Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Exp Mol Pathol ; 125: 104751, 2022 04.
Article in English | MEDLINE | ID: mdl-35122807

ABSTRACT

We have identified the novel protein GASP-1 (G protein coupled receptor-associated sorting protein 1) that appears to be a universal cancer marker and the expression of which in tumor tissue and patient sera is predictive of cancer severity (Tuszynski et al. 2011; Zheng et al. 2012; Zheng 2013; Chang and Tuszynski, 2020). In preliminary results we discovered that a GASP-1 antibody inhibited the growth of the triple negative breast cancer cell line MDA-MB-231 and transient reduction of GASP-1 in these cells decreased their proliferation. To further substantiate these results, we over and under-expressed GASP-1 in stable clones of MDA-MB-231 cells and evaluated their growth and invasive activities. Cells under-expressing GASP-1 failed to grow after 4 days in culture and eventually died. In contrast GASP-1 expressing cells grew exponentially. Similarly, GASP-1 under-expressing cells formed 30% fewer colonies in soft agar as compared to controls and whereas GASP-1 over-expressing cells formed 2-fold more colonies than controls. In tumor cell invasion assays GASP-1 over-expressing cells were over 10-fold more invasive than controls whereas GASP-1 under-expressing cells were over 10-fold less invasive than controls. In IHC staining studies of breast cancer cells, we found that the overexpressed GASP-1 appear in granules of different sizes that are directly correlated with cancer invasiveness. Our results strongly indicate that GASP-1 promotes proliferation and invasion of the triple negative breast cancer cell line MDA-MB-231 and targeting GASP-1 for treatment of breast cancer is indicated.


Subject(s)
Intercellular Signaling Peptides and Proteins , Triple Negative Breast Neoplasms , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Intercellular Signaling Peptides and Proteins/genetics , Neoplasm Invasiveness , Triple Negative Breast Neoplasms/pathology
2.
Exp Mol Pathol ; 95(3): 249-54, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24036285

ABSTRACT

Acute myeloid leukemia (AML) is a malignant proliferative disorder in which leukemic cells fail to terminally differentiate and accumulate in the blood and bone marrow. Standard AML therapy requires intensive chemotherapy with a low rate of durable remission and is associated with significant treatment-related toxicity, especially in elderly patients. Therefore, new therapeutic options for the treatment of AML are urgently needed. We previously reported that the novel angiogenic inhibitor, angiocidin, induces differentiation of monocytes to macrophages. Here we investigate the effects of angiocidin on AML cells lines and primary AML cells. Differentiation was assessed by flow cytometry measuring the increase in expression of cell surface marker characteristic of normal macrophages. Four AML cell lines (THP-1, Mono-mac-1, HL-60 and MV4-11) and 5 of 10 primary human AML samples showed evidence of differentiation when cultured in vitro for 24 h with 10 µg/mL angiocidin. Additionally, we found that angiocidin promoted secretion of a number of cytokines from the cell lines as well as patient cells. We next evaluated the effect of angiocidin on a xenotransplanted primary human AML sample engrafted in NSG mice. We found angiocidin monotherapy reduced the human AML burden in bone marrow by 63% relative to untreated control. Interestingly, angiocidin+cytosine arabinoside (Ara-C) combination therapy reduced human AML in bone marrow by 79%. We believe the combination of in vitro data supporting the capacity of angiocidin to drive differentiation in multiple AML cell lines and primary human AML samples and its activity in a xenotransplantation model that reproduces the human disease is significant. These observations support the continued evaluation and development of angiocidin as a potential novel, non-toxic therapy for AML.


Subject(s)
Cell Differentiation , Cytokines/metabolism , Leukemia, Myeloid, Acute/pathology , Proteasome Endopeptidase Complex/metabolism , Animals , Blotting, Western , Cell Proliferation , Flow Cytometry , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , RNA-Binding Proteins , Tumor Cells, Cultured
3.
Exp Mol Pathol ; 93(1): 111-5, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22483848

