Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
1.
Mol Oncol ; 16(2): 485-507, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34694686

RESUMO

Alteration in glycosylation pattern of MUC1 mucin tandem repeats during carcinomas has been shown to negatively affect adhesive properties of malignant cells and enhance tumor invasiveness and metastasis. In addition, MUC1 overexpression is closely interrelated with angiogenesis, making it a great target for immunotherapy. Alongside, easier interaction of nanobodies (single-domain antibodies) with their antigens, compared to conventional antibodies, is usually associated with superior desirable results. Herein, we evaluated the preclinical efficacy of a recombinant nanobody against MUC1 tandem repeats in suppressing tumor growth, angiogenesis, invasion, and metastasis. Expressed nanobody demonstrated specificity only toward MUC1-overexpressing cancer cells and could internalize in cancer cell lines. The IC50 values (the concentration at which the nanobody exerted half of its maximal inhibitory effect) of the anti-MUC1 nanobody against MUC1-positive human cancer cell lines ranged from 1.2 to 14.3 nm. Similar concentrations could also effectively induce apoptosis in MUC1-positive cancer cells but not in normal cells or MUC1-negative human cancer cells. Immunohistochemical staining of spontaneously developed mouse breast tumors prior to in vivo studies confirmed cross-reactivity of nanobody with mouse MUC1 despite large structural dissimilarities between mouse and human MUC1 tandem repeats. In vivo, a dose of 3 µg nanobody per gram of body weight in tumor-bearing mice could attenuate tumor progression and suppress excessive circulating levels of IL-1a, IL-2, IL-10, IL-12, and IL-17A pro-inflammatory cytokines. Also, a significant decline in expression of Ki-67, MMP9, and VEGFR2 biomarkers, as well as vasculogenesis, was evident in immunohistochemically stained tumor sections of anti-MUC1 nanobody-treated mice. In conclusion, the anti-MUC1 tandem repeat nanobody of the present study could effectively overcome tumor growth, invasion, and metastasis.


Assuntos
Proliferação de Células/genética , Neoplasias Mamárias Animais/patologia , Mucina-1/genética , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Neovascularização Patológica/genética , Anticorpos de Domínio Único/genética , Sequências de Repetição em Tandem , Animais , Apoptose/genética , Linhagem Celular Tumoral , Quimiocinas/metabolismo , Reações Cruzadas , Citocinas/metabolismo , Feminino , Humanos , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mucina-1/imunologia , Ligação Proteica , Anticorpos de Domínio Único/imunologia
2.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-33653008

RESUMO

Gold nanoparticles (AuNPs) are foreseen as a promising tool in nanomedicine, both as drug carriers and radiosensitizers. They have been also proposed as a potential anticancer drug due to the anti-angiogenic effect in tumor tissue. In this work we investigated the effect of citrate-coated AuNPs of nominal diameter 20 nm on the growth and metastatic potential of 4T1 cells originated from a mouse mammary gland tumor inoculated into the mammary fat pad of Balb/ccmdb mice. To evaluate whether AuNPs can prevent the tumor growth, one group of inoculated mice was intragastrically (i.g.) administered with 1 mg/kg of AuNPs daily from day 1 to day 14 after cancer cell implantation. To evaluate whether AuNPs can attenuate the tumor growth, the second group was intravenously (i.v.) administered with 1 or 5 mg/kg of AuNPs, twice on day 5 and day 14 after inoculation. We did not observe any anticancer activity of i.v. nor i.g. administered AuNPs, as they did not affect neither the primary tumor growth rate nor the number of lung metastases. Unexpectedly, both AuNP treatment regimens caused a marked vasodilating effect in the tumor tissue. As no change of potential angiogenic genes (Fgf2, Vegfa) nor inducible nitric oxygenase (Nos2) was observed, we proposed that the vasodilation was caused by AuNP-dependent decomposition of nitrosothiols and direct release of nitric oxide in the tumor tissue.


Assuntos
Ácido Cítrico/uso terapêutico , Ouro/uso terapêutico , Neoplasias Mamárias Animais/irrigação sanguínea , Nanopartículas Metálicas/uso terapêutico , Animais , Linhagem Celular Tumoral , Ácido Cítrico/administração & dosagem , Feminino , Ouro/administração & dosagem , Neoplasias Mamárias Animais/patologia , Neoplasias Mamárias Animais/terapia , Nanopartículas Metálicas/administração & dosagem , Camundongos , Camundongos Endogâmicos BALB C , Nanomedicina , Tamanho da Partícula , Vasodilatação
3.
Mol Oncol ; 14(12): 3198-3210, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33080107

RESUMO

Breast cancer metastasis is a complex process that depends not only on intrinsic characteristics of metastatic stem cells, but also on the particular microenvironment that supports their growth and modulates the plasticity of the system. In search for microenvironmental factors supporting cancer stem cell (CSC) growth and tumour progression to metastasis, we here investigated the role of the matricellular protein transforming growth factor beta induced (TGFBI) in breast cancer. We crossed the MMTV-PyMT model of mammary gland tumorigenesis with a TgfbiΔ/Δ mouse and studied the CSC content of the tumours. We performed RNAseq on wt and ko tumours, and analysed the tumour vasculature and the immune compartment by IHC and FACS. The source of TGFBI expression was determined by qPCR and by bone marrow transplantation experiments. Finally, we performed in silico analyses using the METABRIC cohort to assess the potential prognostic value of TGFBI. We observed that deletion of Tgfbi led to a dramatic decrease in CSC content and lung metastasis. Our results show that lack of TGFBI resulted in tumour vessel normalisation, with improved vessel perfusion and decreased hypoxia, a major factor controlling CSCs and metastasis. Furthermore, human data mining in a cohort of breast cancer patients showed that higher expression of TGFBI correlates with poor prognosis and is associated with the more aggressive subtypes of breast cancer. Overall, these data reveal a novel biological mechanism controlling metastasis that could potentially be exploited to improve the efficacy and delivery of chemotherapeutic agents in breast cancer.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Fator de Crescimento Transformador beta/metabolismo , Hipóxia Tumoral , Animais , Feminino , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neovascularização Patológica/metabolismo , Prognóstico
4.
Nanomedicine ; 21: 102074, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31376571

RESUMO

The assessment of vascular permeability of malignant tumor plays an important role in the diagnosis and treatment of cancer. Dynamic contrast-enhanced magnetic resonance image (DCE-MRI) using Gd-encapsulated carbonaceous dots and Gd-DTPA-BMA as contrast agents was performed in 4T1 mouse breast cancer and HCC827 human non-small-cell lung cancer (NSNLC) xenograft models. Histopathological parameters of tumor vascularity microvessel density (MVD), microvessel area (MVA), endothelial area (EA) and α-SMA CD31 Co-expression (α-SMA/CD31%) were compared with the DCE-MRI parameters. Results demonstrated that DCE-MRI with the new nanoparticle Gd@C-dots can noninvasively evaluate vascular permeability. Ktrans measured by DCE-MRI with Gd@C-dots is an accurate parameter for the characterization of tumor permeability. EA is a reliable microvessel parameter to evaluate vessel permeability.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Meios de Contraste , Endotélio Vascular/diagnóstico por imagem , Gadolínio , Neoplasias Pulmonares/diagnóstico por imagem , Imageamento por Ressonância Magnética , Neoplasias Mamárias Animais/diagnóstico por imagem , Nanopartículas/química , Neovascularização Patológica/diagnóstico por imagem , Animais , Carcinoma Pulmonar de Células não Pequenas/irrigação sanguínea , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Meios de Contraste/química , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Gadolínio/química , Gadolínio/farmacocinética , Gadolínio/farmacologia , Xenoenxertos , Humanos , Neoplasias Pulmonares/irrigação sanguínea , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Camundongos , Transplante de Neoplasias , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Transplante Isogênico
5.
Theranostics ; 9(13): 3918-3939, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281522

RESUMO

Carbon monoxide and nitric oxide are two of the most important vasoprotective mediators. Their downregulation observed during vascular dysfunction, which is associated with cancer progression, leads to uncontrolled platelet activation. Therefore, the aim of our studies was to improve vasoprotection and to decrease platelet activation during progression of mouse mammary gland cancer by concurrent use of CO and NO donors (CORM-A1 and DETA/NO, respectively). Methods: Mice injected intravenously with 4T1-luc2-tdTomato or orthotopically with 4T1 mouse mammary gland cancer cells were treated with CORM-A1 and DETA/NO. Ex vivo aggregation and activation of platelets were assessed in the blood of healthy donors and breast cancer patients. Moreover, we analyzed the compounds' direct effect on 4T1 mouse and MDA-MB-231 human breast cancer cells proliferation, adhesion and migration in vitro. Results: We have observed antimetastatic effect of combination therapy, which was only transient in orthotopic model. During early stages of tumor progression concurrent use of CORM-A1 and DETA/NO demonstrated vasoprotective ability (decreased endothelin-1, sICAM and sE-selectin plasma level) and downregulated platelets activation (decreased bound of fibrinogen and vWf to platelets) as well as inhibited EMT process. Combined treatment with CO and NO donors diminished adhesion and migration of breast cancer cells in vitro and inhibited aggregation as well as TGF-ß release from breast cancer patients' platelets ex vivo. However, antimetastatic effect was not observed at a later stage of tumor progression which was accompanied by increased platelets activation and endothelial dysfunction related to a decrease of VASP level. Conclusion: The therapy was shown to have antimetastatic action and resulted in normalization of endothelial metabolism, diminution of platelet activation and inhibition of EMT process. The effect was more prominent during early stages of tumor dissemination. Such treatment could be applied to inhibit metastasis during the first stages of this process.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Boranos/farmacologia , Carbonatos/farmacologia , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Óxido Nítrico/metabolismo , Compostos Nitrosos/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Boranos/uso terapêutico , Carbonatos/uso terapêutico , Bovinos , Adesão Celular/efeitos dos fármacos , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Progressão da Doença , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Endotélio/efeitos dos fármacos , Endotélio/fisiopatologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Hidrazinas/farmacologia , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos Endogâmicos BALB C , Proteínas dos Microfilamentos/metabolismo , Metástase Neoplásica , Óxido Nítrico/farmacologia , Compostos Nitrosos/uso terapêutico , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Fatores de Tempo
6.
Anticancer Agents Med Chem ; 19(5): 655-666, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30569877

RESUMO

BACKGROUND: The angiogenesis process is regulated by many factors, such as Hypoxia-Inducible Factor-1 (HIF-1) and Vascular Endothelial Growth Factor (VEGF). Metformin has demonstrated its ability to inhibit cell growth and the LY294002 is the major inhibitor of PI3K/AKT/mTOR pathway that has antiangiogenic properties. METHODS: Canine mammary tumor cell lines CMT-U229 and CF41 were treated with metformin and LY294002. Cell viability, protein and gene expression of VEGF and HIF-1 were determined in vitro. For the in vivo study, CF41 cells were inoculated in female athymic nude mice treated with either metformin or LY294002. The microvessel density by immunohistochemistry for CD31 as well as the gene and protein expression of HIF-1 and VEGF were evaluated. RESULTS: The treatment with metformin and LY294002 was able to reduce the cellular viability after 24 hours. The protein and gene expression of HIF-1 and VEGF decreased after treatment with metformin and LY294002. In the in vivo study, there was a decrease in tumor size, protein and gene expression of HIF-1 and VEGFA, in addition to the decreasing of CD31 expression after all treatments. CONCLUSION: Our results demonstrate the effectiveness of metformin and LY294002 in controlling the angiogenesis process in mammary tumors by VEGF and HIF-1, the most important angiogenic markers.


Assuntos
Cromonas/uso terapêutico , Doenças do Cão/tratamento farmacológico , Neoplasias Mamárias Animais/tratamento farmacológico , Metformina/uso terapêutico , Morfolinas/uso terapêutico , Neovascularização Patológica/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Cobalto/administração & dosagem , Cães , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
J Cell Mol Med ; 22(12): 5939-5954, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30291675

RESUMO

The activity of a cell-surface ecto-adenosine deaminase (eADA) is markedly increased in the endothelial activation and vascular inflammation leading to decreased adenosine concentration and alterations in adenosine signalling. Depending on the specific pathway activated, extracellular purines mediate host cell response or regulate growth and cytotoxicity on tumour cells. The aim of this study was to test the effects of adenosine deaminase inhibition by 2'deoxycoformycin (dCF) on the breast cancer development. dCF treatment decreased a tumour growth and a final tumour mass in female BALB/c mice injected orthotopically with 4T1 cancer cells. dCF also counteracted cancer-induced endothelial dysfunction in orthotopic and intravenous 4T1 mouse breast cancer models. In turn, this low dCF dose had a minor effect on immune stimulation exerted by 4T1 cell implantation. In vitro studies revealed that dCF suppressed migration and invasion of 4T1 cells via A2a and A3 adenosine receptor activation as well as 4T1 cell adhesion and transmigration through the endothelial cell layer via A2a receptor stimulation. Similar effects of dCF were observed in human breast cancer cells. Moreover, dCF improved a barrier function of endothelial cells decreasing its permeability. This study highlights beneficial effects of adenosine deaminase inhibition on breast cancer development. The inhibition of adenosine deaminase activity by dCF reduced tumour size that was closely related to the decreased aggressiveness of tumour cells by adenosine receptor-dependent mechanisms and endothelial protection.


Assuntos
Inibidores de Adenosina Desaminase/farmacologia , Progressão da Doença , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Receptores Purinérgicos P1/metabolismo , Adenosina Desaminase/metabolismo , Animais , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Espaço Extracelular/metabolismo , Feminino , Humanos , Neoplasias Mamárias Animais/sangue , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Nucleotídeos/sangue , Pentostatina/farmacologia , Fenótipo , Migração Transendotelial e Transepitelial/efeitos dos fármacos
8.
Dis Model Mech ; 11(9)2018 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-29997220

RESUMO

Cancer invasion programs are adaptive by switching between metastatic collective and single-cell dissemination; however, current intravital microscopy models for epithelial cancer in mice fail to reliably recreate such invasion plasticity. Using microimplantation of breast cancer spheroids into the murine mammary fat pad and live-cell monitoring, we show microenvironmental conditions and cytoskeletal adaptation during collective to single-cell transition in vivo E-cadherin-expressing 4T1 and E-cadherin-negative MMT tumors both initiated collective invasion along stromal structures, reflecting invasion patterns in 3D organotypic culture and human primary ductal and lobular carcinoma. Collectively invading cells developed weakly oscillatory actin dynamics, yet provided zones for single-cell transitions with accentuated, more chaotic actin fluctuations. This identifies collective invasion in vivo as a dynamic niche and efficient source for single-cell release.


Assuntos
Plasticidade Celular , Microscopia Intravital , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/patologia , Actinas/metabolismo , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Células HEK293 , Humanos , Imageamento Tridimensional , Neoplasias Mamárias Animais/irrigação sanguínea , Camundongos Endogâmicos BALB C , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica/patologia , Células Estromais/patologia
9.
Biomaterials ; 175: 110-122, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29803999

RESUMO

Nogo-B receptor (NgBR) plays fundamental roles in regulating angiogenesis, vascular development, and the epithelial-mesenchymal transition (EMT) of cancer cells. However, the therapeutic effect of NgBR blockade on tumor vasculature and malignancy is unknown, investigations on which requires an adequate delivery system for small interfering RNA against NgBR (NgBR siRNA). Here a surface charge switchable polymeric nanoparticle that was sensitive to the slightly acidic tumor microenvironment was developed for steady delivery of NgBR siRNA to tumor tissues. The nanoformulation was constructed by conjugating 2, 3-dimethylmaleic anhydride (DMMA) molecules to the surface amines of micelles formed by cationic co-polymer poly(lactic-co-glycolic acid)2-poly(ethylenimine) and subsequent absorption of NgBR siRNAs. The nanoparticles remained negatively charged in physiological condition and smartly converted to positive surface charge due to tumor-acidity-activated shedding of DMMA. The charge conversion facilitated cellular uptake of siRNAs and in turn efficiently depleted the expression of NgBR in tumor-bearing tissues. Silencing of NgBR suppressed endothelial cell migration and tubule formation, and reverted the EMT process of breast cancer cells. Delivery of the nanoformulation to mice bearing orthotopic breast carcinoma showed no effect on tumor growth, but led to remarkable decrease of distant metastasis by normalizing tumor vessels and suppressing the EMT of breast cancer cells. This study demonstrated that NgBR is a promising therapeutic target in abnormal tumor vasculature and aggressive cancer cells, and the tumor-responsive nanoparticle with the feature of charge transformation offers great potential for tumor-specific delivery of gene therapeutics.


Assuntos
Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/patologia , Nanopartículas/química , RNA Interferente Pequeno/administração & dosagem , Receptores de Superfície Celular/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular , Portadores de Fármacos , Liberação Controlada de Fármacos , Transição Epitelial-Mesenquimal , Feminino , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Anidridos Maleicos/química , Neoplasias Mamárias Animais/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica/terapia , Polietilenoimina/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptores de Superfície Celular/genética , Microambiente Tumoral
10.
Angiogenesis ; 21(3): 653-665, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29691683

RESUMO

Anti-angiogenesis therapy is an established therapeutic strategy for cancer. The endogenous angiogenic inhibitor angiostatin contains the first 3-4 kringle domains of plasminogen and inhibits both angiogenesis and vascular permeability. We present here a 10-residue peptide, Angio-3, derived from plasminogen kringle 3, which retains the functions of angiostatin in inhibiting both angiogenesis and vascular permeability. NMR studies indicate that Angio-3 holds a solution structure similar to the corresponding region of kringle 3. Mechanistically, Angio-3 inhibited both VEGF- and bFGF-induced angiogenesis by inhibiting EC proliferation and migration while inducing apoptosis. Inhibition of VEGF-induced vascular permeability results from its ability to impede VEGF-induced dissociation of adherens junction and tight junction proteins as well as the formation of actin stress fibers. When administered intravenously, Angio-3 inhibited subcutaneous breast cancer and melanoma growth by suppressing both tumor angiogenesis and intra-tumor vascular permeability. Hence, Angio-3 is a novel dual inhibitor of angiogenesis and vascular permeability. It is valuable as a lead peptide that can be further developed as therapeutics for diseases involving excessive angiogenesis and/or vascular permeability.


Assuntos
Permeabilidade Capilar , Células Endoteliais da Veia Umbilical Humana/patologia , Neoplasias Mamárias Animais , Melanoma Experimental , Neovascularização Patológica/metabolismo , Peptídeos/farmacologia , Plasminogênio/farmacologia , Animais , Apoptose/efeitos dos fármacos , Feminino , Fator 2 de Crescimento de Fibroblastos/antagonistas & inibidores , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Imageamento por Ressonância Magnética , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Melanoma Experimental/irrigação sanguínea , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Neovascularização Patológica/patologia , Peptídeos/síntese química , Peptídeos/química , Plasminogênio/química , Fibras de Estresse/metabolismo , Fibras de Estresse/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/metabolismo
11.
Breast Cancer Res ; 20(1): 20, 2018 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-29566737

RESUMO

BACKGROUND: Perfusion of breast cancer tissue limits oxygen availability and metabolism but angiogenesis inhibitors have hitherto been unsuccessful for breast cancer therapy. In order to identify abnormalities and possible therapeutic targets in mature cancer arteries, we here characterize the structure and function of cancer feed arteries and corresponding control arteries from female FVB/N mice with ErbB2-induced breast cancer. METHODS: We investigated the contractile function of breast cancer feed arteries and matched control arteries by isometric myography and evaluated membrane potentials and intracellular [Ca2+] using sharp electrodes and fluorescence microscopy, respectively. Arterial wall structure is assessed by transmission light microscopy of arteries mounted in wire myographs and by evaluation of histological sections using the unbiased stereological disector technique. We determined the expression of messenger RNA by reverse transcription and quantitative polymerase chain reaction and studied receptor expression by confocal microscopy of arteries labelled with the BODIPY-tagged α1-adrenoceptor antagonist prazosin. RESULTS: Breast cancer feed arteries are thin-walled and produce lower tension than control arteries of similar diameter in response to norepinephrine, thromboxane-analog U46619, endothelin-1, and depolarization with elevated [K+]. Fewer layers of similarly-sized vascular smooth muscle cells explain the reduced media thickness of breast cancer arteries. Evidenced by lower media stress, norepinephrine-induced and thromboxane-induced tension development of breast cancer arteries is reduced more than is explained by the thinner media. Conversely, media stress during stimulation with endothelin-1 and elevated [K+] is similar between breast cancer and control arteries. Correspondingly, vascular smooth muscle cell depolarizations and intracellular Ca2+ responses are attenuated in breast cancer feed arteries during norepinephrine but not during endothelin-1 stimulation. Protein expression of α1-adrenoceptors and messenger RNA levels for α1A-adrenoceptors are lower in breast cancer arteries than control arteries. Sympathetic vasocontraction elicited by electrical field stimulation is inhibited by α1-adrenoceptor blockade and reduced in breast cancer feed arteries compared to control arteries. CONCLUSION: Thinner media and lower α1-adrenoceptor expression weaken contractions of breast cancer feed arteries in response to sympathetic activity. We propose that abnormalities in breast cancer arteries can be exploited to modify tumor perfusion and thereby either starve cancer cells or facilitate drug and oxygen delivery during chemotherapy or radiotherapy.


Assuntos
Neoplasias da Mama/genética , Neoplasias Mamárias Animais/genética , Neovascularização Patológica/genética , Receptores Adrenérgicos alfa 1/genética , Antagonistas de Receptores Adrenérgicos alfa 1/administração & dosagem , Animais , Artérias/crescimento & desenvolvimento , Artérias/patologia , Artérias/ultraestrutura , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/patologia , Cálcio/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/patologia , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Contração Muscular/efeitos dos fármacos , Miografia , Neovascularização Patológica/patologia , Norepinefrina/administração & dosagem , Oxigênio/metabolismo , Prazosina/administração & dosagem , RNA Mensageiro/genética , Receptor ErbB-2/genética , Receptores Adrenérgicos alfa 1/administração & dosagem
12.
Sci Rep ; 8(1): 4171, 2018 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-29520098

RESUMO

Many cancers adeptly modulate metabolism to thrive in fluctuating oxygen conditions; however, current tools fail to image metabolic and vascular endpoints at spatial resolutions needed to visualize these adaptations in vivo. We demonstrate a high-resolution intravital microscopy technique to quantify glucose uptake, mitochondrial membrane potential (MMP), and SO2 to characterize the in vivo phentoypes of three distinct murine breast cancer lines. Tetramethyl rhodamine, ethyl ester (TMRE) was thoroughly validated to report on MMP in normal and tumor-bearing mice. Imaging MMP or glucose uptake together with vascular endpoints revealed that metastatic 4T1 tumors maintained increased glucose uptake across all SO2 ("Warburg effect"), and also showed increased MMP relative to normal tissue. Non-metastatic 67NR and 4T07 tumor lines both displayed increased MMP, but comparable glucose uptake, relative to normal tissue. The 4T1 peritumoral areas also showed a significant glycolytic shift relative to the tumor regions. During a hypoxic stress test, 4T1 tumors showed significant increases in MMP with corresponding significant drops in SO2, indicative of intensified mitochondrial metabolism. Conversely, 4T07 and 67NR tumors shifted toward glycolysis during hypoxia. Our findings underscore the importance of imaging metabolic endpoints within the context of a living microenvironment to gain insight into a tumor's adaptive behavior.


Assuntos
Microscopia Intravital/métodos , Neoplasias Mamárias Animais , Neovascularização Patológica , Imagem Óptica/métodos , Compostos Organometálicos/farmacologia , Tomografia Computadorizada por Raios X/métodos , Animais , Linhagem Celular Tumoral , Feminino , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/metabolismo , Camundongos , Camundongos Nus , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Microambiente Tumoral
13.
J Ultrasound Med ; 36(12): 2459-2466, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28646597

RESUMO

OBJECTIVES: This study aimed to evaluate the impact of long-term exercise training on the vascularization of rat mammary tumors. METHODS: Female rats were divided into 4 groups: N-methyl-N-nitrosourea (MNU) treated sedentary, MNU treated exercised, control sedentary, and control exercised. Tumor development was induced in the MNU groups by MNU administration. Exercised groups were trained for 35 weeks. Tumor vascularization was evaluated by pulsed Doppler and contrast-enhanced ultrasonography. RESULTS: The pulsatility and resistive indices were slightly higher in the MNU sedentary group (P > .05). Mammary tumors mainly had centripetal and heterogeneous enhancement of the contrast, clear margins, and the presence of penetrating vessels. The MNU exercised group had a lower arrival time and time to peak and higher peak intensity, wash-in, and wash-out (P > .05). The area under the curve was similar between groups (P > .05). CONCLUSIONS: The contrast-enhanced ultrasonographic study did not detect differences in mammary tumor vascularization between MNU sedentary and MNU exercised groups previously detected by power Doppler imaging, B-flow imaging, and immunohistochemistry.


Assuntos
Meios de Contraste , Aumento da Imagem/métodos , Neoplasias Mamárias Animais/irrigação sanguínea , Condicionamento Físico Animal , Ultrassonografia/métodos , Animais , Modelos Animais de Doenças , Feminino , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/patologia , Neovascularização Patológica/diagnóstico por imagem , Ratos , Ratos Sprague-Dawley , Tempo
14.
Life Sci ; 176: 35-41, 2017 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-28336398

RESUMO

AIMS: The inhibition of mast cells' degranulation may be an approach to prevent the formation of new vessels during the mammary carcinogenesis. MATERIALS AND METHODS: Female Sprague-Dawley rats were randomly divided into five experimental groups. Mammary tumors were induced by intraperitoneal injection of N-methyl-N-nitrosourea (MNU). Animals from group II were treated with ketotifen for 18weeks immediately after the MNU administration, while animals from group III only received the ketotifen after the development of the first mammary tumor. Mammary tumors vascularization was assessed by ultrasonography (Doppler, B Flow and contrast-enhanced ultrasound) and immunohistochemistry (vascular endothelial growth factor-A). KEY FINDINGS AND SIGNIFICANCE: Similar to what occurs in women with breast cancer, the majority of MNU-induced mammary tumors exhibited a centripetal enhancement order of the contrast agent, clear margin and heterogeneous enhancement. Ultrasonographic and immunohistochemical data suggest that the inhibition of mast cells' degranulation did not change the mammary tumors vascularization.


Assuntos
Neoplasias Mamárias Animais , Mastócitos/metabolismo , Metilnitrosoureia/toxicidade , Neovascularização Patológica , Ultrassonografia , Animais , Feminino , Cetotifeno/farmacologia , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/induzido quimicamente , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/metabolismo , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley
15.
Sci Adv ; 3(1): e1600675, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28138543

RESUMO

Understanding the role of the tumor microenvironment in carcinogenesis has reshaped cancer research. Events at both microscopic (molecular) and macroscopic (tissue) scales have been identified in engineered tumor microenvironments involving in vitro cultures, live tissue xenografts, and transgenic animals. However, these events have not been comprehensively observed under unperturbed (authentic) conditions free of exogenous labeling or genetic modification. The lack of a suitable imaging methodology has largely limited our understanding of the complex interrelations and possible causal links involved in carcinogenesis and metastasis within the tumor microenvironment. Using multicontrast nonlinear imaging, we visualize endogenous substances in rat and human mammary tumors through their intrinsic nonlinear optical properties, and simultaneously observe angiogenesis, extracellular matrix reorganization, and non-native cell recruitment. We find that all these macroscopic events in the tumor microenvironment require concurrent enrichment of specific extracellular vesicles and a metabolic switch toward biosynthesis. This concurrence at the microscopic scale provides not only new insights into carcinogenesis and metastasis but also a potentially new strategy for cancer diagnosis, surgery, and therapeutics.


Assuntos
Neoplasias da Mama , Matriz Extracelular , Neoplasias Mamárias Animais , Neovascularização Patológica , Microambiente Tumoral , Animais , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Feminino , Humanos , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/metabolismo , Neoplasias Mamárias Animais/patologia , Microscopia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Ratos , Ratos Endogâmicos F344 , Ratos Transgênicos
16.
Microvasc Res ; 108: 69-74, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27569845

RESUMO

PRIMARY OBJECTIVE: Utilizing the detailed vascular network obtained from micro-computed tomography (µCT) to establish a mathematical model of the temporal molecular distribution within a murine C3H mammary carcinoma. PROCEDURES: Female CDF1 mice with a C3H mammary carcinoma on the right rear foot were used in this study. Dynamic information for each tumour was achieved by Dynamic Contrast Enhanced-Magnetic Resonance Imaging (DCE-MRI) on a 16.4 T system. Detailed morphologic information on the tumour vasculature was obtained by ex vivo µCT and compared to CD34 immunohistochemical staining of tissue sections. The reconstructed vascular network served as origin for the diffusion (described by the apparent diffusion coefficient) within the tumour (the restricted volume described by the interstitial volume fraction derived from DCE-MRI). The resulting partial differential equation was solved using Finite-Element and a combined mathematical graph describing molecular distribution within the tumour was obtained. RESULTS: The established molecular distribution model predicted a heterogeneous distribution throughout the tumour related to the layout of the vascular network. Central tumour section concentration-time curves estimated from the established molecular distribution model were compared with physical measurements obtained by DCE-MRI of the same tumours and showed excellent correlation. CONCLUSIONS: A mathematical model describing temporal molecular distribution based on detailed vascular network structures was established and compared to DCE-MRI. The improved morphological insight will enhance future studies of heterogeneous tumours.


Assuntos
Antineoplásicos/metabolismo , Vasos Sanguíneos/diagnóstico por imagem , Angiografia por Tomografia Computadorizada , Simulação por Computador , Imageamento por Ressonância Magnética , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/diagnóstico por imagem , Modelos Biológicos , Técnicas de Diagnóstico Molecular/métodos , Microtomografia por Raio-X , Animais , Antígenos CD34/metabolismo , Antineoplásicos/administração & dosagem , Transporte Biológico , Vasos Sanguíneos/metabolismo , Difusão , Feminino , Análise de Elementos Finitos , Imuno-Histoquímica , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/metabolismo , Camundongos , Valor Preditivo dos Testes , Reprodutibilidade dos Testes
17.
Vet Immunol Immunopathol ; 178: 1-9, 2016 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-27496736

RESUMO

The activity of regulatory T cells (Tregs) is closely associated with the expression of FoxP3 transcription factor. FoxP3 regulatory T cells (FoxP3Treg) have immunosuppressive properties and can work for prevention of harmful autoimmune responses, however can also interfere with beneficial anti-tumor immunity. In human breast cancer these cells play a crucial role in tumor progression. In canine mammary tumors (CMT) this topic is not well-documented. This study included 80 malignant CMT and studied, by immunohistochemistry, the intratumoral FoxP3 expression together with microvessel density (MVD), vascular endothelial growth factor (VEGF) and several clinicopathological characteristics. Abundant FoxP3Treg cells were associated with tumor necrosis (p=0.001), high mitotic grade (p<0.001), more marked nuclear polymorphism (p=0.001), poor differentiation of tumors (p<0.001), high histological grade of malignancy (HGM) (p<0.001), presence of neoplastic intravascular emboli (p<0.001) and presence of lymph node metastasis (p<0.001). Intratumoral FoxP3 was correlated with MVD (r=0.827; p<0.001) and associated with VEGF (p=0.001). Additionally tumors with abundant FoxP3Treg cells were associated with shorter overall survival (OS) time in univariate and multivariate analysis (p<0.001 Kaplan-Meier curves and 7.97 hazard ratio, p<0.001 Cox proportional hazard model). Results suggest that Treg cells play a role in CMT progression and may contribute to increased angiogenesis and aggression in these tumors. The association of intratumoral FoxP3 expression with shorter OS in multivariate analysis suggests the usefulness of Treg cells as an independent prognostic marker.


Assuntos
Doenças do Cão/imunologia , Fatores de Transcrição Forkhead/imunologia , Neoplasias Mamárias Animais/imunologia , Animais , Biomarcadores Tumorais/imunologia , Biomarcadores Tumorais/metabolismo , Doenças do Cão/metabolismo , Doenças do Cão/patologia , Cães , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imuno-Histoquímica , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/patologia , Microvasos/patologia , Neovascularização Patológica , Prognóstico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Cell Rep ; 15(6): 1161-74, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27134168

RESUMO

Despite the approval of several anti-angiogenic therapies, clinical results remain unsatisfactory, and transient benefits are followed by rapid tumor recurrence. Here, we demonstrate potent anti-angiogenic efficacy of the multi-kinase inhibitors nintedanib and sunitinib in a mouse model of breast cancer. However, after an initial regression, tumors resume growth in the absence of active tumor angiogenesis. Gene expression profiling of tumor cells reveals metabolic reprogramming toward anaerobic glycolysis. Indeed, combinatorial treatment with a glycolysis inhibitor (3PO) efficiently inhibits tumor growth. Moreover, tumors establish metabolic symbiosis, illustrated by the differential expression of MCT1 and MCT4, monocarboxylate transporters active in lactate exchange in glycolytic tumors. Accordingly, genetic ablation of MCT4 expression overcomes adaptive resistance against anti-angiogenic therapy. Hence, targeting metabolic symbiosis may be an attractive avenue to avoid resistance development to anti-angiogenic therapy in patients.


Assuntos
Inibidores da Angiogênese/farmacologia , Resistencia a Medicamentos Antineoplásicos , Neoplasias Mamárias Animais/metabolismo , Neovascularização Patológica/tratamento farmacológico , Inibidores da Angiogênese/uso terapêutico , Animais , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Glicólise/efeitos dos fármacos , Humanos , Indóis/farmacologia , Indóis/uso terapêutico , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/tratamento farmacológico , Neoplasias Mamárias Animais/patologia , Camundongos , Modelos Biológicos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Neovascularização Patológica/patologia
19.
Oncotarget ; 7(14): 18106-15, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26919112

RESUMO

Interleukin-33 (IL-33)/IL-33 receptor (IL-33R, ST2) signaling pathway promotes mammary cancer growth and metastasis by inhibiting anti-tumor immunity. However, the role of IL-33/IL-33R axis in neoangiogenesis and tumor necrosis is not elucidated. Therefore, the aim of this study was to investigate the role of IL-33/IL-33R axis in mammary tumor necrosis. Deletion of IL-33R (ST2) gene in BALB/c mice enhanced tumor necrosis and attenuated tumor growth in 4T1 breast cancer model, which was associated with markedly decreased expression of vascular endothelial growth factor (VEGF) and IL-33 in mammary tumor cells. We next analyzed IL-33, IL-33R and VEGF expression and microvascular density (MVD) in breast tumors from 40 female patients with absent or present tumor necrosis. We found significantly higher expression of IL-33, IL-33R and VEGF in breast cancer tissues with absent tumor necrosis. Both, IL-33 and IL-33R expression correlated with VEGF expression in tumor cells. Further, VEGF expression positively correlated with MVD in perinecrotic zone. Taking together, our data indicate that IL-33/IL-33R pathway is critically involved in mammary tumor growth by facilitating expression of pro-angiogenic VEGF in tumor cells and attenuating tumor necrosis. These data add an unidentified mechanism by which IL-33/IL-33R axis facilitates tumor growth.


Assuntos
Neoplasias da Mama/patologia , Interleucina-33/metabolismo , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/patologia , Necrose/patologia , Receptores de Interleucina/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Proteína 1 Semelhante a Receptor de Interleucina-1 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microvasos/patologia , Pessoa de Meia-Idade , Necrose/genética , Metástase Neoplásica/genética , Metástase Neoplásica/patologia , Neovascularização Patológica/patologia , Receptores de Interleucina/metabolismo
20.
Mol Imaging Biol ; 18(4): 617-26, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26630973

RESUMO

PURPOSE: Transgenic mice expressing the polyoma middle T oncoprotein (PyMT) in the mammary epithelium were explored by multimodal imaging to monitor longitudinally spontaneous tumor growth and response to chemotherapy. PROCEDURES: Positron emission tomography (PET) with 2-deoxy-2-[(18)F]fluoro-D-glucose ([(18)F]FDG) and 3'-deoxy-3'-[(18)F]fluorothymidine ([(18)F]FLT), single photon emission tomography (SPECT) with [(99m)Tc]TcO4 ([(99m)Tc]TEC), X-ray computed tomography, and fluorescent confocal endomicroscopy (FCE) images were acquired during tumor progression in female PyMT mice. Imaging with [(18)F]FDG and [(99m)Tc]TEC was also performed in untreated, doxorubicin-treated, and docetaxel-treated PyMT mice. Total tumor volumes were quantified. Tumors were collected and macroscopic and histological examinations were performed. RESULTS: All PyMT mice developed multifocal tumors of the mammary epithelium that became palpable at 8 weeks of age (W8). Computed tomography (CT) detected tumors at W14, while a clear tumoral uptake of [(99m)Tc]TEC and [(18)F]FDG was present as early as W6 and W8, respectively. No contrast between mammary tumors and surrounding tissue was observed at any stage with [(18)F]FLT. FCE detected an angiogenic switch at W10. Lung metastases were not clearly evidenced by imaging. Doxorubicin and docetaxel treatments delayed tumor growth, as shown by [(18)F]FDG and [(99m)Tc]TEC, but tumor growth resumed upon treatment discontinuation. Tumor growth fitted an exponential model with time constant rates of 0.315, 0.145, and 0.212 week(-1) in untreated, doxorubicin, and docetaxel groups, respectively. CONCLUSIONS: Molecular imaging of mammary tumors in PyMT is precocious, precise, and predictive. [(18)F]FDG-PET and [(99m)Tc]TEC SPECT monitor tumor response to chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Carcinogênese/patologia , Neoplasias Mamárias Animais/tratamento farmacológico , Imagem Multimodal/métodos , Animais , Antineoplásicos/farmacologia , Proliferação de Células , Modelos Animais de Doenças , Progressão da Doença , Feminino , Fluorescência , Fluordesoxiglucose F18 , Antígeno Ki-67/metabolismo , Neoplasias Mamárias Animais/irrigação sanguínea , Neoplasias Mamárias Animais/diagnóstico por imagem , Neoplasias Mamárias Animais/patologia , Camundongos , Camundongos Transgênicos , Tecnécio/química , Fatores de Tempo , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...