Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
1.
Genes Dev ; 31(23-24): 2361-2375, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29317486

ABSTRACT

Both the MRTF-SRF and the YAP-TEAD transcriptional regulatory networks respond to extracellular signals and mechanical stimuli. We show that the MRTF-SRF pathway is activated in cancer-associated fibroblasts (CAFs). The MRTFs are required in addition to the YAP pathway for CAF contractile and proinvasive properties. We compared MRTF-SRF and YAP-TEAD target gene sets and identified genes directly regulated by one pathway, the other, or both. Nevertheless, the two pathways exhibit mutual dependence. In CAFs, expression of direct MRTF-SRF genomic targets is also dependent on YAP-TEAD activity, and, conversely, YAP-TEAD target gene expression is also dependent on MRTF-SRF signaling. In normal fibroblasts, expression of activated MRTF derivatives activates YAP, while activated YAP derivatives activate MRTF. Cross-talk between the pathways requires recruitment of MRTF and YAP to DNA via their respective DNA-binding partners (SRF and TEAD) and is therefore indirect, arising as a consequence of activation of their target genes. In both CAFs and normal fibroblasts, we found that YAP-TEAD activity is sensitive to MRTF-SRF-induced contractility, while MRTF-SRF signaling responds to YAP-TEAD-dependent TGFß signaling. Thus, the MRF-SRF and YAP-TEAD pathways interact indirectly through their ability to control cytoskeletal dynamics.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cancer-Associated Fibroblasts/physiology , Cytoskeleton/metabolism , DNA-Binding Proteins/metabolism , Mammary Neoplasms, Animal/physiopathology , Phosphoproteins/metabolism , Trans-Activators/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins , Cell Line, Tumor , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Mice , Phosphoproteins/genetics , Signal Transduction , TEA Domain Transcription Factors , Trans-Activators/genetics , Transcriptional Activation/genetics , Transforming Growth Factor beta1/metabolism , YAP-Signaling Proteins
2.
Genes Dev ; 27(19): 2086-98, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24065739

ABSTRACT

Serine and cysteine cathepsin (Cts) proteases are an important class of intracellular and pericellular enzymes mediating multiple aspects of tumor development. Emblematic of these is CtsB, reported to play functionally significant roles during pancreatic islet and mammary carcinogenesis. CtsC, on the other hand, while up-regulated during pancreatic islet carcinogenesis, lacks functional significance in mediating neoplastic progression in that organ. Given that protein expression and enzymatic activity of both CtsB and CtsC are increased in numerous tumors, we sought to understand how tissue specificity might factor into their functional significance. Thus, whereas others have reported that CtsB regulates metastasis of mammary carcinomas, we found that development of squamous carcinomas occurs independently of CtsB. In contrast to these findings, our studies found no significant role for CtsC during mammary carcinogenesis but revealed squamous carcinogenesis to be functionally dependent on CtsC. In this context, dermal/stromal fibroblasts and bone marrow-derived cells expressed increased levels of enzymatically active CtsC that regulated the complexity of infiltrating immune cells in neoplastic skin, development of angiogenic vasculature, and overt squamous cell carcinoma growth. These studies highlight the important contribution of tissue/microenvironment context to solid tumor development and indicate that tissue specificity defines functional significance for these two members of the cysteine protease family.


Subject(s)
Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/physiopathology , Cathepsin C/metabolism , Skin Neoplasms/physiopathology , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cathepsin C/genetics , Cell Line, Tumor , Chymases/metabolism , Female , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Humans , Leukocytes/metabolism , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/genetics , Pancreatic Elastase/metabolism
3.
Cell Mol Neurobiol ; 39(5): 605-617, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30850915

ABSTRACT

Severe and poorly treated pain often accompanies breast cancer. Thus, novel mechanisms involved in breast cancer-induced pain should be investigated. Then, it is necessary to characterize animal models that are reliable with the symptoms and progression of the disease as observed in humans. Explaining cancer-induced nociception in a murine model of breast carcinoma was the aim of this study. 4T1 (104) lineage cells were inoculated in the right fourth mammary fat pad of female BALB/c mice; after this, mechanical and cold allodynia, or mouse grimace scale (MGS) were observed for 30 days. To determine the presence of bone metastasis, we performed the metastatic clonogenic test and measure calcium serum levels. At 20 days after tumor induction, the antinociceptive effect of analgesics used to relieve pain in cancer patients (acetaminophen, naproxen, codeine or morphine) or a cannabinoid agonist (WIN 55,212-2) was tested. Mice inoculated with 4T1 cells developed mechanical and cold allodynia and increased MGS. Bone metastasis was confirmed using the clonogenic assay, and hypercalcemia was observed 20 days after cells inoculation. All analgesic drugs reduced the mechanical and cold allodynia, while the MGS was decreased only by the administration of naproxen, codeine, or morphine. Also, WIN 55,212-2 improved all nociceptive measures. This pain model could be a reliable form to observe the mechanisms of breast cancer-induced pain or to observe the efficacy of novel analgesic compounds.


Subject(s)
Mammary Neoplasms, Animal/pathology , Nociception , Acetaminophen/pharmacology , Acetaminophen/therapeutic use , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Benzoxazines/pharmacology , Benzoxazines/therapeutic use , Bone Neoplasms/blood , Bone Neoplasms/secondary , Calcium/blood , Cannabinoids/agonists , Cell Line, Tumor , Codeine/pharmacology , Codeine/therapeutic use , Disease Models, Animal , Female , Hyperalgesia/drug therapy , Hyperalgesia/etiology , Locomotion , Mammary Neoplasms, Animal/blood , Mammary Neoplasms, Animal/complications , Mammary Neoplasms, Animal/physiopathology , Mice, Inbred BALB C , Morphine/pharmacology , Morphine/therapeutic use , Morpholines/pharmacology , Morpholines/therapeutic use , Naphthalenes/pharmacology , Naphthalenes/therapeutic use , Naproxen/pharmacology , Naproxen/therapeutic use , Pain Measurement
4.
BMC Vet Res ; 12(1): 164, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27502594

ABSTRACT

BACKGROUND: Current studies report that aberrations in epigenetic regulators or chromatin modifications are related to tumor development and maintenance. EZH2 (Enhancer of zeste homolog 2) is one of the catalytic subunits of Polycomb repressive complex 2, a crucial epigenetic regulator. EZH2 has a master regulatory function in such processes as cell proliferation, stem cell differentiation, and early embryogenesis. In humans, EZH2 is linked to oncogenic function in several carcinomas, including breast cancer, and dysregulation of EZH2 has been particularly associated with loss of differentiation and the development of poorly differentiated breast cancer. In our present study, we were interested in determining whether EZH2 is increased in canine mammary tumors, which show similarities to human breast cancer. RESULTS: Investigation of the expression of EZH2 in canine mammary tumors revealed that EZH2 protein was overexpressed in canine mammary carcinomas, as in human breast cancer. In addition, the immunohistochemical expression level of EZH2 was associated with the degree of malignancy in canine mammary carcinoma. This is the first report to describe EZH2 expression in canine mammary tumors. CONCLUSIONS: Because the expression of EZH2 was similar in canine mammary carcinoma and human breast cancer, spontaneous canine mammary tumors may be a suitable model for studying EZH2 and treatment development.


Subject(s)
Dog Diseases/physiopathology , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/physiopathology , Animals , Disease Models, Animal , Dogs , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans
5.
EMBO J ; 29(24): 4106-17, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21076392

ABSTRACT

Inflammation and hypoxia are known to promote the metastatic progression of tumours. The CCAAT/enhancer-binding protein-δ (C/EBPδ, CEBPD) is an inflammatory response gene and candidate tumour suppressor, but its physiological role in tumourigenesis in vivo is unknown. Here, we demonstrate a tumour suppressor function of C/EBPδ using transgenic mice overexpressing the Neu/Her2/ERBB2 proto-oncogene in the mammary gland. Unexpectedly, this study also revealed that C/EBPδ is necessary for efficient tumour metastasis. We show that C/EBPδ is induced by hypoxia in tumours in vivo and in breast tumour cells in vitro, and that C/EBPδ-deficient cells exhibit reduced glycolytic metabolism and cell viability under hypoxia. C/EBPδ supports CXCR4 expression. On the other hand, C/EBPδ directly inhibits expression of the tumour suppressor F-box and WD repeat-domain containing 7 gene (FBXW7, FBW7, AGO, Cdc4), encoding an F-box protein that promotes degradation of the mammalian target of rapamycin (mTOR). Consequently, C/EBPδ enhances mTOR/AKT/S6K1 signalling and augments translation and activity of hypoxia-inducible factor-1α (HIF-1α), which is necessary for hypoxia adaptation. This work provides new insight into the mechanisms by which metastasis-promoting signals are induced specifically under hypoxia.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , F-Box Proteins/biosynthesis , Gene Expression Regulation , Hypoxia , Mammary Neoplasms, Animal/secondary , Neoplasm Metastasis/pathology , Ubiquitin-Protein Ligases/biosynthesis , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , F-Box-WD Repeat-Containing Protein 7 , Glycolysis , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Neoplasm Metastasis/physiopathology
6.
Nanotechnology ; 25(46): 465101, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25360614