ABSTRACT

Using an innovative "2-D high performance liquid electrophoresis" (2-D HPLE) technology we identified that a specific fragment of G-protein coupled receptor-associated sorting protein 1 (GASP-1) was present in the sera of breast cancer patients and was over-expressed in early and late stage breast tumors (Tuszynski, G.P. et al., 2011). In this study we further investigated the significance of GASP-1 as a tumor marker by investigating the expression GASP-1 in different kinds of tumors as well as in the sera of patients with various cancers. Over expression of GASP-1 was detected in brain, pancreatic, and breast cancers as compared to their respective normal tissues as assessed by immunohistochemical staining of tissue arrays using a "peptide specific" GASP-1 antibody. We found that across these cancers, GASP-1 was expressed approximately 10 fold more in the cancer as compared to normal tissue. The increase in GASP-1 expression was also seen in hyperplastic and inflammatory lesions of breast and pancreatic cancers as compared to normal tissue. GASP-1 was primarily expressed in the tumor epithelium of the epithelial-derived cancers and in the transformed glial cells of the brain tumors. Using a sensitive "competitive ELISA" for GASP-1, we found that sera from patients with brain, liver, breast and lung cancers expressed 4-7 fold more GASP-1 peptide than sera from normal healthy individuals. These studies identify GASP-1 as a potential new serum and tumor biomarker for several cancers and suggest that GASP-1 may be a novel target for development of cancer therapeutics.


Subject(s)
Biomarkers, Tumor/metabolism , Neoplasms/metabolism , Proteins/metabolism , Antibodies, Neoplasm/immunology , Biomarkers, Tumor/blood , Biomarkers, Tumor/immunology , Brain Neoplasms/blood , Brain Neoplasms/metabolism , Breast Neoplasms/blood , Breast Neoplasms/metabolism , Female , Glioma/blood , Glioma/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Liver Cirrhosis/blood , Lung Neoplasms/blood , Lung Neoplasms/metabolism , Neoplasms/blood , Pancreatic Neoplasms/chemistry , Pancreatic Neoplasms/metabolism , Proteins/analysis , Proteins/immunology
4.
Exp Mol Pathol ; 90(3): 244-51, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21241690

ABSTRACT

Angiocidin, a tumor-associated peptide, has been previously shown to inhibit tumor progression by blocking angiogenesis. We now show that angiocidin has a direct inhibitory effect on tumor cell proliferation. MDA-MB-231 breast cancer cells were inhibited from proliferating in the presence of epidermal growth factor (EGF) and angiocidin. Angiocidin transfected breast cancer cells also displayed growth inhibition in vitro and failed to develop significant tumors in mice as compared to vector controls. The anti-proliferative effect of angiocidin was reversed by treating the cells with the epidermal growth factor receptor (EGFR) inhibitor 4557W, a potent tyrosine kinase inhibitor. Consistent with these results, we found that treatment of breast cancer cells with angiocidin induced a 2.3 fold increase in EGFR tyrosine 845 phosphorylation while no change in phosphorylation was observed in the remaining 16 phosphorylation sites of EGFR and those of its family members as measured by a human EGFR phosphorylation array. Treatment of breast cancer cells with angiocidin also resulted in the activation of nuclear factor ĸB (Nf-ĸB) and the de novo up-regulation of many down-stream genes transcribed by Nf-ĸB, including cytokines, inflammatory mediators and the cell cycle inhibitor p21(waf1). Therefore, angiocidin is a peptide that not only inhibits tumor angiogenesis but also directly induces inhibition of tumor growth progression through the activation of EGFR and down-stream genes transcribed by Nf-ĸB.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation/drug effects , ErbB Receptors/metabolism , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/pharmacology , Animals , Blotting, Western , Breast Neoplasms/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Movement , Female , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Phosphorylation , RNA, Messenger/genetics , RNA-Binding Proteins , Reverse Transcriptase Polymerase Chain Reaction , Transfection
5.
Exp Mol Pathol ; 91(2): 608-13, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21791203

ABSTRACT

An innovative "2-D high performance liquid electrophoresis" (2-D HPLE) technology was used to identify serum biomarkers associated with the early stage of breast cancer in addition to other more advanced stages. 2-D HPLE is a newly developed electrophoretic technology that separates 100s of serum albumin complexes on a polyvinyl membrane based on their surface charges. Association of cancer proteins or their fragments (biomarkers) with pre-existing albumin complexes in the blood of cancer patients results in altered mobility on the membrane. Using 2-D HPLE we identified that a specific fragment of G-protein coupled receptor-associated sorting protein 1 (GASP-1) was present in the sera of patients with early stage disease but absent in sera of normal patients. GASP-1 has been shown to modulate lysosomal sorting and functional down-regulation of a variety of G-protein coupled receptors in neuronal cells. However, no reports have linked GASP-1 to breast cancer pathogenesis. GASP-1 was detected in tumor extracts of 7 cases of Stage 2 and Stage 3 breast cancers, but not in adjacent normal tissue as revealed by western blot analysis using an antibody developed against a GASP-1 peptide identified by our 2-D HPLE technology. Using this antibody, we immunohistochemically detected over-expression of GASP-1 in all of 107 cases of archived ductal breast carcinoma tumor samples, while normal adjacent breast tissue from 12 cases of ductal carcinoma showed little or no staining. Additionally, all 10 cases of metastatic breast carcinoma present in lymph nodes were positive. Strong positive GASP-1 staining was observed in all tumor tissue including ductal carcinoma in situ (DCIS) and invasive ductal carcinoma. Additionally, we observed a wide spectrum of enhanced staining of premalignant ductal epithelial cells present in benign ducts and those found in atypical ductal hyperplasia (ADH). These studies identify GASP-1 as a potential new serum and tumor biomarker for breast cancer and suggest that GASP-1 may be a novel target for the development of breast cancer therapeutics.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Vesicular Transport Proteins/metabolism , Amino Acid Sequence , Breast Neoplasms/pathology , Electrophoresis , Female , Humans , Hydrophobic and Hydrophilic Interactions , Immunohistochemistry , Molecular Sequence Data , Neoplasm Proteins/metabolism , Neoplasm Staging , Peptides/chemistry , Peptides/metabolism , Protein Binding , Serum Albumin , Vesicular Transport Proteins/chemistry
6.
Exp Mol Pathol ; 88(1): 15-25, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19931242

ABSTRACT

Angiocidin, a matrix bound and tumor associated protein, has been shown to inhibit tumor progression and angiogenesis. We previously demonstrated that angiocidin binds to thrombospondin-1 and alpha2beta1 integrin. We now show that angiocidin binds and is a preferred substrate for tissue transglutaminase-2 (tTgase). Angiocidin bound tTgase saturably with a Kd of 26 nM, while an angiocidin deletion mutant missing the matrix binding domain of angiocidin failed to bind tTgase. tTgase colocalized with angiocidin on endothelial cells. tTgase bound anti-angiocidin immunoprecipitates of endothelial cell lysates. Breast cancer cells expressing high levels of tTgase attached to angiocidin immobilized on tissue culture plates. Angiocidin was a preferred substrate for tTgase forming high molecular weight cross-linked multimers when treated with tTgase. Cross-linked angiocidin contained iso-peptide bonds as demonstrated by Western blotting and immunohistochemical colocalization studies using endothelial cells treated with angiocidin. Cross-linked angiocidin inhibited cell migration in contrast to monomeric angiocidin and inhibited localization of fibronectin (FN), a pro-tumorigenic matrix protein, into the extracellular matrix (ECM) of tumor and HUVE cells. Our studies provide an additional explanation for the anti-tumor activity of angiocidin suggesting that cross-linked angiocidin disrupts the tumor ECM making it less permissive for tumor growth.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/enzymology , GTP-Binding Proteins/metabolism , Transglutaminases/metabolism , Animals , Breast Neoplasms/enzymology , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Cell Adhesion/drug effects , Cell Line , Cell Movement/drug effects , Child , Endothelial Cells/drug effects , Extracellular Matrix/metabolism , Fibronectins/metabolism , Gene Deletion , Guinea Pigs , Humans , Neovascularization, Pathologic/physiopathology , Proteasome Endopeptidase Complex , Protein Binding , Protein Glutamine gamma Glutamyltransferase 2 , RNA-Binding Proteins , Recombinant Proteins
7.
Circ Res ; 100(9): 1308-16, 2007 May 11.
Article in English | MEDLINE | ID: mdl-17413041

ABSTRACT

Thrombospondin-1 is a multifunctional protein interacting with several cell surface receptors including integrins. We found that it is a ligand for alpha9beta1 integrin, and has an integrin binding site within its N-terminal domain (NoC1). Interaction of thrombospondin-1 and its recombinant NoC1 domain with alpha9beta1 integrin was confirmed in ELISA and cell adhesion assays. Binding of NoC1 to cells expressing alpha9beta1 integrin activated signaling proteins such as Erk1/2 and paxillin. Blocking of this integrin by monoclonal antibody and the met-leu-asp-disintegrin inhibited dermal human microvascular endothelial cell proliferation and NoC1-induced migration of these cells. Immunohistochemical studies revealed that alpha9beta1 is expressed on microvascular endothelium in several organs including skin, lung, heart and brain. NoC1 induced neovascularization in an experimental quail chorioallantoic membrane system and Matrigel plug formation assay in mice. This proangiogenic activity of NoC1 in vivo was inhibited by alpha9beta1 inhibitors. In summary, our results revealed that alpha9beta1 integrin expressed on microvascular endothelial cells interacts with thrombospondin-1, and this interaction is involved in modulation of angiogenesis.


Subject(s)
Integrins/physiology , Neovascularization, Physiologic , Thrombospondin 1/physiology , Cell Adhesion , Cell Movement , Cell Proliferation , Endothelial Cells/cytology , Humans , K562 Cells , Protein Structure, Tertiary , Thrombospondin 1/chemistry
8.
Exp Mol Pathol ; 87(3): 184-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19747478

ABSTRACT

Thrombospondin-1 (TSP-1), a matrix-bound adhesive glycoprotein, has been shown to modulate tumor progression. We previously demonstrated that TSP-1 up-regulates matrix metalloproteinases MMP-2 and MMP-9. Our studies suggested that the balance between MMPs and tissue inhibitors of metalloproteinases (TIMPs) is a key determinant in tumor cell invasion. We now report that TSP-1 up-regulates TIMP-1 expression in both human breast and prostate cancer cell lines. The effect of TSP-1 on TIMP-1 expression was examined in human breast adenocarcinoma cell lines (MDA-MB-231) and human prostate cancer cell lines (PC3-NI and PC3-ML) treated with exogenous TSP-1. TIMP-1 expression was also examined in TSP-1 stably transfected breast cancer cell line (MDA-MB-435). Northern and western blot analysis revealed TIMP-1 mRNA and TIMP-1 protein expression increased with increasing concentrations of TSP-1. This effect was inhibited by antibodies against the type I repeat domain of TSP-1 further suggesting that TSP-1 mediates TIMP-1 secretion. Inhibition of TSP-1 induced TIMP-1 levels increased tumor cell invasion. We conclude that TSP-1 is involved in influencing the critical balance between MMPs and their inhibitors, maintaining the controlled degradation of the extracellular matrix needed to support metastasis and our results may provide an explanation for the divergent activities reported for TSP-1 in tumor progression.


Subject(s)
Breast Neoplasms/pathology , Carcinoma/pathology , Prostatic Neoplasms/pathology , Thrombospondin 1/metabolism , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Breast Neoplasms/metabolism , Carcinoma/metabolism , Cell Line, Tumor , Female , Humans , Male , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Prostatic Neoplasms/metabolism , Thrombospondin 1/pharmacology , Up-Regulation
9.
Int J Cancer ; 123(9): 2195-203, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18712720

ABSTRACT

The presented results show the effect of targeting of collagen receptor, alpha1beta1 integrin expressed on the endothelial cells on the development of experimental melanoma and pathological angiogenesis. Obtustatin, a snake venom KTS-disintegrin, was applied as a specific inhibitor of this integrin. This low molecular weight peptide revealed a potent therapeutic effect on melanoma progression in 2 animal systems, mouse and quail. Its oncostatic effect was related to the inhibition of angiogenesis. Obtustatin inhibited the neovascularization ratio on the CAM embryo of quail, which was pathologically induced by the developing tumor. The i.v. administration of obtustatin completely blocked cancer growth of MV3 human melanoma in nude mice. In B16F10 syngeneic mouse model treatment with the disintegrin revealed a lower effect, although the development of the tumor was significantly reduced for both dosages. The mechanism of obtustatin action is related to the blocking of microvascular endothelial cell proliferation, which undergoes apoptosis in caspase-dependent manner. Summarizing, we present studies of low molecular weight disintegrin, obtustatin as a potential therapeutic compound for treatment of melanoma that contain a high level of vascularization.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Integrin alpha1beta1/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Viper Venoms/therapeutic use , Animals , Apoptosis/drug effects , Cell Line, Tumor , Chorioallantoic Membrane/drug effects , Coturnix , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Viper Venoms/pharmacology
10.
Neuro Oncol ; 10(6): 968-80, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074980

ABSTRACT

In the present study we described the role of alpha9beta1 integrin in glioblastoma progression following its interaction with nerve growth factor (NGF). The level of expression of alpha9beta1 on astrocytomas is correlated with increased grade of this brain tumor and is highest on glioblastoma, whereas normal astrocytes do not express this integrin. Two glioblastoma cell lines, LN229 and LN18, that are alpha9beta1 integrin positive and negative, respectively, were used for alpha9beta1 integrin-dependent NGF-induced tumor progression. NGF was a significant promoter of promigratory and pro-proliferative activities of glioblastoma cells through direct interaction with alpha9beta1 integrin and activation of MAPK Erk1/2 pathway. The level of NGF increases approximately threefold in the most malignant glioma tissue when compared with normal brain. This increase is related to secretion of NGF by tumor cells. Specific inhibitors of alpha9beta1 integrin or gene silencing inhibited NGF-induced proliferation of LN229 cell line to the level shown by LN18 cells. VLO5 promoted alpha9beta1-dependent programmed cell death by induction of intrinsic apoptosis pathway in cancer cells. LN229 cells were rescued from proapoptotic effect of VLO5 by the presence of NGF. This disintegrin significantly inhibited tumor growth induced by implantation of LN229 cells to the chorioallantoic membrane (CAM) of quail embryonic model, and this inhibitory effect was significantly abolished by the presence of NGF. alpha9beta1 integrin appears to be an interesting target for blocking the progression of malignant gliomas, especially in light of the stimulatory effect of NGF on the development of these tumors and its ability to transfer proapoptotic signals in cancer cells.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Integrins/metabolism , Nerve Growth Factor/metabolism , Animals , Apoptosis , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Glioblastoma/pathology , Humans , Immunohistochemistry , Xenograft Model Antitumor Assays
11.
Cancer Lett ; 251(1): 28-35, 2007 Jun 18.
Article in English | MEDLINE | ID: mdl-17188426

ABSTRACT

Angiocidin, a tumor-secreted protein, was measured in serum of 27 healthy volunteers and 33 hepatocellular carcinoma (HCC) patients. Healthy controls either hepatitis B surface antigen (HBsAg) positive or negative showed undetectable levels. Patients had levels of angiocidin ranging from 15.09 to 195.73 pg/ml. Patients with stages III-IV had higher levels of angiocidin (97+/-13 pg/ml, n=17) compared to those with stages I-II (63+/-37 pg/ml, n=16), p<0.043. Patients with microsatellite tumor nodules had higher average levels (98+/-55 pg/ml, n=17) compared to those without microsatellite nodules (51+/-27 pg/ml, n=20), p<0.032. Our studies suggest that angiocidin predicts advanced stage and intra-hepatic metastasis.


Subject(s)
Carcinoma, Hepatocellular/pathology , Carrier Proteins/blood , Liver Neoplasms/pathology , Blotting, Western , Carcinoma, Hepatocellular/blood , Carrier Proteins/metabolism , Carrier Proteins/standards , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Liver Neoplasms/blood , Male , Middle Aged , Neoplasm Staging , Prognosis , Proteasome Endopeptidase Complex , RNA-Binding Proteins , Reference Standards
12.
Cancer Res ; 63(9): 2020-3, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12727812

ABSTRACT

A novel disintegrin, obtustatin, was purified from the venom of the Vipera lebetina obtusa viper. Obtustatin is the shortest disintegrin yet described, containing only 41 amino acids. It contains a similar pattern of cysteines to the short disintegrins echistatin and eristostatin but contains the sequence KTS rather than RGD in its active site loop. Obtustatin is a potent and selective inhibitor of alpha1beta1 integrin. It does not inhibit the closely related integrin alpha2beta1, nor a panel of other integrins tested. It does not inhibit ligand binding to the recombinant alpha1 I-domain. Importantly, obtustatin potently inhibited angiogenesis in vivo in the chicken chorioallantoic membrane assay, and in the Lewis lung syngeneic mouse model, it reduced tumor development by half, confirming and extending previous results on the relevance of alpha1beta1 integrin to angiogenesis and suggesting novel approaches to the generation of angiogenesis inhibitors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Disintegrins/pharmacology , Integrin alpha1beta1/antagonists & inhibitors , Viper Venoms/pharmacology , Allantois/blood supply , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/isolation & purification , Animals , Carcinoma, Lewis Lung/drug therapy , Cell Adhesion/drug effects , Chick Embryo , Chorion/blood supply , Chromatography, High Pressure Liquid , Disintegrins/chemistry , Disintegrins/isolation & purification , Humans , K562 Cells , Mass Spectrometry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neovascularization, Physiologic/drug effects , Tumor Cells, Cultured , Viper Venoms/chemistry , Viper Venoms/isolation & purification
13.
Cancer Lett ; 373(1): 27-35, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26797420

ABSTRACT

There is considerable direct evidence that calcium binding protein ANX A2 is a potential target for treating aggressive breast cancer. The most compelling data are based on the finding of ANX A2 overexpression in aggressive triple negative human breast cancer (TNBC) cell lines and in human breast cancer tissues. Previously, we and others reported a unique role of ANX A2 in cancer invasion, including breast cancer. Moreover, we demonstrated that anti-ANX A2 mAb-mediated immunoneutralization of ANX A2 inhibited invasive human breast cancer growth in a xenograft model. We further evaluated the long-term effects of multiple treatments with anti-ANX A2 mAb and its mechanism of inhibition on human breast tumor growth. We now demonstrate that three treatments with anti-ANX A2 mAb led to significant inhibition of breast tumor growth in immunodeficient mice, and that the anti-tumor response was demonstrable from day 94. After treatment, we followed tumor growth for 172 days and demonstrated 67% inhibition of tumor growth without detectable adverse effects. Biochemical analysis demonstrated that anti-ANX A2 mAb treatment caused significant inhibition of conversion of tissue plasminogen activator (tPA) in the tumor microenvironment. This led to disruption of plasmin generation that consequently inhibited activation of MMP-9 and MMP-2. These results suggest that ANX A2 plays an important role in aggressive breast tumor growth by regulating proteolytic pathways in the tumor microenvironment. ANX A2 may represent a new target for the development of therapeutics for treatment of aggressive breast cancer.


Subject(s)
Annexin A2/antagonists & inhibitors , Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Animals , Annexin A2/immunology , Annexin A2/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Enzyme Activation , Female , Fibrinolysin/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice, Nude , Signal Transduction/drug effects , Time Factors , Tissue Plasminogen Activator/metabolism , Tumor Burden , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
Clin Cancer Res ; 10(12 Pt 1): 4150-7, 2004 Jun 15.
Article in English | MEDLINE | ID: mdl-15217952

ABSTRACT

PURPOSE: Thrombospondin 1 (THBS 1) is a matricellular protein capable of modulating angiogenesis. However, the actual role of THBS 1 in angiogenesis and tumor progression remains controversial. Hepatocellular carcinoma (HCC) is a hypervascular tumor characterized by neovascularization. The significance of THBS 1 in HCC remains unknown. In this study, the significance of THBS 1 in HCC was evaluated by correlating its expression with clinicopathological data. The possible role of THBS 1 in the angiogenesis of HCC was also studied by correlating its expression with vascular endothelial growth factor (VEGF) expression. EXPERIMENTAL DESIGN: Sixty HCC patients were recruited in this study. THBS 1 and VEGF protein expression in tumorous livers were localized by immunohistochemical staining and quantified by ELISA. THBS 1 mRNA was quantified by quantitative reverse transcription-PCR. RESULTS: Immunohistochemical staining of THBS 1 was positive in HCC cells in 51.7% of patients and in stromal cells in 65% of patients. Tumor THBS 1 protein level was significantly correlated with its mRNA expression (P = 0.001) and was significantly correlated with tumor VEGF protein levels (P = 0.001). Its expression was significantly associated with the presence of venous invasion (P = 0.008) and advanced tumor stage (P = 0.049). High THBS 1 expression was also a prognostic marker of poor survival in HCC patients. CONCLUSIONS: This study shows that high expression of THBS 1 is associated with tumor invasiveness and progression in HCC. THBS 1 appears to be a proangiogenic factor that stimulates angiogenesis in HCC in view of its positive correlation with VEGF expression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Thrombospondin 1/biosynthesis , Aged , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neovascularization, Pathologic , Prognosis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Treatment Outcome , Vascular Endothelial Growth Factor A/biosynthesis
15.
Curr Pharm Des ; 10(1): 27-37, 2004.
Article in English | MEDLINE | ID: mdl-14754403

ABSTRACT

Angiogenesis, the formation of blood vessels from preexisting ones, plays a crucial role in tumor progression. Activation of an "angiogenic switch" allows tumor cells to invade and metastasize. The growing interest in the use of antiangiogenic agents in the treatment and prevention of cancer lies in the theoretical advantages of this molecularly targeted modality of chemotherapy. Delivery of antiangiogenic agents are not complicated by having to penetrate large bulky masses but, instead, have easy access to tumoral endothelial cells. Antiangiogenic drugs may not cause cytopenias and thus will avoid many of the unwarranted toxicities of standard chemotherapeutic agents. Because they act directly on nascent endothelial cells, antiangiogenic agents may avoid tumor resistance mechanisms. If antiangiogenic agents are successful, they might be applicable to many tumor types and not be dependent on cell type or growth fraction of cells within a tumor. However, several important obstacles remain with regards to using antiangiogenic drugs in clinical trials with which we must contend in order to determine accurately the efficacy of these agents. In this article, we review the different classes of antiangiogenic agents available, ongoing clinical trials, as well as potential pitfalls and future directions in this exciting field.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Angiogenesis Inhibitors/classification , Angiogenesis Inhibitors/pharmacology , Animals , Antineoplastic Agents/classification , Antineoplastic Agents/pharmacology , Clinical Trials as Topic , Humans , Neoplasms/blood supply , Neoplasms/prevention & control
16.
Surgery ; 132(2): 220-5, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12219015

ABSTRACT

BACKGROUND: Thrombospondin-1 (TSP-1) promotes breast cancer cell invasion of collagen by upregulating matrix metalloproteinase-9 (MMP-9) production. Stromal TSP-1 may play a role in regulating tumor cell invasion. We hypothesize that fibroblasts promote breast cancer cell invasion by upregulating the production of MMP-9 through TSP-1. METHODS: MDA-MB-231 human breast carcinoma cells were grown alone or in coculture with human fibroblasts. Gelatin zymography and Western immunoblot analysis for MMP-9 were performed on the coculture cell media and the single cell media. Inhibition of fibroblast-mediated breast tumor cell invasion by an anti-TSP-1 or an anti-MMP-9 antibody was evaluated using a modified Boyden chamber. RESULTS: Coculture experiments showed an increased production of MMP-9 when compared with breast cancer single cell culture or fibroblast single cell culture experiments as demonstrated by zymography and Western immunoblot analysis. Fibroblast-stimulated MMP-9 production was comparable with TSP-1-stimulated MMP-9 production. Anti-TSP-1 antibody and anti-MMP-9 antibody inhibited fibroblast-stimulated tumor cell invasion to 30% and 26% of controls, respectively (P <.05). CONCLUSIONS: Fibroblasts may regulate breast cancer cell invasion by promoting tumor MMP-9 production through TSP-1. Inhibition of stromal TSP-1 stimulation of MMP-9 synthesis may prevent matrix degradation necessary for tumor invasion and metastasis.


Subject(s)
Breast Neoplasms , Fibroblasts/enzymology , Fibroblasts/pathology , Matrix Metalloproteinase 9/biosynthesis , Neoplasm Invasiveness , Antibodies/pharmacology , Coculture Techniques , Collagen/metabolism , Dermis/cytology , Female , Humans , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Thrombospondin 1/immunology , Thrombospondin 1/metabolism , Tumor Cells, Cultured/pathology , Up-Regulation
17.
Vasc Endovascular Surg ; 45(1): 55-62, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21193465

ABSTRACT

BACKGROUND: Thrombospondin 1 (TSP-1), fibronectin (Fn), and vitronectin (Vn) promote vascular smooth muscle cell (VSMC) chemotaxis through a variety of second messenger systems, including Ras, ERK1/2, and p38. HYPOTHESIS: Ras, ERK1/2, and p38 differentially affect TSP-1-, Fn-, and Vn-induced VSMC chemotaxis. METHODS: Bovine VSMCs were transfected with Ras N17 or treated with the following inhibitors: a farnesyl protein transferase (FPT) inhibitor, PD098059 (ERK1/2 inhibitor), or SB202190 (p38 inhibitor). Thrombospondin 1, Fn, and Vn were used as chemoattractants. Results were analyzed by analysis of variance (ANOVA) with post hoc testing (P < .05). RESULTS: Ras N17 transfection or FPT inhibitor treatment inhibited TSP-1-, Fn-, and Vn-induced chemotaxis. PD098059 or SB202190 resulted in more inhibition of VSMC migration to TSP-1 than to Fn or Vn. CONCLUSIONS: Ras appears equally relevant in the signal transduction pathways of TSP-1-, Fn-, and Vn-induced VSMC chemotaxis. Thrombospondin 1-induced migration is more dependent upon ERK1/2 and p38 than Fn- or Vn-included migration.


Subject(s)
Chemotaxis , Fibronectins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Thrombospondin 1/metabolism , Vitronectin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , ras Proteins/metabolism , Analysis of Variance , Animals , Cattle , Cells, Cultured , Chemotaxis/drug effects , Humans , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Transfection , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , ras Proteins/genetics
18.
J Oncol ; 2010: 645376, 2010.
Article in English | MEDLINE | ID: mdl-20631908

ABSTRACT

Thrombospondin-1 (TSP-1) is involved in a variety of different cellular processes including cell adhesion, tumor progression, and angiogenesis. This paper reports the novel finding that TSP-1 upregulates integrin alpha6 subunit in human keratinocytes and human breast cancer cells resulting in increased cell adhesion and tumor cell invasion. The effect of TSP-1 on alpha6 subunit expression was examined in human keratinocytes and breast adenocarcinoma cell lines (MDA-MB-231) treated with TSP-1 and in TSP-1 stably transfected breast cancer cells. TSP-1 upregulated alpha6 message and protein in these cells as revealed by differential display, Northern and Western blot analysis and immunohistochemical localization studies. The increased expression of alpha6 was shown to mediate adhesion and invasion of these cells to laminin, a major component of the basement membrane and extracellular matrix (ECM). These data suggest that TSP-1 plays an integral role in the attachment of cells to the ECM facilitating cell motility and angiogenesis.

20.
Cancer Biol Ther ; 8(15): 1507-16, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502781

ABSTRACT

Viperistatin and VP12 isolated from Vipera paleastinae venom showed a potent inhibitory activity against collagen receptors, alpha1beta1 and alpha2beta1 integrins, respectively. Structurally, viperistatin belongs to the disintegrin family of proteins, whereas VP12 is composed of two subunits VP12A and VP12B displaying amino acid sequence homology with heterodimeric C-lectin type proteins. Viperistatin and VP12 used separately and simultaneously inhibited pro-metastatic activities of melanoma cells lines. The level of inhibition of MV3 and HS.939T human cell lines in cell adhesion and migration assays by both compounds was correlated with expression of alpha1beta1 and alpha2beta1 integrins on the cell surface. MV3 cells express collagen receptors to much higher extent than HS.939T and required the application of higher concentrations of inhibitors to block their adhesion to collagen types I and IV. A melanoma cell transmigration assay through a dHMVEC layer revealed that alpha1beta1 integrin plays a significant role in invasion of HS.939T cells, while alpha2beta1 integrin appears to be more important for MV3 cells. In an animal model of hematogenous metastasis of the mouse B16F10 cell line, the inhibitory effect of viperistatin and VP12 was only partial. These data suggest that collagen receptors may be an interesting target for development of new anti-metastatic therapies.


Subject(s)
Antineoplastic Agents/therapeutic use , Integrin alpha1beta1/antagonists & inhibitors , Integrin alpha2beta1/antagonists & inhibitors , Lung Neoplasms/prevention & control , Melanoma, Experimental/secondary , Melanoma/secondary , Neoplasm Proteins/antagonists & inhibitors , Viper Venoms/therapeutic use , Viperidae/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Antineoplastic Agents/pharmacology , Cell Adhesion/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/pathology , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Collagen/physiology , Conserved Sequence , Drug Screening Assays, Antitumor , Humans , Integrin alpha1beta1/physiology , Integrin alpha2beta1/physiology , K562 Cells/drug effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Melanoma/drug therapy , Melanoma/pathology , Melanoma, Experimental/drug therapy , Mice , Molecular Sequence Data , Neoplasm Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Viper Venoms/chemistry , Viper Venoms/isolation & purification , Viper Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL