Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 645
Filter
1.
Biochem Biophys Res Commun ; 569: 10-16, 2021 09 10.
Article in English | MEDLINE | ID: mdl-34216992

ABSTRACT

Cancer cells require oxygen and nutrients for growth, making angiogenesis one of the essential components of tumor growth. Gangliosides, constituting membrane lipid rafts, regulate intracellular signal transduction and are involved in the malignancy of cancer cells. While endothelial cells, as well as cancer cells, express vast amounts of gangliosides, the precise function of endothelial gangliosides in angiogenesis remains unclear. In this study, we focused on gangliosides of vascular endothelial cells and analyzed their functions on tumor angiogenesis. In human breast cancer, GM3 synthase was highly expressed in vascular endothelial cells as well as immune cells. Angiogenesis increased in GM3S-KO mice. In BAEC, RNA interference of GM3S showed increased cellular invasion and oxidative stress tolerance through activation of ERK. In the breast cancer model, GM3-KO mice showed an increase in tumor growth and angiogenesis. These results suggest that the endothelial ganglioside GM3 regulates tumor angiogenesis by suppressing cellular invasion and oxidative stress tolerance in endothelial cells.


Subject(s)
Endothelial Cells/metabolism , G(M3) Ganglioside/metabolism , Neovascularization, Pathologic/metabolism , Animals , Cattle , Cell Line, Tumor , Cell Survival/genetics , Cells, Cultured , Kaplan-Meier Estimate , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Neovascularization, Pathologic/genetics , Sialyltransferases/genetics , Sialyltransferases/metabolism , Tumor Burden/genetics , Polypeptide N-acetylgalactosaminyltransferase
2.
J Immunol ; 205(8): 2301-2311, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32938724

ABSTRACT

Tie2-expressing monocytes/macrophages (TEMs) are a distinct subset of proangiogenic monocytes selectively recruited to tumors in breast cancer. Because of the hypoxic nature of solid tumors, we investigated if oxygen, via hypoxia-inducible transcription factors HIF-1α and HIF-2α, regulates TEM function in the hypoxic tumor microenvironment. We orthotopically implanted PyMT breast tumor cells into the mammary fat pads of syngeneic LysMcre, HIF-1α fl/fl /LysMcre, or HIF-2α fl/fl /LysMcre mice and evaluated the tumor TEM population. There was no difference in the percentage of tumor macrophages among the mouse groups. In contrast, HIF-1α fl/fl /LysMcre mice had a significantly smaller percentage of tumor TEMs compared with control and HIF-2α fl/fl /LysMcre mice. Proangiogenic TEMs in macrophage HIF-2α-deficient tumors presented significantly more CD31+ microvessel density but exacerbated hypoxia and tissue necrosis. Reduced numbers of proangiogenic TEMs in macrophage HIF-1α-deficient tumors presented significantly less microvessel density but tumor vessels that were more functional as lectin injection revealed more perfusion, and functional electron paramagnetic resonance analysis revealed more oxygen in those tumors. Macrophage HIF-1α-deficient tumors also responded significantly to chemotherapy. These data introduce a previously undescribed and counterintuitive prohypoxia role for proangiogenic TEMs in breast cancer which is, in part, suppressed by HIF-2α.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/immunology , Macrophages/immunology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/immunology , Neoplasm Proteins/immunology , Receptor, TIE-2/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Female , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/immunology , Macrophages/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Neoplasm Proteins/genetics , Oxygen/immunology , Receptor, TIE-2/genetics
3.
J Hematol Oncol ; 13(1): 123, 2020 09 14.
Article in English | MEDLINE | ID: mdl-32928251

ABSTRACT

BACKGROUND: Conventional therapeutic approaches for tumor angiogenesis, which are primarily focused on the inhibition of active angiogenesis to starve cancerous cells, target the vascular endothelial growth factor signaling pathway. This aggravates hypoxia within the tumor core and ultimately leads to increased tumor proliferation and metastasis. To overcome this limitation, we developed nanoparticles with antiseptic activity that target tumor vascular abnormalities. METHODS: Ferritin-based protein C nanoparticles (PCNs), known as TFG and TFMG, were generated and tested in Lewis lung carcinoma (LLC) allograft and MMTV-PyMT spontaneous breast cancer models. Immunohistochemical analysis was performed on tumor samples to evaluate the tumor vasculature. Western blot and permeability assays were used to explore the role and mechanism of the antitumor effects of PCNs in vivo. For knocking down proteins of interest, endothelial cells were transfected with siRNAs. Statistical analysis was performed using one-way ANOVA followed by post hoc Dunnett's multiple comparison test. RESULTS: PCNs significantly inhibited hypoxia and increased pericyte coverage, leading to the inhibition of tumor growth and metastasis, while increasing survival in LLC allograft and MMTV-PyMT spontaneous breast cancer models. The coadministration of cisplatin with PCNs induced a synergistic suppression of tumor growth by improving drug delivery as evidenced by increased blood prefusion and decreased vascular permeability. Moreover, PCNs altered the immune cell profiles within the tumor by increasing cytotoxic T cells and M1-like macrophages with antitumor activity. PCNs induced PAR-1/PAR-3 heterodimerization through EPCR occupation and PAR-1 activation, which resulted in Gα13-RhoA-mediated-Tie2 activation and stabilized vascular tight junctions via the Akt-FoxO3a signaling pathway. CONCLUSIONS: Cancer treatment targeting the tumor vasculature by inducing antitumor immune responses and enhancing the delivery of a chemotherapeutic agent with PCNs resulted in tumor regression and may provide an effective therapeutic strategy.


Subject(s)
Apoferritins/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Nanoparticles/therapeutic use , Neovascularization, Pathologic/drug therapy , Protein C/therapeutic use , Receptor, TIE-2/physiology , Vascular Remodeling/drug effects , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/therapeutic use , Apoferritins/administration & dosage , Bevacizumab/therapeutic use , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/pathology , Cell Hypoxia/drug effects , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Coculture Techniques , Drug Delivery Systems , Drug Synergism , Endothelial Cells/drug effects , Female , Male , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Proteins/physiology , Pericytes/metabolism , Protein C/administration & dosage , Specific Pathogen-Free Organisms , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
APMIS ; 128(5): 387-389, 2020 May.
Article in English | MEDLINE | ID: mdl-32105360

ABSTRACT

Tumor tissue often has an insufficient nutritional supply, in part due to compression of the vascular network from an increased interstitial fluid pressure. We have shown that the antisecretory factor peptide AF-16 can reduce this pressure in experimental rat breast tumors. In this work we studied if AF-16 administration opened up to an increased vascular volume in these tumors. Sprague-Dawley rats were given dimethylbenxanthracene and developed mammary tumors which were studied. Evans Blue was used as an intravascular volume indicator. Under anesthesia the rats were given AF-16 or solvent intranasally, and Evans Blue was injected i.v. 45 min later. Tumors and various organs were dissected and Evans Blue was extracted and colorimetrically quantified. Tumors had a significantly higher vascular volume after AF-16 administration as compared to other organs. Liver and renal vascular volumes were also increased but to a lesser degree than in the tumors. The results indicate that AF16 could be a candidate for increasing vascular access for chemotherapy in cancer therapy.


Subject(s)
Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Peptides/administration & dosage , Vascular Patency/drug effects , Animals , Female , Kidney/pathology , Liver/pathology , Neuropeptides , Rats , Rats, Sprague-Dawley
5.
Matrix Biol ; 83: 26-47, 2019 10.
Article in English | MEDLINE | ID: mdl-31288084

ABSTRACT

Metastasis is a major cause of death in cancer patients. The extracellular matrix molecule tenascin-C is a known promoter of metastasis, however the underlying mechanisms are not well understood. To further analyze the impact of tenascin-C on cancer progression we generated MMTV-NeuNT mice that develop spontaneous mammary tumors, on a tenascin-C knockout background. We also developed a syngeneic orthotopic model in which tumor cells derived from a MMTV-NeuNT tumor. Tumor cells were transfected with control shRNA or with shRNA to knockdown tenascin-C expression and, were grafted into the mammary gland of immune competent, wildtype or tenascin-C knockout mice. We show that stromal-derived tenascin-C increases metastasis by reducing apoptosis and inducing the cellular plasticity of cancer cells located in pulmonary blood vessels invasions (BVI), before extravasation. We characterized BVI as organized structures of tightly packed aggregates of proliferating tumor cells with epithelial characteristics, surrounded by Fsp1+ cells, internally located platelets and, a luminal monolayer of endothelial cells. We found extracellular matrix, in particular, tenascin-C, between the stromal cells and the tumor cell cluster. In mice lacking stromal-derived tenascin-C, the organization of pulmonary BVI was significantly affected, revealing novel functions of host-derived tenascin-C in supporting the integrity of the endothelial cell coat, increasing platelet abundance, tumor cell survival, epithelial plasticity, thereby promoting overall lung metastasis. Many effects of tenascin-C observed in BVI including enhancement of cellular plasticity, survival and migration, could be explained by activation of TGF-ß signaling. Finally, in several human cancers, we also observed BVI to be surrounded by an endothelial monolayer and to express tenascin-C. Expression of tenascin-C is specific to BVI and is not observed in lymphatic vascular invasions frequent in breast cancer, which lack an endothelial lining. Given that BVI have prognostic significance for many tumor types, such as shorter cancer patient survival, increased metastasis, vessel occlusion, and organ failure, our data revealing a novel mechanism by which stromal tenascin-C promotes metastasis in human cancer, may have potential for diagnosis and therapy.


Subject(s)
Blood Vessels/pathology , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/genetics , Tenascin/genetics , Animals , Blood Vessels/metabolism , Cell Line, Tumor , Female , Gene Knockout Techniques , Humans , Lung Neoplasms/blood supply , Lung Neoplasms/genetics , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Transgenic , Rats , Signal Transduction , Stromal Cells , Tenascin/metabolism , Transforming Growth Factor beta/metabolism
6.
Exp Cell Res ; 380(2): 141-148, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31034805

ABSTRACT

Increasing preclinical and clinical studies revealed that many tumor models had resistance to anti-VEGF-A and anti-VEGF-R2 therapies. Studies have shown that simultaneously blocked DLL4-Notch and VEGF signaling pathways can synergistically inhibit density and function of tumor blood vessels and reduce tumor growth rate. We successfully developed a bispecific monoclonal antibody (named HB-32) that targeting both human DLL4 and human VEGF. HB-32 showed high binding affinity to VEGF and DLL4. Furthermore, HB-32 inhibited proliferation, migration and tube formation of HUVEC. Finally, in vivo xenograft studies demonstrated that HB-32 inhibited proliferation of breast cancer cells (MDA-MB-231) and induced tumor cell apoptosis more efficiently than an anti-VEGF antibody or anti-DLL4 antibody alone. These findings indicate that our bispecific antibody provide a potential treatment for breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/antagonists & inhibitors , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/isolation & purification , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/isolation & purification , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Female , HEK293 Cells , Humans , Injections, Subcutaneous , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Vascular Endothelial Growth Factor A/metabolism
7.
J Clin Invest ; 129(4): 1654-1670, 2019 03 11.
Article in English | MEDLINE | ID: mdl-30855280

ABSTRACT

In tumors, extravascular fibrin forms provisional scaffolds for endothelial cell (EC) growth and motility during angiogenesis. We report that fibrin-mediated angiogenesis was inhibited and tumor growth delayed following postnatal deletion of Tgfbr2 in the endothelium of Cdh5-CreERT2 Tgfbr2fl/fl mice (Tgfbr2iECKO mice). ECs from Tgfbr2iECKO mice failed to upregulate the fibrinolysis inhibitor plasminogen activator inhibitor 1 (Serpine1, also known as PAI-1), due in part to uncoupled TGF-ß-mediated suppression of miR-30c. Bypassing TGF-ß signaling with vascular tropic nanoparticles that deliver miR-30c antagomiRs promoted PAI-1-dependent tumor growth and increased fibrin abundance, whereas miR-30c mimics inhibited tumor growth and promoted vascular-directed fibrinolysis in vivo. Using single-cell RNA-Seq and a NanoString miRNA array, we also found that subtypes of ECs in tumors showed spectrums of Serpine1 and miR-30c expression levels, suggesting functional diversity in ECs at the level of individual cells; indeed, fresh EC isolates from lung and mammary tumor models had differential abilities to degrade fibrin and launch new vessel sprouts, a finding that was linked to their inverse expression patterns of miR-30c and Serpine1 (i.e., miR-30chi Serpine1lo ECs were poorly angiogenic and miR-30clo Serpine1hi ECs were highly angiogenic). Thus, by balancing Serpine1 expression in ECs downstream of TGF-ß, miR-30c functions as a tumor suppressor in the tumor microenvironment through its ability to promote fibrin degradation and inhibit blood vessel formation.


Subject(s)
Endothelial Cells/metabolism , Mammary Neoplasms, Experimental/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/metabolism , Plasminogen Activator Inhibitor 1/metabolism , RNA, Neoplasm/metabolism , Transforming Growth Factor beta/metabolism , Animals , Endothelial Cells/pathology , Female , Gene Deletion , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Plasminogen Activator Inhibitor 1/genetics , RNA, Neoplasm/genetics , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/metabolism , Transforming Growth Factor beta/genetics
8.
Nat Cell Biol ; 21(2): 238-250, 2019 02.
Article in English | MEDLINE | ID: mdl-30664790

ABSTRACT

The presence of disseminated tumour cells (DTCs) in bone marrow is predictive of poor metastasis-free survival of patients with breast cancer with localized disease. DTCs persist in distant tissues despite systemic administration of adjuvant chemotherapy. Many assume that this is because the majority of DTCs are quiescent. Here, we challenge this notion and provide evidence that the microenvironment of DTCs protects them from chemotherapy, independent of cell cycle status. We show that chemoresistant DTCs occupy the perivascular niche (PVN) of distant tissues, where they are protected from therapy by vascular endothelium. Inhibiting integrin-mediated interactions between DTCs and the PVN, driven partly by endothelial-derived von Willebrand factor and vascular cell adhesion molecule 1, sensitizes DTCs to chemotherapy. Importantly, chemosensitization is achieved without inducing DTC proliferation or exacerbating chemotherapy-associated toxicities, and ultimately results in prevention of bone metastasis. This suggests that prefacing adjuvant therapy with integrin inhibitors is a viable clinical strategy to eradicate DTCs and prevent metastasis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Blood Vessels/drug effects , Mammary Neoplasms, Experimental/drug therapy , Tumor Microenvironment/drug effects , Animals , Blood Vessels/metabolism , Blood Vessels/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Integrins/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Transgenic , Paclitaxel/administration & dosage
9.
Nanomedicine ; 15(1): 252-263, 2019 01.
Article in English | MEDLINE | ID: mdl-30359756

ABSTRACT

Nonintrusive and precise imaging for tumor angiogenesis is critical in accurate assessment of cancer diagnosis and prognosis. However, reticulo-endothelial system (RES) capture and inadequate accumulation remain major bottlenecks for current nanoparticle to retain at tumor angiogenesis site. Herein, we report the ultrasmall contrast agent (cNGR-Au:Gd@GSH NMs) could accumulate at tumor vasculature site and enhance the tumor angiogenesis-contrast. It is demonstrated that by loading Au and Gd atom into the naturally-occurring glutathione (GSH) shell with cNGR peptide modification, cNGR-Au:Gd@GSH NMs exhibit the high X-ray photon absorption, longer rotational correlation time and efficient tumor vascular endothelia cell targeting. In vivo studies further indicate the cNGR-Au:Gd@GSH NMs prominently enhance tumor angiogenesis-contrast both on the computed tomography (CT) and magnetic resonance imaging (MRI) modalities by escaping the RES capture and target delivering. Our data imply that the cNGR-Au:Gd@GSH NMs may serve as the high-efficiency contrast agent to assess tumor angiogenesis in a nonintrusive technique.


Subject(s)
Contrast Media/pharmacokinetics , Gadolinium/chemistry , Gold/chemistry , Mammary Neoplasms, Experimental/blood supply , Metal Nanoparticles/chemistry , Neovascularization, Pathologic/diagnosis , Animals , Apoptosis , Cell Proliferation , Female , HT29 Cells , Hep G2 Cells , Humans , Magnetic Resonance Imaging , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Imaging , Neovascularization, Pathologic/diagnostic imaging , Tissue Distribution , Tomography, X-Ray Computed , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Mol Pharm ; 15(11): 5277-5290, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30234992

ABSTRACT

Vascular endothelial dysfunction and platelet activation play a key role in tumor metastasis, and therefore, both of these processes are considered important therapeutic targets in cancer. The aim of our studies was to analyze antimetastatic activity of combination therapy using nitric oxide donor DETA/NO and antiplatelet drug clopidogrel. Nitric oxide acts as a vasoprotective mediator, while clopidogrel inhibits ADP-mediated platelet aggregation. 4T1-luc2-tdTomato cell line transplanted intravenously (i.v.) and 4T1 cell line transplanted orthotopically were used as metastatic mammary gland cancer models. Moreover, antiaggregation action of compounds was tested ex vivo on the blood samples taken from breast cancer patients. We have shown that in selected dosage regimes, DETA/NO combined with clopidogrel significantly reduced lung metastatic foci formation in an i.v. model, and such inhibition was transiently observed also in an orthotopic model. The antimetastatic effect was correlated with a significant increase of prostacyclin (PGI2) metabolite and reduction of endothelin-1, sE-selectin, sI-CAM, and TGF-ß plasma levels as well as decreased V-CAM expression on the endothelium. Combination therapy decreased fibrinogen binding to the resting platelets at the early stage of tumor progression (day 14). However, at the later stages (days 21 and 28), the markers of platelet activation were detected (increased JON/A antibody bound, P-selectin level, binding of fibrinogen, and vWf). Decreased aggregation as well as a lower release of TGF-ß were detected in platelets incubated ex vivo with compounds tested from metastatic breast cancer patients. Although combination therapy increases E-cadherin, the increase of N-cadherin and α-SMA in tumor tissue was also observed. The results showed that at the early stages of tumor progression, combined therapy with DETA/NO and clopidogrel improves vasoprotective and antiplatelet activity. However, in advanced tumors, some adverse effects toward platelet activation can be observed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/drug therapy , Mammary Neoplasms, Experimental/drug therapy , Nitric Oxide Donors/administration & dosage , Platelet Aggregation Inhibitors/administration & dosage , Animals , Breast Neoplasms/blood , Cell Line, Tumor/transplantation , Clopidogrel/administration & dosage , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Female , Humans , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Platelet Activation/drug effects , Platelet-Rich Plasma/drug effects , Triazenes/administration & dosage
11.
Anticancer Drugs ; 29(10): 956-964, 2018 11.
Article in English | MEDLINE | ID: mdl-30134286

ABSTRACT

OBJECTIVE: Cetirizine (CET) and thalidomide (THA) have been previously found to influence angiogenesis. The present study aimed to assess the ability of these drugs to influence mammary carcinogenesis in rats. MATERIALS AND METHODS: Sixty Sprague-Dawley female rats, aged 8 weeks, received 15 mg of 7,12-dimethylbenz(a)anthracene (DMBA) intragastrically. CET and THA (1.0 and 3.0 mg/kg, respectively) were administered orally for 118 days after DMBA administration. At the end of the treatment period, mammary tumors were counted and weighed, and their morphology was analyzed using light microscopy. In tumor tissue, proliferation and apoptotic indices and microvessel density were determined using immunohistochemical techniques; the levels of angiogenic factors, vascular endothelial growth factor and basic fibroblast growth factor, were measured by western blotting. RESULTS: CET and THA, administered separately, failed to influence tumor formation and angiogenesis. In contrast, the drug combination decreased latency to first tumor (significant difference from vehicle-treated control and groups that received either drug alone, P<0.01) and significantly lowered tumor number per rat, number of malignant tumors per rat, tumor burden, and tumor number per tumor-bearing animal (P<0.05 or <0.01). In tissue of malignant tumors, the drug combination decreased the number of proliferating cells, microvessel density, and levels of vascular endothelial growth factor and basic fibroblast growth factor and stimulated apoptosis (difference from all other groups, P<0.01). CONCLUSION: It was shown for the first time that H1-antagonist and THA synergistically inhibit DMBA-induced mammary carcinogenesis; this effect was associated with a decrease in tumor angiogenesis. Further study of the anticancer and antiangiogenic activity of the combination may provide a new approach to breast cancer treatment.


Subject(s)
Cetirizine/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Thalidomide/pharmacology , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , Cetirizine/administration & dosage , Drug Synergism , Female , Fibroblast Growth Factor 2/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley , Thalidomide/administration & dosage , Vascular Endothelial Growth Factor A/metabolism
12.
Br J Cancer ; 118(8): 1098-1106, 2018 04.
Article in English | MEDLINE | ID: mdl-29576623

ABSTRACT

BACKGROUND: Optoacoustic tomography (OT) of breast tumour oxygenation is a promising new technique, currently in clinical trials, which may help to determine disease stage and therapeutic response. However, the ability of OT to distinguish breast tumours displaying different vascular characteristics has yet to be established. The aim of the study is to prove OT as a sensitive technique for differentiating breast tumour models with manifestly different vasculatures. METHODS: Multispectral OT (MSOT) was performed in oestrogen-dependent (MCF-7) and oestrogen-independent (MDA-MB-231) orthotopic breast cancer xenografts. Total haemoglobin (THb) and oxygen saturation (SO2MSOT) were calculated. Pathological and biochemical evaluation of the tumour vascular phenotype was performed for validation. RESULTS: MCF-7 tumours show SO2MSOT similar to healthy tissue in both rim and core, despite significantly lower THb in the core. MDA-MB-231 tumours show markedly lower SO2MSOT with a significant rim-core disparity. Ex vivo analysis revealed that MCF-7 tumours contain fewer blood vessels (CD31+) that are more mature (CD31+/aSMA+) than MDA-MB-231. MCF-7 presented higher levels of stromal VEGF and iNOS, with increased NO serum levels. The vasculogenic process observed in MCF-7 was consistent with angiogenesis, while MDA-MB-231 appeared to rely more on vascular mimicry. CONCLUSIONS: OT is sensitive to differences in the vascular phenotypes of our breast cancer models.


Subject(s)
Biological Mimicry/physiology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/diagnosis , Mammary Neoplasms, Experimental/pathology , Neovascularization, Pathologic/diagnosis , Photoacoustic Techniques/methods , Tomography/methods , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Monitoring/methods , Female , Humans , MCF-7 Cells , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Neovascularization, Pathologic/pathology , Oxygen Consumption/physiology , Sensitivity and Specificity , Tumor Hypoxia/physiology , Xenograft Model Antitumor Assays
13.
Microvasc Res ; 116: 45-49, 2018 03.
Article in English | MEDLINE | ID: mdl-29069572

ABSTRACT

OBJECTIVE: To study blood flow, vascular volume and arterio-venous passages in induced mammary tumours of the rat to characterize parameters possibly responsible for tumour hyponutrition. METHOD: Dimethylbenzanthracene-induced mammary tumours in Sprague-Dawley rats were studied. Regional blood flow was studied by use of the radioactive microsphere tracer technique using 141Cerium-labelled 15µm spheres coinjected into the left cardiac ventricle with 125Iodine-labelled 25µm spheres. Blood volume was studied by use of 125Iodine- or 99mTechnetium-labelled human serum albumin, the latter allowing autoradiography of tumour sections for visualization of flow and volume. RESULTS: Twenty-seven rats with 170 tumours had a mean tumour blood flow of 48 and 67mL×min-1×100g-1 using 15 and 25µm sphere data, respectively, indicating a significant passage through vessels between 15 and 25µm. The lungs showed a "nominal bronchial" blood flow of 260 and 135mL×min-1×100g-1 for the 15 and 25µm spheres, respectively, indicating pulmonary trapping, particularly of small spheres passing the systemic circulation in vessels larger than 15µm. There was a positive correlation between the total tumour blood flow within individual rats and trapped spheres of both dimensions in the lungs, indicating shunts also larger than 25µm. Normal tissues disclosed only small differences in regional blood flow as measured by the two spheres. Blood volume was studied in 20 rats with 120 tumours, with a vascular volume of 3.6mL×100g-1 representing a blood turnover >15 times/min. Blood volume co-localized with perfusion as seen in autoradiographs. CONCLUSION: In induced rat mammary tumours, a high fraction of blood, 28%, passes arterio-venous vessels between 15 and 25µm and there also exist passages >25µm. These findings indicate that the functional capacity of the tumour vascular bed might be impaired, adding to the abnormal microenvironment of tumours.


Subject(s)
Arteries/physiopathology , Blood Volume , Mammary Neoplasms, Experimental/blood supply , Neovascularization, Pathologic , Veins/physiopathology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Arteries/diagnostic imaging , Autoradiography , Blood Flow Velocity , Blood Volume Determination/methods , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/physiopathology , Perfusion Imaging/methods , Pulmonary Circulation , Rats, Sprague-Dawley , Regional Blood Flow , Time Factors , Tumor Microenvironment , Veins/diagnostic imaging
14.
Int J Oncol ; 52(1): 103-126, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29115583

ABSTRACT

Low vitamin D status is considered as a risk factor for breast cancer and has prognostic significance. Furthermore, vitamin D deficiency increases after adjuvant cancer therapy, which alters bone metabolism increasing the risk of osteoporosis. It is now postulated that vitamin D supplementation in breast cancer treatment delays the recurrence of cancer thereby extending survival. We evaluated the impact of calcitriol and its low-calcemic analogs, PRI­2191 and PRI­2205, on the tumor growth, angiogenesis, and metastasis of 4T1 mouse mammary gland cancer. Gene expression analysis related to cancer invasion/metastasis, real­time PCR, ELISA, western blotting, and histochemical studies were performed. In vitro studies were conducted to compare the effects of calcitriol and its analogs on 4T1 and 67NR cell proliferation and expression of selected proteins. Calcitriol and its analogs increased lung metastasis without influencing the growth of primary tumor. The levels of plasma 17ß-estradiol and transforming growth factor ß (TGFß) were found to be elevated after treatment. Moreover, the results showed that tumor blood perfusion improved and osteopontin (OPN) levels increased, whereas vascular endothelial growth factor (VEGF) and TGFß levels decreased in tumors from treated mice. All the studied treatments resulted in increased collagen content in the tumor tissue in the early step of tumor progression, and calcitriol caused an increase in collagen content in lung tissue. In addition, in vitro proliferation of 4T1 tumor cells was not found to be affected by calcitriol or its analogs in contrast to non-metastatic 67NR cells. Calcitriol and its analogs enhanced the metastatic potential of 4T1 mouse mammary gland cancer by inducing the secretion of OPN probably via host cells. In addition, OPN tumor overexpression prevailed over the decreasing tumor TGFß level and blood vessel normalization via tumor VEGF deprivation induced by calcitriol and its analogs. Moreover, the increased plasma TGFß and 17ß-estradiol levels contributed to the facilitation of metastatic process.


Subject(s)
Calcitriol/analogs & derivatives , Calcitriol/pharmacology , Dihydroxycholecalciferols/pharmacology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/pathology , Animals , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Real-Time Polymerase Chain Reaction , Tumor Microenvironment/drug effects
15.
Asian Pac J Cancer Prev ; 18(12): 3231-3238, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29281877

ABSTRACT

Background: Angiogenic activity has been considered to reflect important molecular events during breast tumour development. The present study concerned cellular and molecular changes of MNU-induced breast tumours subjected to promotion and suppression of angiogenesis. Methods: Female Sprague Dawley rats at the age of 21 days received MNU at the dose 70 mg/kg of body weight by intraperitoneal injection. Three months post-carcinogen initiation, mammary tumours were palpated and their growth was monitored. When the tumour diameter reached 1.0 ± 0.05 cm, rats were given bFGF or PF4 intratumourally at a dose of 10 µg/tumour. Entire palpable tumour were subsequently excised and subjected to histology examination, IHC staining, and RT-PCR. Results: No critical morphological changes were observed between pro-angiogenic factor, bFGF, and control groups. However, increase of tumour size with more necrotic and diffuse areas was notable in tumours after anti-angiogenic PF4 intervention. ER and PR mRNA expression was significantly up- and down-regulated in bFGF and PF4 groups, respectively. The trends were significantly associated with peri- and intratumoural MVD counts. However, irrespective of whether we promoted or inhibited angiogenesis, the expression of EGFR and ERBB2 continued to be significantly increased but this was not significantly associated with the MVD score. No significant differences in E-cadherin and LR gene expression were noted between intervention and control groups. Conclusion: ER and PR receptor expression shows consistent responses when tumour angiogenesis is manipulated either positively or negatively. Our study adds to current understanding that not only do we need to target hormonal receptors, as presently practiced, but we also need to target endothelial receptors to successfully treat breast cancer.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Methylnitrosourea/toxicity , Neovascularization, Pathologic/prevention & control , Platelet Factor 4/administration & dosage , Alkylating Agents/toxicity , Animals , Female , Injections, Intralesional , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/chemically induced , Neovascularization, Pathologic/pathology , Rats , Rats, Sprague-Dawley
16.
Cell Rep ; 21(1): 274-288, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978480

ABSTRACT

The small GTPase RhoA is involved in a variety of fundamental processes in normal tissue. Spatiotemporal control of RhoA is thought to govern mechanosensing, growth, and motility of cells, while its deregulation is associated with disease development. Here, we describe the generation of a RhoA-fluorescence resonance energy transfer (FRET) biosensor mouse and its utility for monitoring real-time activity of RhoA in a variety of native tissues in vivo. We assess changes in RhoA activity during mechanosensing of osteocytes within the bone and during neutrophil migration. We also demonstrate spatiotemporal order of RhoA activity within crypt cells of the small intestine and during different stages of mammary gestation. Subsequently, we reveal co-option of RhoA activity in both invasive breast and pancreatic cancers, and we assess drug targeting in these disease settings, illustrating the potential for utilizing this mouse to study RhoA activity in vivo in real time.


Subject(s)
Biosensing Techniques , Fluorescence Resonance Energy Transfer/methods , Intravital Microscopy/methods , Time-Lapse Imaging/methods , rho GTP-Binding Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Movement/drug effects , Dasatinib/pharmacology , Erlotinib Hydrochloride/pharmacology , Female , Fluorescence Resonance Energy Transfer/instrumentation , Gene Expression Regulation , Intestine, Small/metabolism , Intestine, Small/ultrastructure , Intravital Microscopy/instrumentation , Mammary Glands, Animal/blood supply , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/ultrastructure , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/ultrastructure , Mechanotransduction, Cellular , Mice , Mice, Transgenic , Neutrophils/metabolism , Neutrophils/ultrastructure , Osteocytes/metabolism , Osteocytes/ultrastructure , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/ultrastructure , Time-Lapse Imaging/instrumentation , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
17.
Radiat Res ; 188(6): 605-614, 2017 12.
Article in English | MEDLINE | ID: mdl-28956695

ABSTRACT

Radiation stimulates the expression of inflammatory mediators known to increase cancer cell invasion. Therefore, it is important to determine whether anti-inflammatory drugs can prevent this adverse effect of radiation. Since cyclooxygenase-2 (COX-2) is a central player in the inflammatory response, we performed studies to determine whether the COX-2 inhibitor NS-398 can reduce the radiation enhancement of cancer cell invasion. Thighs of Balb/c mice treated with NS-398 were irradiated with either daily fractions of 7.5 Gy for five consecutive days or a single 30 Gy dose prior to subcutaneous injection of nonirradiated MC7-L1 mammary cancer cells. Five weeks later, tumor invasion, blood vessel permeability and interstitial volumes were assessed using magnetic resonance imaging (MRI). Matrix metalloproteinase-2 (MMP-2) was measured in tissues by zymography at 21 days postirradiation. Cancer cell invasion in the mouse thighs was increased by 12-fold after fractionated irradiations (5 × 7.5 Gy) and by 17-fold after a single 30 Gy dose of radiation. This stimulation of cancer cell invasion was accompanied by a significant increase in the interstitial volume and a higher level of the protease MMP-2. NS-398 treatment largely prevented the stimulation of cancer cell invasion, which was associated with a reduction in interstitial volume in the irradiated thighs and a complete suppression of MMP-2 stimulation. In conclusion, this animal model using MC7-L1 cells demonstrates that radiation-induced cancer cell invasion can be largely prevented with the COX-2 inhibitor NS-398.


Subject(s)
Gamma Rays/adverse effects , Mammary Neoplasms, Experimental/pathology , Neoplasm Invasiveness/prevention & control , Nitrobenzenes/therapeutic use , Sulfonamides/therapeutic use , Animals , Cell Line, Tumor , Cell Movement/radiation effects , Cyclooxygenase 2 Inhibitors/pharmacology , Drug Screening Assays, Antitumor , Inflammation Mediators/metabolism , Mammary Neoplasms, Experimental/blood supply , Matrix Metalloproteinase 2/analysis , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/prevention & control , Neoplasm Proteins/analysis , Neoplasm Transplantation , Nitrobenzenes/pharmacology , Radiotherapy/adverse effects , Sulfonamides/pharmacology , Thigh , Transplantation, Heterotopic , Tumor Burden/drug effects , Tumor Burden/radiation effects
18.
Nutr Cancer ; 69(7): 1036-1042, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28937793

ABSTRACT

BACKGROUND: One major concern in the treatment of cancer patients during chemotherapy is drug resistance. Here we investigated the effects of soy isoflavone extracts alone or in combination with Docetaxel on the drug resistance, angiogenesis, apoptosis, and tumor volume in mouse 4T1 breast tumor model. METHODS: Sixty female BALB/c mice were randomly divided into 4 groups: control, dietary soy isoflavone extract [Iso, 100 mg/kg diet (0.01%)], Docetaxel (10 mg/kg) injection, and the combination of dietary soy isoflavone extract and intravenous Docetaxel injection (Docetaxel + Iso). One week after the third injection, the breast tumors of eight mice from each group were excised to analyze NF-κBp65' vascular endothelial growth factor receptor-2 (VEGFR2) and Pgp gene and protein expressions and the other seven mice were monitored for survival rate analysis until they died. RESULTS: NF-κBp65 gene and protein expressions were significantly lower in the Docetaxel + Iso group in comparison with that of the Docetaxel group. VEGFR2 protein expression in the Docetaxel + Iso and Iso groups was significantly lower than that of the Docetaxel group. CONCLUSION: These findings may indicate that the combined use of isoflavone extracts together with chemotherapeutic agents has more efficient anti-carcinogenic effects than their individual use.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm/drug effects , Glycine max/chemistry , Mammary Neoplasms, Experimental/drug therapy , Plant Extracts/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Docetaxel , Eating/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Isoflavones/chemistry , Isoflavones/pharmacology , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Plant Extracts/administration & dosage , Plant Extracts/chemistry , Survival Rate , Taxoids/administration & dosage , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
19.
Adv Exp Med Biol ; 977: 169-174, 2017.
Article in English | MEDLINE | ID: mdl-28685442

ABSTRACT

We hypothesized that cancer cells actively migrate toward intratumor microvessels, guided by tissue gradients of metabolic substrates (such as O2) and/or metabolites (such as CO2/H+). To test this hypothesis, we developed an in vitro model in which cellular energy metabolism establishes gradients of O2/nutrient/metabolite in monolayer cells cultured in a conventional culture dish. When gradients of O2 ranging from 3% to ~0% were produced, MDA-MB-231 cells located at 300, 500 and 1500 µm downstream in the gradient demonstrated significant directional migrations (Rayleigh z test). We also found a similar directionality in cell migration at the same location even when the initial O2 level in the O2 gradient was raised from 3% to 21%. Interestingly, such directionalities were no longer demonstrated when the cell density was lowered from 1.8 × 106 to 0.9 × 106 cells/ml. In the former, the magnitude of the extracellular pH gradient in regions 300 and 500 µm downstream in the gradient was significantly larger. Thus, the direction of cell migrations appeared to depend on the gradient of extracellular pH rather than on O2.


Subject(s)
Chemotaxis , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Oxygen/metabolism , Cell Count , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Chemotaxis/drug effects , Energy Metabolism/physiology , Female , Humans , Hydrogen-Ion Concentration , Mammary Neoplasms, Experimental/blood supply , Microvessels/metabolism , Microvessels/pathology , Neoplasm Metastasis , Osmolar Concentration , Oxygen/pharmacology
20.
J Control Release ; 261: 216-222, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28576640

ABSTRACT

Over the last decade, the benefits of drug vectors to treat cancer have been well recognized. However, drug delivery and vector distribution differences in tumor-associated capillary bed at different stages of disease progression are not well understood. To obtain further insights into drug vector distribution changes in vasculature during tumor progression, we combined intra-vital imaging of metastatic tumors in mice, microfluidics-based artificial tumor capillary models, and Computational Fluid Dynamics (CFD) modeling. Microfluidic and CFD circulation models were designed to mimic tumor progression by escalating flow complexity and chaoticity. We examined flow of 0.5 and 2µm spherical particles, and tested the effects of hematocrit on particle local accessibility to flow area of capillary beds by co-circulating red blood cells (RBC). Results showed that tumor progression modulated drug vector distribution in tumor-associated capillaries. Both particles shared 80-90% common flow area, while 0.5 and 2µm particles had 2-9% and 1-2% specific flow area, respectively. Interestingly, the effects of hematocrit on specific circulation area was opposite for 0.5 and 2µm particles. Dysfunctional capillaries with no flow, a result of tumor progression, limited access to all particles, while diffusion was shown to be the only prevailing transport mechanism. In view of drug vector distribution in tumors, independent of formulation and other pharmacokinetic aspects, our results suggest that the evolution of tumor vasculature during progression may influence drug delivery efficiency. Therefore, optimized drug vectors will need to consider primary vs metastatic tumor setting, or early vs late stage metastatic disease, when undergoing vector design.


Subject(s)
Capillaries/metabolism , Drug Delivery Systems , Mammary Neoplasms, Experimental/pathology , Microfluidics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Disease Progression , Erythrocytes , Female , Hematocrit , Hydrodynamics , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Models, Theoretical , Neoplasm Metastasis , Neoplasm Staging
SELECTION OF CITATIONS
SEARCH DETAIL
...