ABSTRACT

Nanoparticles have recently gained increased attention as drug delivery systems for the treatment of cancer due to their minute size and unique chemical properties. However, very few studies have tested the biophysical changes associated with nanoparticles on metastatic cancer cells at the cellular and sub-cellular scales. Here, we investigated the mechanical and morphological properties of cancer cells by measuring the changes in cell Young's Modulus using AFM, filopodial retraction (FR) by time lapse optical light microscopy imaging and filopodial disorganization by high resolution AFM imaging of cells upon treatment with nanoparticles. In the current study, nanomechanical changes in live murine metastatic breast cancer cells (4T1) post exposure to a nanodiamond/nanoplatinum mixture dispersed in aqueous solution (DPV576), were monitored. Results showed a decrease in Young's modulus at two hours post treatment with DPV576 in a dose dependent manner. Partial FR at 20 min and complete FR at 40 min were observed. Moreover, analysis of the retraction distance (in microns) measured over time (minutes), showed that a DPV576 concentration of 15%v/v yielded the highest FR rate. In addition, DPV576 treated cells showed early signs of filopodial disorganization and disintegration. This study demonstrates the changes in cell stiffness and tracks early structural alterations of metastatic breast cancer cells post treatment with DPV576, which may have important implications in the role of nanodiamond/nanoplatinum based cancer cell therapy and sensitization to chemotherapy drugs.


Subject(s)
Drug Carriers/administration & dosage , Drug Carriers/chemistry , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Animals , Biophysical Phenomena , Cell Line, Tumor , Diamond , Mammary Neoplasms, Animal/drug therapy , Mice, Inbred BALB C , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Particle Size , Platinum , Pseudopodia/pathology , Pseudopodia/physiology
7.
BMC Vet Res ; 10: 296, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25515784

ABSTRACT

BACKGROUND: Progesterone receptors play a key role in the development of canine mammary tumours, and recent research has focussed on their possible value as therapeutic targets using antiprogestins. Cloning and sequencing of the progesterone receptor gene has shown that the receptor has two isoforms, A and B, transcribed from a single gene. Experimental studies in human breast cancer suggest that the differential expression of progesterone receptor isoforms has implications for hormone therapy responsiveness. This study examined the effects of the antiprogestin aglepristone on cell proliferation and mRNA expression of progesterone receptor isoforms A and B in mammary carcinomas in dogs treated with 20 mg/Kg of aglepristone (n = 22) or vehicle (n = 5) twice before surgery. RESULTS: Formalin-fixed, paraffin-embedded tissue samples taken before and after treatment were used to analyse total progesterone receptor and both isoforms by RT-qPCR and Ki67 antigen labelling. Both total progesterone receptor and isoform A mRNA expression levels decreased after treatment with aglepristone. Furthermore, a significant decrease in the proliferation index (percentage of Ki67-labelled cells) was observed in progesterone-receptor positive and isoform-A positive tumours in aglepristone-treated dogs. CONCLUSIONS: These findings suggest that the antiproliferative effects of aglepristone in canine mammary carcinomas are mediated by progesterone receptor isoform A.


Subject(s)
Dog Diseases/drug therapy , Estrenes/therapeutic use , Mammary Neoplasms, Animal/drug therapy , Neoadjuvant Therapy/veterinary , Receptors, Progesterone/physiology , Animals , Cell Proliferation/drug effects , Dog Diseases/physiopathology , Dogs , Female , Mammary Neoplasms, Animal/physiopathology , Neoadjuvant Therapy/methods , Polymerase Chain Reaction/veterinary , Receptors, Progesterone/metabolism
8.
Vet Pathol ; 51(4): 737-48, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24048323

ABSTRACT

Human inflammatory breast carcinoma (IBC) and canine inflammatory mammary carcinoma (IMC) are considered the most malignant types of breast cancer. IMC has similar characteristics to IBC; hence, IMC has been suggested as a model to study the human disease. To compare the angiogenic and angioinvasive features of IMC with non-IMC, 3 canine mammary tumor xenograft models in female SCID mice were developed: IMC, comedocarcinoma, and osteosarcoma. Histopathological and immunohistochemical characterization of both primary canine tumors and xenografts using cellular markers pancytokeratin, cytokeratin 14, vimentin, and α-smooth muscle actin and vascular factors (VEGF-A, VEGF-D, VEGFR-3, and COX-2) was performed. Tumor cell proliferation index was measured by the Ki-67 marker. The xenograft models reproduced histological features found in the primary canine tumor and preserved the original immunophenotype. IMC xenografts showed a high invasive character with tumor emboli in the dermis, edema, and occasional observations of ulceration. In addition, compared with osteosarcoma and comedocarcinoma, the IMC model showed the highest vascular factor expression associated with a high proliferation index. Likewise, IMC xenografts showed higher COX-2 expression associated with VEGF-D and VEGFR-3, as well as a higher presence of dermal lymphatic tumor emboli, suggesting COX-2 participation in IMC lymphangiogenesis. These results provide additional evidence to consider vascular factors, their receptors, and COX-2 as therapeutic targets for IBC.


Subject(s)
Disease Models, Animal , Dog Diseases/metabolism , Dog Diseases/physiopathology , Gene Expression Regulation, Neoplastic/physiology , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/physiopathology , Peptides/metabolism , Animals , Cyclooxygenase 2/metabolism , Dogs , Female , Heterografts/pathology , Heterografts/transplantation , Immunohistochemistry/veterinary , Intercellular Signaling Peptides and Proteins , Mice , Mice, SCID
9.
Am J Pathol ; 181(1): 278-93, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22698676

ABSTRACT

Increasing chronological age is the most significant risk factor for human cancer development. To examine the effects of host aging on mammary tumor growth, we used caveolin (Cav)-1 knockout mice as a bona fide model of accelerated host aging. Mammary tumor cells were orthotopically implanted into these distinct microenvironments (Cav-1(+/+) versus Cav-1(-/-) age-matched young female mice). Mammary tumors grown in a Cav-1-deficient tumor microenvironment have an increased stromal content, with vimentin-positive myofibroblasts (a marker associated with oxidative stress) that are also positive for S6-kinase activation (a marker associated with aging). Mammary tumors grown in a Cav-1-deficient tumor microenvironment were more than fivefold larger than tumors grown in a wild-type microenvironment. Thus, a Cav-1-deficient tumor microenvironment provides a fertile soil for breast cancer tumor growth. Interestingly, the mammary tumor-promoting effects of a Cav-1-deficient microenvironment were estrogen and progesterone independent. In this context, chemoprevention was achieved by using the mammalian target of rapamycin (mTOR) inhibitor and anti-aging drug, rapamycin. Systemic rapamycin treatment of mammary tumors grown in a Cav-1-deficient microenvironment significantly inhibited their tumor growth, decreased their stromal content, and reduced the levels of both vimentin and phospho-S6 in Cav-1-deficient cancer-associated fibroblasts. Since stromal loss of Cav-1 is a marker of a lethal tumor microenvironment in breast tumors, these high-risk patients might benefit from treatment with mTOR inhibitors, such as rapamycin or other rapamycin-related compounds (rapalogues).


Subject(s)
Aging/physiology , Anticarcinogenic Agents/therapeutic use , Caveolin 1/physiology , Mammary Neoplasms, Animal/prevention & control , Sirolimus/therapeutic use , Animals , Caveolin 1/deficiency , Female , Mammary Neoplasms, Animal/blood supply , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Knockout , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Ovariectomy , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Signal Transduction/physiology , Stromal Cells/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Microenvironment/physiology , Xenograft Model Antitumor Assays
10.
BMC Vet Res ; 9: 65, 2013 Apr 05.
Article in English | MEDLINE | ID: mdl-23561040

ABSTRACT

BACKGROUND: Tumor-associated macrophages (TAMs) have high impact on the cancer development because they can facilitate matrix invasion, angiogenesis, and tumor cell motility. It gives cancer cells the capacity to invade normal tissues and metastasize. The signaling of colony-stimulating factor-1 receptor (CSF-1R) which is an important regulator of proliferation and differentiation of monocytes and macrophages regulates most of the tissue macrophages. However, CSF-1R is expressed also in breast epithelial tissue during some physiological stages i.g.: pregnancy and lactation. Its expression has been also detected in various cancers. Our previous study has showed the expression of CSF-1R in all examined canine mammary tumors. Moreover, it strongly correlated with grade of malignancy and ability to metastasis. This study was therefore designed to characterize the role of CSF-1R in canine mammary cancer cells proliferation, apoptosis, migration, and invasion. As far as we know, the study presented hereby is a pioneering experiment in this field of veterinary medicine. RESULTS: We showed that csf-1r silencing significantly increased apoptosis (Annexin V test), decreased proliferation (measured as Ki67 expression) and decreased migration ("wound healing" assay) of canine mammary cancer cells. Treatment of these cells with CSF-1 caused opposite effect. Moreover, csf-1r knock-down changed growth characteristics of highly invasive cell lines on Matrigel matrix, and significantly decreased the ability of these cells to invade matrix. CSF-1 treatment increased invasion of cancer cells. CONCLUSION: The evidence of the expression and functional role of the CSF-1R in canine mammary cancer cells indicate that CSF-1R targeting may be a good therapeutic approach.


Subject(s)
Dog Diseases/physiopathology , Mammary Neoplasms, Animal/physiopathology , Receptor, Macrophage Colony-Stimulating Factor/physiology , Animals , Apoptosis/physiology , Blotting, Western/veterinary , Cell Line, Tumor , Cell Proliferation , Dogs , Female , Flow Cytometry/veterinary , Neoplasm Invasiveness/physiopathology , Neoplasm Metastasis/physiopathology , Real-Time Polymerase Chain Reaction/veterinary , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis
11.
FASEB J ; 25(3): 979-89, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21135040

ABSTRACT

Telomerase is tightly regulated in humans relative to mice, owing to the differential regulation of TERT genes. To explore hTERT regulation in vivo, we engineered mice with a 160-kb transgenic bacterial artificial chromosome (BAC) spanning the hTERT locus with a Renilla luciferase (Rluc) cassette downstream of its promoter. Analysis of multiple founder lines revealed that the Rluc expression profile from the transgenic hTERT reporter locus reproduced that of the native hTERT gene in all tissues and organs examined, demonstrating that genetic sequence determined the species-specific developmental regulation of the hTERT gene and that mouse epigenetic and transcription machineries faithfully regulated hTERT transcription. Thus, these mice allowed detailed analyses of developmental hTERT regulation. Both the transgenic hTERT reporter and the endogenous mTERT locus were expressed in early embryonic stages, and their mRNA levels progressively decreased throughout embryonic and postnatal development. Whereas hTERT transcription was much lower than mTERT expression in most organs, it increased significantly during postnatal development of thymus, testis, and ovary. In testis, the Rluc mRNA was enriched in elongating spermatids of seminiferous tubules. In addition, the transcription of transgenic hTERT reporter, but surprisingly not the endogenous mTERT gene, was activated during Wnt1-induced mammary tumorigenesis, allowing the monitoring of tumor development via noninvasive bioluminescent imaging. Collectively, our results demonstrate that the hTERT transgenic reporter system recapitulates the developmental regulation of the hTERT gene in a chromosomal position-independent manner and serves as a legitimate model to explore telomerase regulation in the development of normal and neoplastic tissues in vivo.


Subject(s)
Chromosomes, Artificial, Bacterial/genetics , Genes, Reporter/genetics , Mammary Neoplasms, Animal/genetics , Telomerase/genetics , Age Factors , Animals , Disease Models, Animal , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Gene Silencing , Genetic Complementation Test , Humans , Luciferases/genetics , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Transgenic , Telomerase/metabolism , Wnt1 Protein/metabolism
12.
FASEB J ; 25(8): 2626-37, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21518852

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are significantly increased in cancer patients and tumor bearing-animals. MDSCs infiltrate into tumors and promote tumor invasion and metastasis. To identify the mediator responsible for the prometastatic property of MDSCs, we used proteomics. We found neutrophilic granule protein (NGP) was decreased >2-fold in MDSCs from metastatic 4T1 tumor-bearing mice compared to nonmetastatic 67NR controls. NGP mRNA levels were decreased in bone marrow and in tumor-infiltrating MDSCs by 45 and 66%, respectively, in 4T1 tumor-bearing mice compared to 67NR controls. Interestingly, 4T1-conditioned medium reduced myeloid cell NGP expression by ∼ 40%, suggesting that a secreted factor mediates gene reduction. Sequence analysis shows a putative cystatin domain in NGP, and biochemical analysis confirms NGP a novel cathepsin inhibitor. It inhibited cathepsin B activity by nearly 40% in vitro. NGP expression in 4T1 tumor cells suppressed cell invasion, delayed primary tumor growth, and greatly reduced lung metastasis in vivo. A 2.8-fold reduction of cathepsin activity was found in tumors expressing NGP compared to controls. NGP significantly reduced tumor angiogenesis to 12.6 from 19.6 and lymphangiogenesis to 4.6 from 9.1 vessels/field. Necrosis was detectable only in NGP-expressing tumors, and the number of apoptotic cells increased to 22.4 from 8.3 in controls. Taken together, this study identifies a negative regulator of tumor metastasis in MDSCs, NGP, which is down-regulated in metastatic conditions. The finding suggests that malignant tumors promote invasion/metastasis not only through up-regulation of proteases but also down-regulation of protease inhibitors.


Subject(s)
Cystatins/physiology , Myeloid Cells/physiology , Neoplasm Metastasis/prevention & control , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cell Line, Tumor , Cell Proliferation , Cystatins/genetics , Female , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/blood supply , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Invasiveness/prevention & control , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Metastasis/physiopathology , Neovascularization, Pathologic , Protease Inhibitors/metabolism , Proteomics
13.
J Biomed Biotechnol ; 2012: 786417, 2012.
Article in English | MEDLINE | ID: mdl-23093865

ABSTRACT

Animal surface temperature profile captured using infrared camera is helpful for the assessment of physiological responses associated with the regulation of body temperature. Diagnosing breast cancer in early stage itself has a greater effect on the prognosis. In this work, asymmetrical temperature distribution analysis of chemical carcinogen 7,12-dimethyl benz(a)anthracene-induced in the lower right flank region of Wistar rats (n = 6) was carried out to test the potential of thermography in diagnosing mammary cancer and tumor growth over a period of nine weeks in comparison with histopathology results as standard. Temperature difference between the tumor induced lower right and left side of flank region was significant (with P value <0.001), whereas in the abdomen and shoulder there was no significant difference in temperature between right and left sides. Percentage of asymmetrical temperature difference in the tumor induced lower flank region was 0.5 to 2%, whereas in the other regions it was <0.5%. Green pixel distribution in RGB color histogram was asymmetrical in the tumor induced lower flank region. Temperature reduction was observed in the tumor induced region after the seventh day of carcinogen induction. Asymmetrical thermogram analysis is the best method of diagnosing mammary cancer and for studying tumor development.


Subject(s)
Biomarkers, Tumor/blood , Carcinoembryonic Antigen/blood , Mammary Neoplasms, Animal/diagnosis , Mammary Neoplasms, Animal/physiopathology , Thermography/methods , 9,10-Dimethyl-1,2-benzanthracene , Animals , Diagnosis, Computer-Assisted/methods , Female , Mammary Neoplasms, Animal/chemically induced , Rats , Rats, Wistar , Reproducibility of Results , Sensitivity and Specificity
14.
Cancer Res ; 82(3): 365-376, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34903599

ABSTRACT

In breast cancer, the type and distribution of infiltrating immune cells are associated with clinical outcome. Moreover, cancers with abundant tumor-infiltrating lymphocytes (TIL) are more likely to respond to immunotherapy, whereas those in which CD8+ T cells are completely absent (deserts) or excluded are less likely to respond. Detailed understanding of this biology is limited by a lack of preclinical breast cancer models that recapitulate TIL distributions and their associated biology. Here we established mammary tumor-derived transplants (mTDT) from 12 Trp53-null mammary tumors in syngeneic BALB/cJ mice and examined the stability of their growth rate, TIL distribution, and transcriptomic profiles. All mTDTs were estrogen receptor negative. Half of the parental tumors were classified as infiltrated, and the rest were divided between excluded and desert phenotypes. After two orthotopic passages, most (70%) mTDT from infiltrated parents recapitulated this pattern, whereas the desert or excluded parental patterns were maintained in about half of daughter mTDT. Approximately 30% of mTDT gave rise to lung or liver metastases, although metastasis was not associated with a TIL phenotype. Unsupervised transcriptomic analysis clustered mTDT according to their TIL spatial patterns. Infiltrated mTDT transplanted subcutaneously or orthotopically were resistant to anti-PD-L1. Profiling implicated prolonged antigen stimulation and loss of effector function of lymphocytes rather than T-cell exhaustion in the lack of response of infiltrated mTDT to checkpoint blockade. In summary, the molecular diversity and immune complexity of mTDT should facilitate the dissection of mechanisms of breast cancer response to immunotherapies. SIGNIFICANCE: A set of diverse preclinical models of breast cancer is characterized to enable mechanistic dissection of tumor-immune interactions and to improve the efficacy of immunotherapies.


Subject(s)
Breast Neoplasms/physiopathology , Lymphocytes, Tumor-Infiltrating/immunology , Mammary Neoplasms, Animal/physiopathology , Animals , Disease Models, Animal , Female , Mice , Mice, Nude , Tumor Microenvironment
15.
Histochem Cell Biol ; 136(6): 699-708, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22005837

ABSTRACT

Gel-forming mucins are large, high molecular weight, and heavily O-glycosylated proteins that are responsible for the rheological properties of mucus gel. Among them, the mucin MUC5B has been implicated in breast cancer and cystic fibrosis. We obtained a new polyclonal serum, named CP1, which was isolated from a rabbit immunized with a mouse Muc5b peptide. The immunoprofile of Muc5b was determined on paraffin-embedded and frozen mouse tissue sections and showed a similar expression pattern in mouse to that in the human. The "nonmammary" mucin Muc5b was detected in all mammary tumors analyzed from MMTV-ras mice, suggesting that the CP1 antibody is a valuable tool for investigating the involvement of this mucin in mammary cancer. We also found that uninfected Cftr( -/- ) mice harbored more Clara cells, which were Muc5b-positive, than did their wild-type control littermates. The number of Muc5b-positive cells increased in Cftr( -/- ) mice infected experimentally with Pseudomonas aeruginosa, and the mice developed mucus plugs in their bronchi and bronchioles with a high frequency of Muc5b content (87%, Cohen's kappa = 0.82; p < 0.0001). These findings suggest that mice genetically deficient in the Cftr gene are predisposed to develop mucus plugs and that MUC5B may provide a valuable target for decreasing mucus viscosity in cystic fibrosis.


Subject(s)
Gene Expression Regulation , Mammary Neoplasms, Animal/physiopathology , Mucin-5B/metabolism , ras Proteins/metabolism , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Specificity/genetics , Female , Immunohistochemistry , Mammary Neoplasms, Animal/metabolism , Mammary Tumor Virus, Mouse , Mice , Mice, Inbred C57BL , Mice, Inbred CFTR , Mice, Knockout , Mucin-5B/genetics , Real-Time Polymerase Chain Reaction
16.
Sci Rep ; 11(1): 23277, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857853

ABSTRACT

The aim of this study was to evaluate renal hemodynamics, routine clinical and laboratory parameters used to estimate renal function, and clinical evolution during six months in bitches with mammary carcinomas that underwent mastectomy and were treated (TG) or not (CG) with carprofen for three months after surgery. Twenty-six bitches with mammary carcinoma were equally distributed into TG that received carprofen 4.4 mg/kg/day for 90 days and CG that did not receive anti-inflammatory medication. Renal artery Doppler flowmetry, contrast-enhanced ultrasound (CEUS) of renal parenchyma, haematological, biochemical and clinical analyses were obtained once a month. These data were compared between groups and time via analysis of variance (ANOVA) in a completely randomized design with repeated measures (P < 0.05). On B-mode ultrasound, the area of the renal artery was greater (P = 0.0003) in the TG. Regarding laboratory findings, haematocrit and haemoglobin were similar in both groups, showing a significant and gradual increase after three months of treatment; MCV, MHC, and MCHC were increased (P < 0.05) and lymphocyte and band counts decreased (P < 0.05) in the TG. Regarding biochemical tests, ALT was the only parameter with a significant difference, being higher (P = 0.0272) in the treated group. It can be concluded that the use of carprofen for 90 days causes minimal changes in renal perfusion, erythrocyte parameters and ALT activity, and reduces the proportion of blood inflammatory cells. Therefore, use of this medication can be carried out safely in patients who require auxiliary cancer treatment.


Subject(s)
Carbazoles/administration & dosage , Carbazoles/adverse effects , Carcinoma/drug therapy , Dog Diseases/drug therapy , Kidney/blood supply , Kidney/diagnostic imaging , Mammary Neoplasms, Animal/drug therapy , Renal Circulation/drug effects , Ultrasonography, Doppler , Animals , Carcinoma/physiopathology , Carcinoma/surgery , Dog Diseases/physiopathology , Dog Diseases/surgery , Dogs , Female , Mammary Glands, Animal/surgery , Mammary Neoplasms, Animal/physiopathology , Mammary Neoplasms, Animal/surgery , Time Factors
17.
J Exp Med ; 193(6): 727-40, 2001 Mar 19.
Article in English | MEDLINE | ID: mdl-11257139

ABSTRACT

In human breast carcinomas, overexpression of the macrophage colony-stimulating factor (CSF-1) and its receptor (CSF-1R) correlates with poor prognosis. To establish if there is a causal relationship between CSF-1 and breast cancer progression, we crossed a transgenic mouse susceptible to mammary cancer with mice containing a recessive null mutation in the CSF-1 gene (Csf1(op)) and followed tumor progression in wild-type and null mutant mice. The absence of CSF-1 affects neither the incidence nor the growth of the primary tumors but delayed their development to invasive, metastatic carcinomas. Transgenic expression of CSF-1 in the mammary epithelium of both Csf1(op)/Csf1(op) and wild-type tumor-prone mice led to an acceleration to the late stages of carcinoma and to a significant increase in pulmonary metastasis. This was associated with an enhanced infiltration of macrophages into the primary tumor. These studies demonstrate that the growth of mammary tumors and the development to malignancy are separate processes and that CSF-1 selectively promotes the latter process. CSF-1 may promote metastatic potential by regulating the infiltration and function of tumor-associated macrophages as, at the tumor site, CSF-1R expression was restricted to macrophages. Our data suggest that agents directed at CSF-1/CSF-1R activity could have important therapeutic effects.


Subject(s)
Lung Neoplasms/secondary , Macrophage Colony-Stimulating Factor/physiology , Mammary Neoplasms, Animal/physiopathology , Animals , Disease Progression , Female , Macrophage Colony-Stimulating Factor/genetics , Male , Mammary Neoplasms, Animal/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic
18.
BMC Vet Res ; 6: 15, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20226097

ABSTRACT

BACKGROUND: Cell adhesion is an important regulator of cell growth and motility. Recently the hepatocyte cell adhesion molecules 1 and 2 (HEPACAM1 and 2), members of the immunoglobulin family of adhesion genes, have been identified. HEPACAM1 is involved in negative cell cycle regulation via p53, p21 and p27 signalling but also mediates increased human breast cancer cell spread. The role and expression pattern of HEPACAM2 has not been analyzed so far. In the present study we quantified gene expression levels of HEPACAM1 and 2 to evaluate their possible role during the carcinogenesis of canine mammary tumours. RESULTS: Adenomas displayed increased HEPACAM1 and 2 mRNA expression levels and decreased HEPACAM1 protein expression levels when compared to normal gland, carcinomas and lymph node metastases. In contrast, metastatic carcinomas, intravascular tumour cells and lymph node metastases had HEPACAM 1 protein and mRNA expression levels similar to normal gland but decreased HEPACAM2 mRNA expression when compared to normal gland of the same dog. CONCLUSIONS: HEPACAM1 and 2 seem to be important for cell-cell adhesion of normal and neoplastic canine mammary cells. The loss of HEPACAM1 protein expression in adenomas but not in carcinomas questions its role as a tumour suppressor at late stages of malignant transformation and indicates that it might rather be involved in physiologic mammary cell adhesion and canine mammary tumour metastasis. Furthermore, it can be speculated, whether HEPACAM2 plays a different role in malignancy and metastasis of canine mammary tumours since its transcriptional levels are different in carcinomas and their lymph node metastases when compared to HEPACAM1.


Subject(s)
Adenoma/physiopathology , Dog Diseases/physiopathology , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/physiopathology , Mammary Neoplasms, Animal/physiopathology , Proteins/metabolism , Animals , Dogs , Female , Lymphatic Metastasis/physiopathology
19.
Vet Pathol ; 47(1): 98-101, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20080488

ABSTRACT

RAD51 is a key enzyme of homologous recombination and repair of DNA double-strand breaks. RAD51 mRNA expression levels are significantly increased in laser-microdissected mammary simple carcinomas and their lymph node metastases when compared to adenomas or nonneoplastic mammary gland of the same dog. Here, RAD51 protein expression was analyzed by immunohistochemistry in paraffin-embedded mammary carcinomas and their lymph node metastases of 40 dogs, adenomas of 48 dogs, and nonneoplastic mammary gland of 88 dogs. Number of cells with nuclear RAD51 expression was significantly (P < or = .05) increased in carcinomas when compared to adenomas and metastases. In contrast, no significant differences in the number of RAD51-expressing cells were detected when metastases were compared with adenomas and nonneoplastic gland. RAD51 expression in carcinomas was correlated with expression in metastases but not with histologic grade. In conclusion, the increased number of RAD51-expressing cells in carcinomas might indicate genomic instability in these cells. Nevertheless, the increased RAD51 mRNA expression in metastases could not be confirmed by immunohistochemistry.


Subject(s)
Dog Diseases/physiopathology , Mammary Neoplasms, Animal/physiopathology , Rad51 Recombinase/biosynthesis , Adenoma/enzymology , Adenoma/pathology , Adenoma/physiopathology , Animals , Carcinoma/enzymology , Carcinoma/pathology , Carcinoma/physiopathology , Dog Diseases/enzymology , Dog Diseases/pathology , Dogs , Female , Gene Expression Regulation, Neoplastic/physiology , Lymphatic Metastasis , Mammary Glands, Animal/enzymology , Mammary Glands, Animal/pathology , Mammary Glands, Animal/physiopathology , Mammary Neoplasms, Animal/enzymology , Mammary Neoplasms, Animal/pathology , Rad51 Recombinase/physiology
20.
Vet Comp Oncol ; 18(3): 281-291, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31606921

ABSTRACT

The present work describes the microtomographic characterization of macro- and microcalcifications present in excised canine mammary glands. In human breast cancer, microcalcifications are highly relevant for diagnosis and prognosis, often being the sole element determining biopsy. Canine mammary tumours are considered a model for human breast cancer, but the morphological features of calcifications had still to be studied in this species. The objective of this research is to contribute to the characterization of the mineralization features of the canine mammary gland. In the present study, the excised mammary glands of 33 bitches underwent fluoroscopic examination. In 30 of the samples, the presence of calcification was suspected, and multiple biopsies were taken of these areas. Biopsy fragments underwent microtomographic scanning. Microcalcifications were found in non-neoplastic glandular tissue, benign and malign lesions, as it is known to happen in humans. Qualitative evaluation regarding morphology of the imaged calcifications showed similarities to breast cancer findings, based on the BI-RADS 2013 classification, such as pleomorphism and shape. No differences in the quantitative morphological parameters of volume, surface, surface/volume, SMI and structure thickness were found when macrocalcifications were considered. However, although significant differences existed in these parameters between microcalcifications from malignant canine mammary tumours and the two other groups, none were found between non-neoplastic and benign tumours. Findings further support the use of this spontaneous animal model for the study of human breast cancer, considering how clinically relevant microcalcifications are in humans.


Subject(s)
Calcinosis/veterinary , Dog Diseases/physiopathology , Mammary Neoplasms, Animal/physiopathology , Animals , Biopsy/veterinary , Calcinosis/physiopathology , Dogs , Female , Fluoroscopy/veterinary , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL