+)/CK7(+)/HSP27(+), tumorigenic, immortalized, canine mammary tumor cell line. Bupivacaine induced apoptosis in DTK-SME cells in a time- and concentration-dependent manner. Apoptosis-associated morphological changes, including cell shrinkage and rounding, chromatin condensation, and formation of apoptotic bodies, were observed in the bupivacaine-treated DTK-SME cells. Apoptosis was further confirmed with annexin V staining, TUNEL staining, and DNA laddering assays. At the molecular level, the activation of caspases-3, -8, and -9 corresponded well to the degree of DNA fragmentation triggered by bupivacaine. We also demonstrated that the pan-caspase inhibitor, z-VAD-fmk, only partially inhibited the apoptosis induced by bupivacaine. Moreover, treated cells increased expression of endonuclease G, a death effector that acts independently of caspases. Our data suggested that bupivacaine-induced apoptosis occurs through both caspase-dependent and caspase-independent apoptotic pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bupivacaine/pharmacology , Caspases/metabolism , Dog Diseases/drug therapy , Mammary Neoplasms, Animal/drug therapy , Amino Acid Chloromethyl Ketones/metabolism , Animals , Caspase Inhibitors/metabolism , Cell Line, Tumor , DNA Fragmentation/drug effects , Dog Diseases/etiology , Dogs , Female , Mammary Neoplasms, Animal/etiology
20.
Breast Cancer Res ; 17: 14, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25848982

ABSTRACT

INTRODUCTION: Insulin analogues are structurally modified molecules with altered pharmaco-kinetic and -dynamic properties compared to regular human insulin used by diabetic patients. While these compounds are tested for undesired mitogenic effects, an epidemiological discussion is ongoing regarding an association between insulin analogue therapy and increased cancer incidence, including breast cancer. Standard in vivo rodent carcinogenesis assays do not pick up this possible increased carcinogenic potential. METHODS: Here we studied the role of insulin analogues in breast cancer development. For this we used the human relevant mammary gland specific p53R270H/⁺WAPCre mouse model. Animals received life long repeated treatment with four different insulin (-like) molecules: normal insulin, insulin glargine, insulin X10 (AspB10) or insulin-like growth factor 1 (IGF1). RESULTS: Insulin-like molecules with strong mitogenic signaling, insulin X10 and IGF1, significantly decreased the time for tumor development. Yet, insulin glargine and normal insulin, did not significantly decrease the latency time for (mammary gland) tumor development. The majority of tumors had an epithelial to mesenchymal transition phenotype (EMT), irrespective of treatment condition. Enhanced extracellular signaling related kinase (Erk) or serine/threonine kinase (Akt) mitogenic signaling was in particular present in tumors from the insulin X10 and IGF1 treatment groups. CONCLUSIONS: These data indicate that insulin-like molecules with enhanced mitogenic signaling increase the risk of breast cancer development. Moreover, the use of a tissue specific cancer model, like the p53R270H/⁺WAPCre mouse model, is relevant to assess the intrinsic pro-carcinogenic potential of mitogenic and non-mitogenic biologicals such as insulin analogues.


Subject(s)
Insulin-Like Growth Factor I/administration & dosage , Insulin/administration & dosage , Mammary Neoplasms, Animal/etiology , Milk Proteins/genetics , Tumor Suppressor Protein p53/genetics , Animals , Body Weight/drug effects , Body Weight/genetics , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cluster Analysis , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Insulin/analogs & derivatives , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...