Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters











Publication year range
1.
J Dairy Sci ; 103(6): 5532-5549, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32229120

ABSTRACT

A previous study in the bovine mammary epithelial cell line BME-UV1 demonstrated that suppression of the phosphatidylinositol-4,5-biphosphate 3 kinase (PI3K)/AKT (somatotropic) signaling pathway was required for transforming growth factor ß1 (TGFß1)-induced programmed cell death (PCD). To investigate whether this is a universal mechanism for TGFß1 to induce PCD in bovine mammary epithelium, we compared TGFß1 modulation of PI3K/AKT and its role in PCD in 2 bovine mammary epithelial cell lines: MAC-T and BME-UV1. In MAC-T cells, TGFß1 promoted cell survival, and this paralleled a reduction in PI3K/AKT activity, rather than an increase. In BME-UV1 cells, TGFß1 induced PCD, and this was accompanied by a time-dependent effect on PI3K/AKT activity, including an initial significant increase in the phosphorylation of AKT at 3 h, followed by a reduction between 12 and 24 h, and then an increase at 48 h. Inhibition of AKT activity enhanced TGFß1-induced PCD in BME-UV1 cells but had no effect on MAC-T cells, suggesting that TGFß1 mediates PCD in BME-UV1 cells through suppression of AKT activity. Inhibition of TGFß receptor type I (TßRI) kinase activity completely abrogated TGFß1-induced PCD in BME-UV1 cells but had no effect on TGFß1-induced suppression of PCD in MAC-T cells, demonstrating that TGFß1-induced PCD in BME-UV1 cells is dependent on TßRI/SMAD signaling. These and previous observations suggest that the different effects of TGFß1 on PCD in these cell lines might involve noncanonical signaling pathways other than PI3K/AKT, and may reflect their different lineages. Future studies should address this finding, taking into consideration the effect that different culture conditions might have on cell phenotype.


Subject(s)
Apoptosis , Cattle/physiology , Signal Transduction , Transforming Growth Factor beta1/metabolism , Animals , Cell Line , Cell Survival , Epithelial Cells/metabolism , Female , Mammary Glands, Animal/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation
2.
Vet Comp Oncol ; 15(4): 1572-1584, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28322030

ABSTRACT

BACKGROUND: Melatonin has oncostatic actions and IL-25 is active in inflammatory processes that induce apoptosis in tumor cells AIM: The aim of this study was to evaluate melatonin and IL-25 in metastatic (CF-41) and non-metastatic (CMT-U229) canine mammary tumor cells cultured as monolayers and tridimensional structures. MATERIALS AND METHODS: The cells were treated with melatonin, IL-25 and IL-17B silencing gene and performed cell viability, gene and protein expression of caspase-3 and VEGFA (Vascular endothelial growth factor A) and an apoptosis membrane protein array. RESULTS: Treatment with 1 mM of melatonin reduced cell viability of both tumor cell lines, all treatments alone and combined significantly increased caspase-3 cleaved and proteins involved in the apoptotic pathway and reduced pro-angiogenic VEGFA, confirming the effectiveness of these potential promising treatments. CONCLUSION: This is the first study evaluating the potential use of these strategies in CF-41 and CMT-U229 cell lines and together encourages subsequent in vitro and in vivo studies for further exploration of clinical applications.


Subject(s)
Apoptosis/drug effects , Dog Diseases/drug therapy , Interleukin-17/pharmacology , Mammary Neoplasms, Animal/drug therapy , Melatonin/pharmacology , Animals , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dog Diseases/pathology , Dogs , Female , Fluorescent Antibody Technique/veterinary , Gene Silencing , Mammary Neoplasms, Animal/pathology , Vascular Endothelial Growth Factor A/metabolism
3.
BMC Cell Biol ; 18(1): 10, 2017 01 23.
Article in English | MEDLINE | ID: mdl-28114883

ABSTRACT

BACKGROUND: Sprouting angiogenesis requires vascular endothelial proliferation, migration and morphogenesis. The process is regulated by soluble factors, principally vascular endothelial growth factor (VEGF), and via bidirectional signaling through the Jagged/Notch system, leading to assignment of tip cell and stalk cell identity. The cytokine transforming growth factor beta (TGF-ß) can either stimulate or inhibit angiogenesis via its differential surface receptor signaling. Here we evaluate changes in expression of angiogenic signaling receptors when bovine aortic endothelial cells were exposed to TGF-ß1 under low serum conditions. RESULTS: TGF-ß1 induced a dose dependent inhibition of tip cell assignment and subsequent angiogenesis on Matrigel, maximal at 5.0 ng/ml. This occurred via ALK5-dependent pathways and was accompanied by significant upregulation of the TGF-ß co-receptor endoglin, and SMAD2 phosphorylation, but no alteration in Smad1/5 activation. TGF-ß1 also induced ALK5-dependent downregulation of Notch1 but not of its ligand delta-like ligand 4. Cell associated VEGFR2 (but not VEGFR1) was significantly downregulated and accompanied by reciprocal upregulation of VEGFR2 in conditioned medium. Quantitative polymerase chain reaction analysis revealed that this soluble VEGFR2 was not generated by a selective shift in mRNA isoform transcription. This VEGFR2 in conditioned medium was full-length protein and was associated with increased soluble HSP-90, consistent with a possible shedding of microvesicles/exosomes. CONCLUSIONS: Taken together, our results suggest that endothelial cells exposed to TGF-ß1 lose both tip and stalk cell identity, possibly mediated by loss of VEGFR2 signaling. The role of these events in physiological and pathological angiogenesis requires further investigation.


Subject(s)
Endothelial Cells/metabolism , Neovascularization, Physiologic , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cattle , Endothelial Cells/drug effects , Humans , Neovascularization, Physiologic/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology
4.
Genet Mol Res ; 10(4): 3901-13, 2011 Dec 08.
Article in English | MEDLINE | ID: mdl-22183949

ABSTRACT

Breast cancer is one of the leading causes of cancer-related deaths amongst women in the USA. The tumor microenvironment has been suggested to be an attractive therapeutic target for treatment of cancers. The glycosaminoglycan chondroitin sulfate, as part of the cellular microenvironment, consists of long linear chains of repeating disaccharide units, which are covalently attached to core proteins to form chondroitin sulfate-proteoglycans. In vitro studies have implicated chondroitin sulfate in various aspects of carcinogenesis, whereas the in vivo roles of chondroitin sulfate are less clear. Drastically elevated levels of chondroitin sulfate have been observed within the stromal compartment of many solid tumors, including human breast carcinomas, the significance of which is unknown. We examined the role of tumor-associated chondroitin sulfate in breast cancer progression. Enzymatic elimination of endogenous chondroitin sulfate by intra-tumor injections of chondroitinase ABC leads to the development of secondary tumors and increased lung metastases, while primary orthotopic tumor growth was not affected. These results establish a metastasis-inhibiting effect of primary breast tumor-associated chondroitin sulfate, which may open novel carbohydrate-based therapeutic strategies to combat breast cancer.


Subject(s)
Chondroitin Sulfates/metabolism , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Chondroitin ABC Lyase/administration & dosage , Chondroitin ABC Lyase/pharmacology , Female , Injections , Mice
5.
Cancer Res ; 61(13): 5090-101, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11431346

ABSTRACT

Inhibitors of epidermal growth factor receptor (EGFR) signaling are among the novel drugs showing great promise for cancer treatment in the clinic. However, the possibility of acquired resistance to such drugs because of tumor cell genetic instabilities has not yet been explored. Here we report the experimental derivation and properties of such cell variants obtained from recurrent tumor xenografts of the human A431 squamous cell carcinoma, after two consecutive cycles of therapy with one of three different anti-EGFR monoclonal antibodies: mR3, hR3, or C225. Initial response to a 2-week period of treatment was generally total tumor regression and was not significantly different among the three antibody groups. However, tumors often reappeared at the site of inoculation, generally after prolonged latency periods, and most of the tumors became refractory to a second round of therapy. Cell lines established from such resistant tumors retained high EGFR expression, normal sensitivity to anti-EGFR antibody or ligand, and unaltered growth rate when compared with the parental line in vitro. In contrast, the A431 cell variants exhibited an accelerated growth rate and a significantly attenuated response to anti-EGFR antibodies in vivo relative to the parental line. Because of the reported suppressive effect of EGFR inhibitors on vascular endothelial growth factor (VEGF) expression, and the demonstrated role of VEGF in the angiogenesis and growth of A431 tumor xenografts, relative VEGF expression was examined. Five of six resistant variants expressed increased levels of VEGF, which paralleled an increase in both angiogenic potential in vitro and tumor angiogenesis in vivo. In addition, elevated expression of VEGF in variants of A431 cells obtained by gene transfection rendered the cells significantly resistant to anti-EGFR antibodies in vivo. Taken together, the results suggest that, at least in the A431 system, variants displaying acquired resistance to anti-EGFR antibodies can emerge in vivo and can do so, at least in part, by mechanisms involving the selection of tumor cell subpopulations with increased angiogenic potential.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/blood supply , ErbB Receptors/antagonists & inhibitors , Neovascularization, Pathologic/pathology , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Drug Resistance, Neoplasm , Endothelial Growth Factors/biosynthesis , Endothelial Growth Factors/genetics , Endothelial Growth Factors/physiology , ErbB Receptors/immunology , Humans , Lymphokines/biosynthesis , Lymphokines/genetics , Lymphokines/physiology , Mice , Mice, SCID , Neoplasm Recurrence, Local , Neovascularization, Pathologic/metabolism , Tumor Cells, Cultured , Up-Regulation , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Xenograft Model Antitumor Assays
6.
Cancer Metastasis Rev ; 20(1-2): 79-86, 2001.
Article in English | MEDLINE | ID: mdl-11831651

ABSTRACT

The ultimate target of anti-angiogenic drugs is the genetically stable, activated endothelial cell of a newly forming tumor blood vessel, rather than the genetically unstable tumor cell population per se. This led to the notion that acquired resistance to such drugs may not develop as readily, if at all. While there is some evidence that this lack of resistance development may be the case for some direct-acting angiogenesis inhibitors, it is becoming apparent that resistance can develop over time to many types of angiogenesis inhibitors including, possibly, some direct inhibitors, especially when used as monotherapies. Possible mechanisms for such acquired or induced resistance include: (i) redundancy of pro-angiogenic growth factors when the drug used targets a single such growth factor or its cognate endothelial cell-associated receptor tyrosine kinase; (ii) the anti-apoptotic/pro-survival function of growth factors such as VEGF, which, in high local concentrations, can antagonize the pro-apoptotic effects of various angiogenesis inhibitors; (iii) epigenetic, transient upregulation, or induction, of various anti-apoptotic effector molecules in host-endothelial cells; and (iv) heterogeneous vascular dependence of tumor cell populations. It is suggested that long-term disease control with anti-angiogenic drugs can be best achieved by judicious combination therapy. In this regard, the great molecular diversity of anti-angiogenic drug targets, in contrast to chemotherapy, makes this a particularly attractive therapeutic option, especially when approved, commercially available drugs considered to have anti-angiogenic effects are used in such combination treatment strategies.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Drug Resistance, Neoplasm , Animals , Drug Therapy, Combination , Humans , Neoplasms/blood supply , Neoplasms/drug therapy
7.
Oncogene ; 19(40): 4611-20, 2000 Sep 21.
Article in English | MEDLINE | ID: mdl-11030150

ABSTRACT

Like other types of pre-malignant lesions and carcinoma, angiogenesis is associated with high-grade cervical dysplasia and with invasive squamous carcinoma of the cervix. Vascular endothelial cell growth factor (VEGF) is known to be one of the most important inducers of angiogenesis and is upregulated in carcinoma of the cervix. Human Papilloma Virus 16 (HPV-16) has been etiologically linked to human cervical cancer, and the major oncogenic proteins encoded by the viral genome, E6 and E7, are involved in the immortalization of target cells. Because several oncogenes including mutant ras, EGF receptor, ErbB2/Her2, c-myc and v-src upregulate VEGF expression, we asked whether HVP-16 E6 oncoprotein could act in a similar fashion. We found that HPV-16 E6-positive cells generally express high levels of VEGF message. Furthermore, co-expression of the VEGF promoter-Luc (luciferase) reporter gene with E6 in both human keratinocytes and mouse fibroblast showed that E6 oncoprotein upregulates VEGF promoter activity, and does so in a p53 independent manner. An E6 responsive region which comprises four Sp-1 sites, between -194 and -50 bp of the VEGF promoter, is also necessary for constitutive VEGF transcription. Taken together, our results suggest the possibility that the HPV oncoprotein E6 may contribute to tumor angiogenesis by direct stimulation of the VEGF gene.


Subject(s)
Endothelial Growth Factors/genetics , Genes, p53 , Lymphokines/genetics , Neovascularization, Pathologic/genetics , Oncogene Proteins, Viral/physiology , Papillomaviridae/genetics , Promoter Regions, Genetic , Repressor Proteins , Tumor Suppressor Protein p53/physiology , Autocrine Communication , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Endothelial Growth Factors/metabolism , ErbB Receptors/physiology , Female , HeLa Cells/metabolism , HeLa Cells/virology , Humans , Keratinocytes/virology , Lymphokines/metabolism , Neoplasm Proteins/physiology , Neovascularization, Pathologic/physiopathology , Oncogene Proteins, Viral/genetics , Papillomaviridae/physiology , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Recombinant Fusion Proteins/biosynthesis , Transcription, Genetic , Transcriptional Activation , Transforming Growth Factor alpha/physiology , Tumor Cells, Cultured/metabolism , Tumor Cells, Cultured/virology , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vulvar Neoplasms/metabolism , Vulvar Neoplasms/pathology , Vulvar Neoplasms/virology
8.
Eur J Cancer ; 36(10): 1248-57, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10882863

ABSTRACT

A number of drugs currently being tested in clinical trials as possible angiogenesis inhibitors were not originally developed with the intention of suppressing tumour angiogenesis. Thalidomide and interferon alpha are obvious examples of such drugs. This list of 'accidental' angiogenesis inhibitors may include established agents such as conventional cytotoxic chemotherapeutic drugs as well as the new generation of anticancer drugs known as anti-oncoprotein signal transduction inhibitors. With respect to the former, the potential of such drugs to inhibit angiogenesis could be the result of their ability to cause collateral damaging effects on cycling endothelial cells found in newly formed blood vessels, or inhibiting other vital endothelial cell functions necessary for angiogenesis. The antitumour vascular side-effects of chemotherapy may be optimised by administering such drugs continuously on a more frequent (e.g. weekly or even daily) basis at levels well below the maximum tolerated dose (MTD), especially when this is done in combination with newly developed anti-angiogenic drugs such as vascular endothelial cell growth factor (VEGF) receptor blocking antibodies. This strategy may minimise or delay the problems of host toxicity and acquired drug resistance. The possibility of anti-angiogenic effects mediated by signal transduction inhibitors such as ras farnesyltransferase inhibitors (ras FTI's), or drugs which block receptor tyrosine kinases (e.g. ErbB2/neu) such as Herceptin, may be the consequence of such oncogenes inducing or upregulating various pro-angiogenic molecules such as VEGF (vascular endothelial cell growth factor) in tumour cells. Hence, treatment of tumour cells with such drugs can lead to downregulation of tumour cell-associated VEGF expression and this can contribute to an anti-angiogenic effect of the drug in vivo. In addition, some of these drugs may also affect certain 'activated' endothelial cell functions directly so as to block angiogenesis. An awareness of the potential of such conventional or experimental anticancer drugs to affect tumour growth through blockade or suppression of angiogenesis has implications for how anticancer drugs may be used clinically, either alone, or in combination with other drugs to optimally treat cancer.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Neoplasms/drug therapy , Neuroblastoma/drug therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Drug Therapy, Combination , Endothelial Growth Factors/metabolism , Humans , Lymphokines/metabolism , Mice , Mice, SCID , Neovascularization, Pathologic , Signal Transduction , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors , Vinblastine/antagonists & inhibitors , Vinblastine/therapeutic use
9.
Cancer Res ; 60(2): 490-8, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10667605

ABSTRACT

A possible link between oncogenes and tumor angiogenesis has been implicated by the finding that expression of various oncogenes, particularly mutant ras, can lead to a marked induction of a potent paracrine stimulator of angiogenesis, vascular endothelial growth factor (VEGF). We sought to determine how oncogenic ras induction of VEGF is mediated at the molecular level and whether the mechanisms involved differ fundamentally between transformed epithelial cells and fibroblasts. Our results suggest that in a subline (called RAS-3) of immortalized nontumorigenic rat intestinal epithelial cells (IEC-18) that acquired a tumorigenic phenotype upon transfection of mutant ras, up-regulation of VEGF occurs in the absence of an autocrine growth factor circuit. The expression of VEGF mRNA and protein by RAS-3 cells was strongly suppressed in the presence of LY294002, an inhibitor of phosphatidylinositol 3'-kinase, but remained largely unaffected in the same cells treated with an inhibitor (PD98059) of mitogen-activated protein/extracellular signal-regulated kinase kinase 1 (MKK/MEK-1). This is consistent with the observation that overexpression of a constitutively activated mutant of MEK-1 (AN3/ S222D) in the parental IEC-18 cells did not result in up-regulation of VEGF production. The impact of mutant ras on VEGF expression was also significantly amplified at high cell density, conditions under which RAS-3 cells became less sensitive to LY294002-induced VEGF down-regulation. In marked contrast to cells of epithelial origin, ras-transformed murine fibroblasts (3T3RAS) up-regulated VEGF in a manner that was strongly inhibitable by MEK-1 blockade (ie. treatment with PD98059), whereas these cells were relatively unaffected by treatment with the phosphatidylinositol 3'-kinase inhibitor LY294002. In addition, VEGF was up-regulated by 2-3-fold in NIH3T3 cells overexpressing mutant MEK-1. Collectively, the data suggest that the stimulatory effect of mutant ras on VEGF expression is executed in a nonautocrine and cell type-dependent manner and that it can be significantly exacerbated by physiological/ environmental influences such as high cell density.


Subject(s)
Cell Transformation, Neoplastic , Endothelial Growth Factors/genetics , Gene Expression Regulation , Genes, ras , Intestinal Mucosa/physiology , Lymphokines/genetics , Neoplasms, Experimental/genetics , Neovascularization, Pathologic , 3T3 Cells , Animals , Cell Division , Cell Line , Chromones/pharmacology , Enzyme Inhibitors/pharmacology , Epithelial Cells/pathology , Epithelial Cells/physiology , Fibroblasts/pathology , Fibroblasts/physiology , Flavonoids/pharmacology , Intestinal Mucosa/pathology , Kinetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Morpholines/pharmacology , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombospondin 1/genetics , Transfection , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factors
10.
Mol Med ; 4(5): 286-95, 1998 May.
Article in English | MEDLINE | ID: mdl-9642680

ABSTRACT

We have tried to stress that mutant oncogenes or overexpressed, nonmutated proto-oncogenes, in addition to their direct affect on promoting aberrant tumor cell proliferation (and survival), may possess a crucial indirect means of stimulating tumor cell growth through regulation of angiogenesis. This effect would never be observed in tissue culture studies of oncogene function using pure cultures of tumor cells, which probably helps explain why the pro-angiogenic function of oncogenes has not been appreciated until only relatively recently. Indeed, the very first indication of a possible contributory role of oncogenes, such as ras and myc, to tumor angiogenesis was first reported by Thompson et al. in 1989, who used reconstituted organ cultures of the mouse prostate gland for their studies (69). This potentially important contribution of oncogenes to tumor growth and development may prove to have an impact on how various signal transduction inhibitors that are now in early phase clinical trials, e.g., monoclonal neutralizing antibodies to the human EGF receptor (70), function in vivo as anti-tumor agents.


Subject(s)
Neoplasms/blood supply , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Oncogenes , Animals , Genes, Tumor Suppressor , Humans , Neovascularization, Pathologic/prevention & control , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL