201
|
Serum Containing Tao-Hong-Si-Wu Decoction Induces Human Endothelial Cell VEGF Production via PI3K/Akt-eNOS Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:195158. [PMID: 23762109 PMCID: PMC3673321 DOI: 10.1155/2013/195158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Revised: 04/11/2013] [Accepted: 04/21/2013] [Indexed: 01/05/2023]
Abstract
Tao-Hong-Si-Wu decoction (TSD) is a famous traditional Chinese medicine (TCM) and widely used for ischemic disease in China. TSD medicated serum was prepared after oral administration of TSD (1.6 g/kg) twice a day for 3 days in rats. TSD medicated serum induced human umbilical vein endothelial cells (HUVECs) proliferation, VEGF secretion, and nitric oxide (NO) production. These promoted effects of TSD were partly inhibited by treatment with PI3K inhibitor (LY294002) or eNOS inhibitor (L-NAME), respectively, and completely inhibited by treatment with LY294002 and L-NAME simultaneously. Western blot analysis findings further indicated that TSD medicated serum upregulated p-Akt and p-eNOS expressions, which were significantly inhibited by LY294002 or L-NAME and completely inhibited by both LY294002 and L-NAME; these results indicated that TSD medicated serum induced HUVECs VEGF expression via PI3K/Akt-eNOS signaling. TSD medicated serum contains hydroxysafflor yellow A, ferulic acid, and ligustilide detected by UPLC with standards, so these effect of TSD medicated serum may be associated with these three active compounds absorbed in serum.
Collapse
|
202
|
Chinese Herbal Formula Huo-Luo-Xiao-Ling Dan Protects against Bone Damage in Adjuvant Arthritis by Modulating the Mediators of Bone Remodeling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:429606. [PMID: 23762133 PMCID: PMC3670518 DOI: 10.1155/2013/429606] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 03/08/2013] [Accepted: 03/08/2013] [Indexed: 01/03/2023]
Abstract
Huo-luo-xiao-ling dan (HLXL) is an herbal mixture that has long been used in traditional Chinese medicine for the treatment of rheumatoid arthritis (RA) and other inflammatory disorders. Despite the availability of potent conventionally used drugs for RA, their limited efficacy in a proportion of patients coupled with their high cost and severe adverse effects has necessitated the search for novel therapeutics for this debilitating disease. Further, the control of both inflammation and bone damage is essential for effective management of arthritis. The aim of our study was to evaluate the efficacy of HLXL against arthritic bone damage in adjuvant arthritis (AA) model of RA. Our results show that HLXL treatment suppressed inflammatory arthritis and reduced bone and cartilage damage in the joints of arthritic Lewis rats. HLXL-induced protection against bone damage was mediated primarily via inhibition of mediators of osteoclastic bone remodeling (e.g., receptor activator of nuclear factor kappa-B ligand; RANKL), skewing of RANKL/osteoprotegerin (OPG) ratio in favor of antiosteoclastic activity, reduction in the number of osteoclasts in the arthrodial joint's bone, and inhibition of cytokine production and MMP activity. Our results suggest that HLXL might offer a promising alternative/adjunct treatment for both inflammation and bone damage in RA.
Collapse
|
203
|
Zeng L, Xiao Q, Chen M, Margariti A, Martin D, Ivetic A, Xu H, Mason J, Wang W, Cockerill G, Mori K, Li JYS, Chien S, Hu Y, Xu Q. Vascular endothelial cell growth-activated XBP1 splicing in endothelial cells is crucial for angiogenesis. Circulation 2013; 127:1712-22. [PMID: 23529610 DOI: 10.1161/circulationaha.112.001337] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Vascular endothelial cell growth factor plays a pivotal role in angiogenesis via regulating endothelial cell proliferation. The X-box binding protein 1 (XBP1) is believed to be a signal transducer in the endoplasmic reticulum stress response. It is unknown whether there is crosstalk between vascular endothelial cell growth factor signaling and XBP1 pathway. METHODS AND RESULTS We found that vascular endothelial cell growth factor induced the kinase insert domain receptor internalization and interaction through C-terminal domain with the unspliced XBP1 and the inositol requiring enzyme 1 α in the endoplasmic reticulum, leading to inositol requiring enzyme 1 α phosphorylation and XBP1 mRNA splicing, which was abolished by siRNA-mediated knockdown of kinase insert domain receptor. Spliced XBP1 regulated endothelial cell proliferation in a PI3K/Akt/GSK3β/β-catenin/E2F2-dependent manner and modulated the cell size increase in a PI3K/Akt/GSK3β/β-catenin/E2F2-independent manner. Knockdown of XBP1 or inositol requiring enzyme 1 α decreased endothelial cell proliferation via suppression of Akt/GSK3β phosphorylation, β-catenin nuclear translocation, and E2F2 expression. Endothelial cell-specific knockout of XBP1 (XBP1ecko) in mice retarded the retinal vasculogenesis in the first 2 postnatal weeks and impaired the angiogenesis triggered by ischemia. Reconstitution of XBP1 by Ad-XBP1s gene transfer significantly improved angiogenesis in ischemic tissue in XBP1ecko mice. Transplantation of bone marrow from wild-type o XBP1ecko mice could also slightly improve the foot blood reperfusion in ischemic XBP1ecko mice. CONCLUSIONS These results suggest that XBP1 can function via growth factor signaling pathways to regulate endothelial proliferation and angiogenesis.
Collapse
Affiliation(s)
- Lingfang Zeng
- Cardiovascular Division, King's College London, 125 Coldharbour Lane, London, UK. or
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
204
|
Parathyroid hormone-related protein protects renal tubuloepithelial cells from apoptosis by activating transcription factor Runx2. Kidney Int 2013; 83:825-34. [PMID: 23364519 DOI: 10.1038/ki.2012.476] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Runx2 is a key transcription factor in bone development regulating several processes, including osteoblast apoptosis. The antiapoptotic effects of parathyroid hormone (PTH) in osteoblasts depend on Runx2-mediated transcription of prosurvival genes. In the kidney, PTH-related protein (PTHrP) promotes tubulointerstitial cell survival by activating the PTH/PTHrP type 1 receptor. We found that Runx2 is expressed in renal tubuloepithelial MCT and HK2 cell lines in vitro and in the mouse kidney tubuloepithelium in vivo. The 1-36 amino-acid fragment of PTHrP was found to increase the expression and nuclear translocation of Runx2 in both cell lines in a dose- and time-dependent manner. PTHrP(1-36) protected renal tubuloepithelial cells from folic acid toxicity and serum deprivation, an effect inhibited by a dominant-negative Runx2 construct or a Runx2 siRNA. Furthermore, PTHrP(1-36) upregulated the antiapoptotic proteins Bcl-2 and osteopontin, and these effects were abolished by Runx2 siRNA. Runx2, osteopontin, and Bcl-2 were increased in tubuloepithelial cells from transgenic mice with PTHrP overexpression and in wild-type mice with acute or chronic renal failure. Thus, PTHrP regulates renal tubuloepithelial cell survival via Runx2 in the mammalian kidney.
Collapse
|
205
|
Du J, Lu X, Long Z, Zhang Z, Zhu X, Yang Y, Xu J. In vitro and in vivo anticancer activity of aconitine on melanoma cell line B16. Molecules 2013; 18:757-67. [PMID: 23299553 PMCID: PMC6270132 DOI: 10.3390/molecules18010757] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2012] [Revised: 11/29/2012] [Accepted: 01/05/2013] [Indexed: 01/07/2023] Open
Abstract
The anti-tumor effect of aconitine in melanoma cell line B16 has been studied in this paper. We found that B16 cells showed significantly reduced growth rates and increased apoptotic effects in the presence of aconitine. Furthermore, aconitine inhibited the PI3K/AKT and MAPK/ERK1/2 signaling pathways, thus regulating the levels of protein and mRNA of PCNA and apoptotic related signaling molecules. Above all, we found that aconitine showed an anti-melanoma effect in suppressing tumor growth in vivo. In conclusion, we show that aconitine may be a useful anticancer drug in the future.
Collapse
Affiliation(s)
- Juan Du
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaonian Lu
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ziwen Long
- Shanghai Cancer Center, Fudan University, Shanghai 200040, China
| | - Zhen Zhang
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaohua Zhu
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yongsheng Yang
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jinhua Xu
- Dermatology Department, Huashan Hospital, Fudan University, Shanghai 200040, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel./Fax: +86-21-5288-7776
| |
Collapse
|
206
|
Das SK, Bhutia SK, Azab B, Kegelman TP, Peachy L, Santhekadur PK, Dasgupta S, Dash R, Dent P, Grant S, Emdad L, Pellecchia M, Sarkar D, Fisher PB. MDA-9/syntenin and IGFBP-2 promote angiogenesis in human melanoma. Cancer Res 2012; 73:844-54. [PMID: 23233738 DOI: 10.1158/0008-5472.can-12-1681] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Melanoma differentiation-associated gene-9 (mda-9/syntenin) encodes an adapter scaffold protein whose expression correlates with and mediates melanoma progression and metastasis. Tumor angiogenesis represents an integral component of cancer metastasis prompting us to investigate a possible role of mda-9/syntenin in inducing angiogenesis. Genetic (gain-of-function and loss-of-function) and pharmacologic approaches were used to modify mda-9/syntenin expression in normal immortal melanocytes, early radial growth phase melanoma, and metastatic melanoma cells. The consequence of modifying mda-9/syntenin expression on angiogenesis was evaluated using both in vitro and in vivo assays, including tube formation assays using human vascular endothelial cells, chorioallantoic membrane (CAM) assays and xenograft tumor animal models. Gain-of-function and loss-of-function experiments confirm that MDA-9/syntenin induces angiogenesis by augmenting expression of several proangiogenic factors/genes. Experimental evidence is provided for a model of angiogenesis induction by MDA-9/syntenin in which MDA-9/syntenin interacts with the extracellular matrix (ECM), activating Src and FAK resulting in activation by phosphorylation of Akt, which induces hypoxia inducible factor 1-α (HIF-1α). The HIF-1α activates transcription of insulin growth factor-binding protein-2 (IGFBP-2), which is secreted thereby promoting angiogenesis and further induces endothelial cells to produce and secrete VEGF-A augmenting tumor angiogenesis. Our studies delineate an unanticipated cell nonautonomous function of MDA-9/syntenin in the context of angiogenesis, which may directly contribute to its metastasis-promoting properties. As a result, targeting MDA-9/syntenin or its downstream-regulated molecules may provide a means of simultaneously impeding metastasis by both directly inhibiting tumor cell transformed properties (autonomous) and indirectly by blocking angiogenesis (nonautonomous).
Collapse
Affiliation(s)
- Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia 23298, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
207
|
Activation of Ras/MEK/ERK signaling in chronic subdural hematoma outer membranes. Brain Res 2012; 1489:98-103. [DOI: 10.1016/j.brainres.2012.10.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 10/07/2012] [Indexed: 01/01/2023]
|
208
|
Bhattacharya SD, Mi Z, Talbot LJ, Guo H, Kuo PC. Human mesenchymal stem cell and epithelial hepatic carcinoma cell lines in admixture: concurrent stimulation of cancer-associated fibroblasts and epithelial-to-mesenchymal transition markers. Surgery 2012; 152:449-54. [PMID: 22938903 DOI: 10.1016/j.surg.2012.06.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 06/07/2012] [Indexed: 01/03/2023]
Abstract
BACKGROUND The microenvironments of neoplasms influence both mesenchymal stem cell differentiation into cancer-associated fibroblasts (CAF) and tumor cell line differentiation to mesenchymal phenotypes via epithelial-to-mesenchymal transition (EMT). Using direct cell-cell contact approximating the microenvironment of a neoplasm, we investigated the role of this interaction in human mesenchymal stem cells (hMSCs) and epithelial hepatic carcinoma SK-Hep1 cells by evaluating CAF differentiation and EMT. METHODS hMSCs and SK-Hep1 cells were homogenously cultured for 12 hours with media only, OPN-R3 aptamer blockade of OPN, or RGD peptide blockade of integrin receptor, negative control mutant OPN-R3 aptamer, and RGE peptide blockade. mRNA was isolated from each subpopulation, and real-time-polymerase chain reaction was performed for CAF markers and EMT transcription factors and structural proteins. RESULTS SK-Hep1 cells in admixture with hMSCs showed increased EMT marker vimentin expression that was ablated with OPN-R3 aptamer or RGD blockade. SK-Hep1 cells when cultured with hMSC admixture increased Snail and Slug expression that was hindered with OPN-R3 aptamer. hMSCs acquired CAF markers tenascin-c and SDF-1 in admixture that was ablated with either OPN-R3 aptamer or RGD blockade. All SK-Hep1 and hMSC negative control subpopulations were statistically equivalent to media-only groups. Fluorescence photography exhibited the critical cell-cell interfaces and acquired EMT traits of SK-Hep1. CONCLUSION We conclude that direct interaction of cell lines closely replicates the native neoplasm microenvironment. Our experiments reveal soluble OPN or integrin receptor blockade independently prevents progression to metastatic phenotype by acquisition of CAF and EMT markers.
Collapse
|
209
|
Marsh T, Pietras K, McAllister SS. Fibroblasts as architects of cancer pathogenesis. Biochim Biophys Acta Mol Basis Dis 2012; 1832:1070-8. [PMID: 23123598 DOI: 10.1016/j.bbadis.2012.10.013] [Citation(s) in RCA: 187] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 10/16/2012] [Accepted: 10/19/2012] [Indexed: 12/19/2022]
Abstract
Studies of epithelial cancers (i.e., carcinomas) traditionally focused on transformation of the epithelium (i.e., the cancer cells) and how aberrant signaling within the cancer cells modulates the surrounding tissue of origin. In more recent decades, the normal cells, blood vessels, molecules, and extracellular components that surround the tumor cells, collectively known as the "tumor microenvironment" or "stroma", have received increasing attention and are now thought to be key regulators of tumor initiation and progression. Of particular relevance to the work reviewed herein are the fibroblasts, which make up the major cell type within the microenvironment of most carcinomas. Due to their inherent heterogeneity, plasticity, and function, it is perhaps not surprising that fibroblasts are ideal modulators of normal and cancerous epithelium; however, these aspects also present challenges if we are to interrupt their tumor-supportive functions. Here, we review the current body of knowledge and the many questions that still remain about the special entity known as the cancer-associated fibroblast. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Timothy Marsh
- Hematology Division, Brigham & Women's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
210
|
Tong B, Lu D, Wei Z, Wang T, Xia Y, Dai Y. Gleditsioside B, a triterpene saponin isolated from the anomalous fruits of Gleditsia sinensis Lam., abrogates bFGF-induced endothelial cell migration through preventing the activation of MMP-2 and FAK via inhibiting ERK and PI3K/AKT signaling pathways. Vascul Pharmacol 2012; 58:118-26. [PMID: 23026290 DOI: 10.1016/j.vph.2012.09.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 07/31/2012] [Accepted: 09/23/2012] [Indexed: 01/15/2023]
Abstract
Angiogenesis has become an attractive target for the treatment of certain diseases such as cancer and rheumatoid arthritis. Our previous studies demonstrated that the saponin fraction from Gleditsia sinensis fruits had anti-angiogenic potential, and Gleditsiosides B (GB) was probably the main active constituent. In the present study, we assessed the effect of GB on endothelial cell migration, a crucial event in angiogenesis, and explored the underlying mechanisms. The migration of endothelial cells was assessed by transwell. The expressions of MMP-2/-9 and TIMP-1/-2 were analyzed by Western blotting, and the activities of MMP-2/-9 were detected by gelatin zymography assay. Moreover, migration-related proteins and signaling pathways, including FAK, MAPKs and PI3K/AKT, were analyzed by Western blotting. It was shown that GB, at a concentration of 10 μM without significant cytotoxicity, could effectively abrogate the migration of human umbilical vein endothelial cells (HUVECs) induced by bFGF. GB also inhibited the expression and activity of MMP-2, elevated the expression of TIMP-1, and restrained the phosphorylations of FAK, ERK, PI3K and AKT in a concentration-dependent manner. The findings suggest that GB was able to abrogate the migration of endothelial cells through down-regulating the activation of MMP-2 and FAK via preventing ERK and PI3K/AKT signaling pathways.
Collapse
Affiliation(s)
- Bei Tong
- Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | | | | | | | | | | |
Collapse
|
211
|
Osteopontin and other regulators of angiogenesis and fibrogenesis in the vitreous from patients with proliferative vitreoretinal disorders. Mediators Inflamm 2012; 2012:493043. [PMID: 23055574 PMCID: PMC3465918 DOI: 10.1155/2012/493043] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2012] [Accepted: 08/30/2012] [Indexed: 02/06/2023] Open
Abstract
The aim of this study was to determine the levels of the angiogenic and fibrogenic factors osteopontin (OPN), high-mobility group box-1 (HMGB1), and connective tissue growth factor (CTGF) and the antiangiogenic and antifibrogenic pigment epithelium-derived factor (PEDF) in the vitreous fluid from patients with proliferative diabetic retinopathy (PDR), proliferative vitreoretinopathy (PVR), and rhegmatogenous retinal detachment with no PVR (RD). Vitreous samples from 48 PDR, 17 PVR and 30 RD patients were studied by enzyme-linked immunosorbent assay. OPN, HMGB1, CTGF, and PEDF levels were significantly higher in PDR patients than in RD patients (P < 0.001; 0.002; <0.001; <0.001, resp.). CTGF and PEDF levels were significantly higher in PVR patients than in RD patients (P < 0.001; 0.004, resp.). Exploratory logistic regression analysis identified significant associations between PDR and high levels of HMGB1, CTGF and PEDF, between PDR with active neovascularization and high levels of CTGF and PEDF, and between PDR with traction retinal detachment and high levels of HMGB1. In patients with PDR, there were significant correlations between the levels of PEDF and the levels of OPN (r = 0.544, P = 0.001), HMGB1 (r = 0.719, P < 0.001), and CTGF (r = 0.715, P < 0.001). In patients with PVR, there were significant correlations between the levels of OPN and the levels of HMGB1 (r = 0.484, P = 0.049) and PEDF (r = 0.559, P = 0.02). Our findings suggest that OPN, HMGB1, and CTGF contribute to the pathogenesis of proliferative vitreoretinal disorders and that increased levels of PEDF may be a response to counterbalance the activity of angiogenic and fibrogenic factors in PDR and PVR.
Collapse
|
212
|
Vong S, Kalluri R. The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis. Genes Cancer 2012; 2:1139-45. [PMID: 22866205 DOI: 10.1177/1947601911423940] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor angiogenesis, the building of blood vessels in an expanding tumor mass, is an elegantly coordinated process that dictates tumor growth and progression. Stromal components of the tumor microenvironment, such as myofibroblasts and the extracellular matrix, collaborate with tumor cells in regulating development. Such myofibroblasts and the extracellular matrix have ever-expanding roles in the angiogenic process as well. This review summarizes how stromal myofibroblasts and the extracellular matrix can modulate tumor angiogenesis, highlighting recent findings.
Collapse
Affiliation(s)
- Sylvia Vong
- Division of Matrix Biology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
213
|
Kim KJ, Choi JS, Kim KW, Jeong JW. The anti-angiogenic activities of glycyrrhizic acid in tumor progression. Phytother Res 2012; 27:841-6. [PMID: 22899320 DOI: 10.1002/ptr.4800] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 07/03/2012] [Accepted: 07/16/2012] [Indexed: 12/24/2022]
Abstract
Glycyrrhizic acid (GA) is the bioactive compound of licorice and has been used as a herbal medicine because of its anti-viral, anti-cancer, and anti-inflammatory properties. This study was designed to investigate the effects of GA on tumor growth, angiogenesis, and the mechanisms underlying the anti-angiogenic activities of GA. We observed that GA inhibited tumor growth and angiogenesis in mice. GA decreased angiogenic activities, such as the migration, invasion, and tube formation of endothelial cells. We also demonstrated that GA reduced the production of reactive oxygen species and activation of ERK in endothelial cells. Our findings suggest that GA is a promising anti-angiogenic therapeutic agent that targets the ERK pathway. Considering that angiogenesis is highly stimulated in the majority of cancers, GA could offer a potent therapeutic agent for cancer.
Collapse
Affiliation(s)
- Kil-Jung Kim
- Department of Biomedical Science, Kyung Hee University, Seoul, Korea
| | | | | | | |
Collapse
|
214
|
Li XD, Chen J, Ruan CC, Zhu DL, Gao PJ. Vascular endothelial growth factor-induced osteopontin expression mediates vascular inflammation and neointima formation via Flt-1 in adventitial fibroblasts. Arterioscler Thromb Vasc Biol 2012; 32:2250-8. [PMID: 22814749 DOI: 10.1161/atvbaha.112.255216] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Adventitia acts as an active participant in vascular inflammation but the precise mechanism underlying adventitia-mediated vascular inflammation is not fully understood. In this study, we sought to determine whether vascular endothelial growth factor (VEGF) regulates osteopontin (OPN) expression through Flt-1 in adventitial fibroblasts (AFs) to mediate vascular inflammation and neointima formation. METHODS AND RESULTS In primary cultured AFs, VEGF increased intracellular and secreted OPN expression in a time- and dose-dependent manner, which was effectively suppressed by a specific anti-Flt-1 hexapeptide. Interestingly, VEGF treatment of AFs enhanced the capability of AF-conditioned medium to stimulate macrophages chemotaxis, and this effect was attenuated after blockade of OPN from AF-conditioned medium. Furthermore, perivascular delivery of anti-Flt-1 peptide preferentially concentrated in the adventitia resulted in a decrease of neointima formation after balloon injury in carotid arteries. The inhibition of neointima formation was preceded by significant reduction of VEGF and OPN expression with concurrent macrophage infiltration into adventitia after injury. Activation of extracellular signal-regulated kinase 1/2 pathway was involved in OPN upregulation and macrophage chemotaxis. CONCLUSIONS These results demonstrate that VEGF/Flt-1 signaling plays a significant role in vascular inflammation and neointima formation by regulating OPN expression in AFs and provide insight into Flt-1 as a potential therapeutic target for vascular diseases.
Collapse
Affiliation(s)
- Xiao-Dong Li
- Laboratory of Vascular Biology and Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | |
Collapse
|
215
|
|
216
|
Zhong ZF, Hoi PM, Wu GS, Xu ZT, Tan W, Chen XP, Cui L, Wu T, Wang YT. Anti-angiogenic effect of furanodiene on HUVECs in vitro and on zebrafish in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2012; 141:721-727. [PMID: 21911050 DOI: 10.1016/j.jep.2011.08.052] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 08/17/2011] [Accepted: 08/25/2011] [Indexed: 05/31/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Furanodiene is an active ingredient of the traditional Chinese medicine, Rhizoma Curcumae, commonly used for the treatment of cancer in China. AIM OF THE STUDY To investigate the anti-cancer property of Rhizoma Curcumae, this study describes the anti-angiogenic activities of furanodiene in human umbilical vein endothelial cells (HUVECs) in vitro and in zebrafish in vivo. MATERIALS AND METHODS HUVECs were treated with different doses of furanodiene in the presence or absence of vascular endothelial growth factor (VEGF). The anti-proliferative effect of furanodiene was measured using the XTT assay. The anti-migration and anti-invasion activities of this compound were investigated with a wound-healing migration model and a three-dimensional cell invasion model, respectively. The effects of furanodiene on HUVEC differentiation were assessed by in vitro tube formation in Matrigel™. The expression of related proteins was detected by Western blot. Morphological observations of zebrafish were evaluated in transgenic Tg (fli1: EGFP) zebrafish embryos. RESULTS Our results showed that furanodiene exposure could significantly inhibit the proliferation of HUVECs in a dose-dependent manner and inhibit VEGF-induced proliferation at a low dose. Relative to the VEGF-induced control, the number of invading and migrating cells was significantly reduced in the furanodiene-treated groups. Furanodiene also dramatically suppressed tube formation and p-Akt (Ser473), p-Erk 1/2 (Thr202/Tyr204), ICAM-1, p-p85 (Ser428) as well as p85 protein expression. Furthermore, exposure to furanodiene inhibited angiogenesis in the zebrafish model. CONCLUSIONS This study demonstrated that furanodiene exposure exhibits a potential anti-angiogenic effect through suppression of endothelial cell growth, invasion, migration and tube formation via regulation of the PI3K pathway. This potential anti-angiogenic effect of furanodiene may play an important role in the anti-tumor activity of the traditional Chinese medicine, Rhizoma Curcumae.
Collapse
MESH Headings
- Angiogenesis Inhibitors/isolation & purification
- Angiogenesis Inhibitors/pharmacology
- Animals
- Animals, Genetically Modified
- Antineoplastic Agents, Phytogenic/isolation & purification
- Antineoplastic Agents, Phytogenic/pharmacology
- Blotting, Western
- Cell Differentiation/drug effects
- Cell Movement/drug effects
- Cell Proliferation/drug effects
- Class Ia Phosphatidylinositol 3-Kinase/metabolism
- Curcuma/chemistry
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Furans/isolation & purification
- Furans/pharmacology
- Green Fluorescent Proteins/biosynthesis
- Green Fluorescent Proteins/genetics
- Heterocyclic Compounds, 2-Ring/isolation & purification
- Heterocyclic Compounds, 2-Ring/pharmacology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Intercellular Adhesion Molecule-1/metabolism
- Medicine, Chinese Traditional
- Mitogen-Activated Protein Kinase 1/metabolism
- Mitogen-Activated Protein Kinase 3/metabolism
- Neovascularization, Physiologic/drug effects
- Phosphorylation
- Plants, Medicinal
- Proto-Oncogene Protein c-fli-1/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Serine
- Signal Transduction/drug effects
- Threonine
- Tyrosine
- Vascular Endothelial Growth Factor A/metabolism
- Wound Healing/drug effects
- Zebrafish/embryology
- Zebrafish/genetics
- Zebrafish/metabolism
Collapse
Affiliation(s)
- Zhang-Feng Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau, China
| | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Park D, Kim Y, Kim H, Kim K, Lee YS, Choe J, Hahn JH, Lee H, Jeon J, Choi C, Kim YM, Jeoung D. Hyaluronic acid promotes angiogenesis by inducing RHAMM-TGFβ receptor interaction via CD44-PKCδ. Mol Cells 2012; 33:563-74. [PMID: 22610405 PMCID: PMC3887750 DOI: 10.1007/s10059-012-2294-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 04/06/2012] [Accepted: 04/23/2012] [Indexed: 12/12/2022] Open
Abstract
Hyaluronic acid (HA) has been shown to promote angiogenesis. However, the mechanism behind this effect remains largely unknown. Therefore, in this study, the mechanism of HA-induced angiogenesis was examined. CD44 and PKCδ were shown to be necessary for induction of the receptor for HA-mediated cell motility (RHAMM), a HA-binding protein. RHAMM was necessary for HA-promoted cellular invasion and endothelial cell tube formation. Cytokine arrays showed that HA induced the expression of plasminogen activator-inhibitor-1 (PAI), a downstream target of TGFβ receptor signaling. The induction of PAI-1 was dependent on CD44 and PKCδ. HA also induced an interaction between RHAMM and TGFβ receptor I, and induction of PAI-1 was dependent on RHAMM and TGFβ receptor I. Histone deacetylase 3 (HDAC3), which is decreased by HA via rac1, reduced induction of plasminogen activator inhibitor-1 (PAI-1) by HA. ERK, which interacts with RHAMM, was necessary for induction of PAI-1 by HA. Snail, a downstream target of TGFβ signaling, was also necessary for induction of PAI-1. The down regulation of PAI-1 prevented HA from enhancing endothelial cell tube formation and from inducing expression of angiogenic factors, such as ICAM-1, VCAM-1 and MMP-2. HDAC3 also exerted reduced expression of MMP-2. In this study, we provide a novel mechanism of HA-promoted angiogenesis, which involved RHAMM-TGFβRI signaling necessary for induction of PAI-1.
Collapse
Affiliation(s)
- Deokbum Park
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| | - Youngmi Kim
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| | - Hyunah Kim
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| | - kyungjong Kim
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| | - Yun-Sil Lee
- College of Pharmacy, Ewha Womans University, Seoul 120-750,
Korea
| | - Jongseon Choe
- School of Medicine, Kangwon National University, Chunchon 200-701,
Korea
| | - Jang-Hee Hahn
- School of Medicine, Kangwon National University, Chunchon 200-701,
Korea
| | - Hansoo Lee
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| | - Jongwook Jeon
- Cell Signaling and BioImaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Chulhee Choi
- Cell Signaling and BioImaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon 305-701,
Korea
| | - Young-Myeong Kim
- School of Medicine, Kangwon National University, Chunchon 200-701,
Korea
| | - Dooil Jeoung
- School of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 200-701,
Korea
| |
Collapse
|
218
|
Itraconazole inhibits HMEC-1 angiogenesis. Biomed Pharmacother 2012; 66:312-7. [DOI: 10.1016/j.biopha.2011.11.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 11/21/2011] [Indexed: 12/12/2022] Open
|
219
|
Kong CS, Jeong CH, Choi JS, Kim KJ, Jeong JW. Antiangiogenic Effects of P
-Coumaric Acid in Human Endothelial Cells. Phytother Res 2012; 27:317-23. [DOI: 10.1002/ptr.4718] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 03/29/2012] [Accepted: 04/13/2012] [Indexed: 11/08/2022]
Affiliation(s)
- Chang-Seok Kong
- Department of Biomedical Science, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
| | - Chul-Ho Jeong
- College of Pharmacy; Keimyung University; Daegu 704-701 Korea
| | - Jae-Sun Choi
- Department of Biomedical Science, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
| | - Kil-Jung Kim
- Department of Biomedical Science, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
| | - Joo-Won Jeong
- Department of Biomedical Science, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
- Department of Anatomy and Neurobiology, Biomedical Science Institute, School of Medicine; Kyung Hee University; Seoul 130-701 Korea
| |
Collapse
|
220
|
Fitzpatrick LE, Lisovsky A, Sefton MV. The expression of sonic hedgehog in diabetic wounds following treatment with poly(methacrylic acid-co-methyl methacrylate) beads. Biomaterials 2012; 33:5297-307. [PMID: 22541537 DOI: 10.1016/j.biomaterials.2012.04.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2012] [Accepted: 04/01/2012] [Indexed: 12/15/2022]
Abstract
The expression of native sonic hedgehog (Shh) was significantly increased in poly(methacrylic acid-co-methyl methacrylate) bead (MAA) treated wounds at day 4 compared to both poly(methyl methacrylate) bead (PMMA) treated and untreated wounds in diabetic db/db mice. MAA beads also increased the expression of the Shh transcription factor Gli3 at day 4. Previously, topical application of MAA beads (45 mol % methacrylic acid) improved wound closure and blood vessel density in excisional wounds in these mice, while PMMA beads did not. Gene expression within the granulation tissue of healing wounds was studied to provide insight into the mechanism of vessel formation and wound healing in the presence of MAA beads. In addition to the increased expression of Shh, MAA-treated wounds had increased expression of osteopontin (OPN), IL-1β and TNF-α, (at day 7) similar to the previously reported MAA response of macrophage-like and endothelial cells in vitro.
Collapse
Affiliation(s)
- Lindsay E Fitzpatrick
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada.
| | | | | |
Collapse
|
221
|
Psallidas I, Stathopoulos GT, Maniatis NA, Magkouta S, Moschos C, Karabela SP, Kollintza A, Simoes DCM, Kardara M, Vassiliou S, Papiris SA, Roussos C, Kalomenidis I. Secreted phosphoprotein-1 directly provokes vascular leakage to foster malignant pleural effusion. Oncogene 2012; 32:528-35. [PMID: 22370646 DOI: 10.1038/onc.2012.57] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Secreted phosphoprotein-1 (SPP1) promotes cancer cell survival and regulates tumor-associated angiogenesis and inflammation, both central to the pathogenesis of malignant pleural effusion (MPE). Here, we examined the impact of tumor- and host-derived SPP1 in MPE formation and explored the mechanisms by which the cytokine exerts its effects. We used a syngeneic murine model of lung adenocarcinoma-induced MPE. To dissect the effects of tumor- versus host-derived SPP1, we intrapleurally injected wild-type and SPP1-knockout C57/BL/6 mice with either wild-type or SPP1-deficient syngeneic lung cancer cells. We demonstrated that both tumor- and host-derived SPP1 promoted pleural fluid accumulation and tumor dissemination in a synergistic manner (P<0.001). SPP1 of host origin elicited macrophage recruitment into the cancer-affected pleural cavity and boosted tumor angiogenesis, whereas tumor-derived SPP1 curtailed cancer cell apoptosis in vivo. Moreover, the cytokine directly promoted vascular hyper-permeability independently of vascular endothelial growth factor. In addition, SPP1 of tumor and host origin differentially affected the expression of proinflammatory and angiogenic mediators in the tumor microenvironment. These results suggest that SPP1 of tumor and host origin impact distinct aspects of MPE pathobiology to synergistically promote pleural fluid formation and pleural tumor progression. SPP1 may present an attractive target of therapeutic interventions for patients with MPE.
Collapse
Affiliation(s)
- I Psallidas
- Marianthi Simou Laboratory, 1st Department of Critical Care & Pulmonary Services, Athens Medical School, Evangelismos Hospital, Athens, Greece.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
222
|
Li J, Yang GZ, Zhu ZM, Zhou ZY, Li L. Osteopontin is overexpressed in colorectal carcinoma and is correlated with P53 by immunohistochemistry. Exp Ther Med 2012; 3:621-624. [PMID: 22969940 DOI: 10.3892/etm.2012.465] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 01/17/2012] [Indexed: 12/27/2022] Open
Abstract
Osteopontin (OPN), a secreted phosphorylated glycoprotein, has been found to be involved in carcinogenesis, progression and metastasis of several types of cancers. The aim of the present study was to investigate the immunohistochemical expression of OPN in colorectal carcinoma (CRC) and its relationship with clinicopathological parameters and P53. Expression of OPN, Ki-67 and TP53 was detected in 77 cases of CRC by immunohistochemistry and the correlation of the expression of OPN with clinicopathological features, Ki-67 and P53 staining was investigated. Thirty-eight cases (49.4%) of CRC demonstrated OPN overexpression. Overexpression of OPN was associated with lymph node metastasis (P=0.025) and Dukes' stages (P=0.031), but not with gender, histological differentiation, depth of tumor invasion, TNM stages or Ki-67 index. The correlation between expression of OPN and TP53 was statistically significant (P=0.030). In conclusion, OPN is overexpressed in CRC, and plays a role in tumor progression and metastasis, which is possibly regulated by P53.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, the General Hospital of Beijing Military Command, Beijing 100700
| | | | | | | | | |
Collapse
|
223
|
Fitzpatrick LE, Chan JW, Sefton MV. On the mechanism of poly(methacrylic acid –co– methyl methacrylate)-induced angiogenesis: Gene expression analysis of dTHP-1 cells. Biomaterials 2011; 32:8957-67. [DOI: 10.1016/j.biomaterials.2011.08.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/09/2011] [Indexed: 01/13/2023]
|
224
|
Toward an integrative analysis of the tumor microenvironment in ovarian epithelial carcinoma. CANCER MICROENVIRONMENT 2011; 5:173-83. [PMID: 22109660 DOI: 10.1007/s12307-011-0092-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 11/10/2011] [Indexed: 12/11/2022]
Abstract
Ovarian epithelial carcinomas are heterogeneous malignancies exhibiting great diversity in histological phenotypes as well as genetic and epigenetic aberrations. A general early event in tumorigenesis is regional dissemination into the peritoneal cavity. Initial spread to the peritoneum is made possible by cooperative signaling between a wide array of molecules constituting the tissue microenvironment in the coelomic epithelium. Changes in the activity of key microenvironmental components not constitutively expressed in normal tissue, including several disclosed adhesion molecules, growth factors, proteases, and G-protein coupled receptors (GPCRs), coordinate the transition. Remodeling of the extracellular matrix (ECM) and subsequent cell surface interactions enable transformation by promoting chromosomal instability (CIN) and stimulating several common signal transduction cascades to prepare the tissue for harboring and facilitating growth, angiogenesis and metastasis of the developing tumor.
Collapse
|
225
|
Yuen HF, Chan KK, Grills C, Murray JT, Platt-Higgins A, Eldin OS, O'Byrne K, Janne P, Fennell DA, Johnston PG, Rudland PS, El-Tanani M. Ran is a potential therapeutic target for cancer cells with molecular changes associated with activation of the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways. Clin Cancer Res 2011; 18:380-91. [PMID: 22090358 DOI: 10.1158/1078-0432.ccr-11-2035] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE Cancer cells have been shown to be more susceptible to Ran knockdown than normal cells. We now investigate whether Ran is a potential therapeutic target of cancers with frequently found mutations that lead to higher Ras/MEK/ERK [mitogen-activated protein/extracellular signal-regulated kinase (ERK; MEK)] and phosphoinositide 3-kinase (PI3K)/Akt/mTORC1 activities. EXPERIMENTAL DESIGN Apoptosis was measured by flow cytometry [propidium iodide (PI) and Annexin V staining] and MTT assay in cancer cells grown under different conditions after knockdown of Ran. The correlations between Ran expression and patient survival were examined in breast and lung cancers. RESULTS Cancer cells with their PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways inhibited are less susceptible to Ran silencing-induced apoptosis. K-Ras-mutated, c-Met-amplified, and Pten-deleted cancer cells are also more susceptible to Ran silencing-induced apoptosis than their wild-type counterparts and this effect is reduced by inhibitors of the PI3K/Akt/mTORC1 and MEK/ERK pathways. Overexpression of Ran in clinical specimens is significantly associated with poor patient outcome in both breast and lung cancers. This association is dramatically enhanced in cancers with increased c-Met or osteopontin expression, or with oncogenic mutations of K-Ras or PIK3CA, all of which are mutations that potentially correlate with activation of the PI3K/Akt/mTORC1 and/or Ras/MEK/ERK pathways. Silencing Ran also results in dysregulation of nucleocytoplasmic transport of transcription factors and downregulation of Mcl-1 expression, at the transcriptional level, which are reversed by inhibitors of the PI3K/Akt/mTORC1 and MEK/ERK pathways. CONCLUSION Ran is a potential therapeutic target for treatment of cancers with mutations/changes of expression in protooncogenes that lead to activation of the PI3K/Akt/mTORC1 and Ras/MEK/ERK pathways.
Collapse
Affiliation(s)
- Hiu-Fung Yuen
- Center for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Imano M, Okuno K, Itoh T, Satou T, Ishimaru E, Yasuda T, Hida JI, Imamoto H, Takeyama Y, Shiozaki H. Osteopontin Induced by Macrophages Contribute to Metachronous Liver Metastases in Colorectal Cancer. Am Surg 2011. [DOI: 10.1177/000313481107701143] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Even after radical surgery for stage II and stage III colorectal cancer, metachronous liver metastasis is frequently observed. The aim of this study was to identify the risk of metachronous liver metastasis with retrospective clinicopathological study. Immunohistochemistry was performed to evaluate the expression of Osteopontin (OPN), CD-68, and CD105 in 41 cases of stage II and stage III colorectal cancer tissue. Stage II and stage III colorectal cancer patients who had undergone R0 resection were classified into two groups: with metachronous liver metastasis (m-LM; n = 17) and without liver metastases (control; n = 24). Additionally, double-immunofluorescence staining was performed using antibodies to OPN and CD68. OPN-positive cells were frequently colocalized with CD68 immunoreactivity. OPN and microvascular density expression in the central area were significantly higher in the m-LM (OPN; control 4.3 ± 0.56, m-LV 10.8 ± 1.48, P < 0.05; microvascular density control 18.5 ± 2.86, m-LV 31.4 ± 4.39, P < 0.05), while CD68 expression in the invasive margin was significantly higher in the control group (control 98.9 ± 7.31, m-LV 28.2 ± 3.18, P < 0.05). These results suggest that the risk of metachronous liver metastasis could be well predicted by immunohistochemical staining of OPN in the central areas, and CD68 in the invasive margins of tumors.
Collapse
Affiliation(s)
- Motohiro Imano
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
- Ambulatory Treatment Center, Kinki University Hospital, Ohno-higashi Osaka-Sayama, Osaka, Japan
| | - Kiyotaka Okuno
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Tatsuki Itoh
- Departments of Pathology, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Takao Satou
- Departments of Pathology, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Eizaburo Ishimaru
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Takushi Yasuda
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Jin-Ichi Hida
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Haruhiko Imamoto
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| | - Yoshifumi Takeyama
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
- Ambulatory Treatment Center, Kinki University Hospital, Ohno-higashi Osaka-Sayama, Osaka, Japan
| | - Hitoshi Shiozaki
- Departments of Surgery, Kinki University Faculty of Medicine, Ohno-Higashi Osaka-Sayama, Osaka, Japan
| |
Collapse
|
227
|
PERIOSTIN regulates MMP-2 expression via the αvβ3 integrin/ERK pathway in human periodontal ligament cells. Arch Oral Biol 2011; 57:52-9. [PMID: 21885032 DOI: 10.1016/j.archoralbio.2011.07.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/14/2011] [Accepted: 07/28/2011] [Indexed: 01/05/2023]
Abstract
OBJECTIVE During orthodontic tooth movement, activation of the vascular system in the compressed periodontal ligament (PDL), which becomes hypoxic, is essential for periodontal tissue remodelling. PERIOSTIN, an extracellular matrix protein, is expressed in PDL and its concentration is increased on the compressive side during orthodontic tooth movement. PERIOSTIN promotes angiogenesis through upregulation of matrix metalloproteinase (MMP)-2, which has been shown to be expressed via αvβ3 integrin/extracellular signal-related kinase (ERK) signalling pathway and vascular endothelial growth factor (VEGF). Therefore, we hypothesized that hypoxia-induced PERIOSTIN promotes MMP-2 expression via αvβ3 integrin/ERK signalling and VEGF in PDL cells. METHODS Human PDL cells were cultured in condition medium containing desferrioxamine (DFO) to mimic hypoxia. The total RNA, cell lysates or supernatant were collected, and MMP2 and VEGF expression, PERIOSTIN expression and ERK phosphorylation, and MMP-2 activity were analysed by real-time RT-PCR, western blot analysis, and zymography, respectively. A recombinant human PERIOSTIN or PERIOSTIN siRNA was applied to the cells, then the total RNA was extracted to measure MMP-2 and VEGF expression. The cells were treated with αvβ3 integrin-blocking antibody or ERK inhibitor followed by PERIOSTIN stimulation. MMP-2 expression was measured by real-time RT-PCR. RESULTS PERIOSTIN was upregulated in a time-dependent manner in human PDL cells treated with DFO, a chemical hypoxia mimic. MMP-2 and VEGF expression, and MMP-2 activity were increased by DFO or PERIOSTIN treatment, and decreased by PERIOSTIN silencing. PERIOSTIN treatment also induced ERK phosphorylation, and PERIOSTIN-induced MMP-2 was reduced by αvβ3 integrin-blocking antibody or ERK inhibitor. CONCLUSION These data suggest that PERIOSTIN upregulates MMP-2 expression via the αvβ3 integrin/ERK signalling pathway and VEGF expression in human PDL cells.
Collapse
|
228
|
Berge G, Pettersen S, Grotterød I, Bettum IJ, Boye K, Mælandsmo GM. Osteopontin--an important downstream effector of S100A4-mediated invasion and metastasis. Int J Cancer 2011; 129:780-90. [PMID: 20957651 DOI: 10.1002/ijc.25735] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Accepted: 09/21/2010] [Indexed: 01/10/2023]
Abstract
Substantial evidence has linked the small calcium-binding protein S100A4 to metastatic progression. S100A4-mediated effects include stimulation of angiogenesis, regulation of cell death and increased cell motility and invasion, but the exact molecular mechanisms by which the protein exerts these effects are incompletely elucidated. In the present study, we demonstrate that S100A4 induces NF-κB-dependent expression and secretion of osteopontin (OPN) in a selection of osteosarcoma cell lines. OPN is, as S100A4, known to result in a variety of cellular effects potentially leading to increased angiogenesis and metastasis, and several of the activated signaling pathways are common for the two proteins. In our study, extracellular S100A4 was found to upregulate enzymes of the plasminogen activator system and matrix metalloproteinase (MMP) family, especially urokinase plasminogen activator and MMP-13. Furthermore, increased motility and invasion was observed in vitro as a result of S100A4 treatment. OPN expression was inhibited by the use of siRNA molecules, and a partial blocking of S100A4-induced effects on protease expression and invasive capacity was detected. In conclusion, our results suggest regulation of OPN as a downstream molecular mechanism of S100A4 signaling. This novel finding adds more information to how S100A4 mediates tumor development and metastatic progression. The observation of a link between S100A4 and OPN, and also identification of common downstream effect molecules, highlights them, their receptors or downstream proteins, as targets for therapeutic approaches.
Collapse
Affiliation(s)
- Gisle Berge
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
229
|
Ghanaati S, Unger RE, Webber MJ, Barbeck M, Orth C, Kirkpatrick JA, Booms P, Motta A, Migliaresi C, Sader RA, Kirkpatrick CJ. Scaffold vascularization in vivo driven by primary human osteoblasts in concert with host inflammatory cells. Biomaterials 2011; 32:8150-60. [PMID: 21821280 DOI: 10.1016/j.biomaterials.2011.07.041] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Accepted: 07/13/2011] [Indexed: 10/17/2022]
Abstract
Successful cell-based tissue engineering requires a rapid and thorough vascularization in order to ensure long-term implant survival and tissue integration. The vascularization of a scaffold is a complex process, and is modulated by the presence of transplanted cells, exogenous and endogenous signaling proteins, and the host tissue reaction, among other influencing factors. This paper presents evidence for the significance of pre-seeded osteoblasts for the in vivo vascularization of a biodegradable scaffold. Human osteoblasts, cultured on silk fibroin micronets in vitro, migrated throughout the interconnected pores of the scaffold and produced extensive bone matrix. When these constructs were implanted in SCID mice, a rapid and thorough vascularization of the scaffold by the host blood capillaries occurred. This profound response was not seen for the silk fibroin scaffold alone. Moreover, when the pre-cultivation time of human osteoblasts was reduced from 14 days to only 24 h, the significant effect these cells exerted on vascularization rate in vivo was still detectable. From these studies, we conclude that matrix and soluble factors produced by osteoblasts can serve to instruct host endothelial cells to migrate, proliferate, and initiate the process of scaffold vascularization. This finding represents a potential paradigm shift for the field of tissue engineering, especially in bone, as traditional strategies to enhance scaffold vascularization have focused on endovascular cells and regarded osteoblasts primarily as cell targets for mineralization. In addition, the migration of host macrophages and multinucleated giant cells into the scaffold was also found to influence the vascularization of the biomaterial. Therefore, the robust effect on scaffold vascularization seen by pre-culturing with osteoblasts appears to occur in concert with the pro-angiogenic stimuli arising from host immune cells.
Collapse
Affiliation(s)
- Shahram Ghanaati
- REPAIR-Lab, Institute of Pathology, Johannes Gutenberg University, Langenbeckstrasse. 1, 55101 Mainz, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
230
|
Wang Y, Yan W, Lu X, Qian C, Zhang J, Li P, Shi L, Zhao P, Fu Z, Pu P, Kang C, Jiang T, Liu N, You Y. Overexpression of osteopontin induces angiogenesis of endothelial progenitor cells via the avβ3/PI3K/AKT/eNOS/NO signaling pathway in glioma cells. Eur J Cell Biol 2011; 90:642-8. [DOI: 10.1016/j.ejcb.2011.03.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2010] [Revised: 03/21/2011] [Accepted: 03/21/2011] [Indexed: 01/10/2023] Open
|
231
|
Poggio P, Grau JB, Field BC, Sainger R, Seefried WF, Rizzolio F, Ferrari G. Osteopontin controls endothelial cell migration in vitro and in excised human valvular tissue from patients with calcific aortic stenosis and controls. J Cell Physiol 2011; 226:2139-49. [PMID: 21520066 DOI: 10.1002/jcp.22549] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Calcific aortic stenosis (CAS) is a pathological condition of the aortic valve characterized by dystrophic calcification of the valve leaflets. Despite the high prevalence and mortality associated with CAS, little is known about its pathogenetic mechanisms. Characterized by progressive dystrophic calcification of the valve leaflets, the early stages of aortic valve degeneration are similar to the active inflammatory process of atherosclerosis including endothelial disruption, inflammatory cell infiltration, lipid deposition, neo-vascularization and calcification. In the vascular system, the endothelium is an important regulator of physiological and pathological conditions; however, the contribution of endothelial dysfunction to valvular degeneration at the cellular and molecular level has received little attention. Endothelial cell (EC) activation and neo-vascularization of the cusps characterizes all stages of aortic valvular degeneration from aortic sclerosis to aortic stenosis. Here we reported the role of osteopontin (OPN) in the regulation of EC activation in vitro and in excised tissue from CAS patients and controls. OPN is an important pro-angiogenic factor in several pathologies. High levels of OPN have been demonstrated in both tissue and plasma of patients with aortic valve sclerosis and stenosis. The characterization of valvular ECs as a cellular target for OPN will help us uncover the pathogenesis of aortic valve degeneration and stenosis, opening new perspectives for the prevention and therapy of this prevalent disease.
Collapse
Affiliation(s)
- Paolo Poggio
- Division of Cardiovascular Surgery, Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania 19036, USA
| | | | | | | | | | | | | |
Collapse
|
232
|
Ahmed M, Behera R, Chakraborty G, Jain S, Kumar V, Sharma P, Bulbule A, Kale S, Kumar S, Mishra R, Raja R, Saraswati S, Kaur R, Soundararajan G, Kumar D, Thorat D, Sanyal M, Ramdasi A, Ghosh P, Kundu GC. Osteopontin: a potentially important therapeutic target in cancer. Expert Opin Ther Targets 2011; 15:1113-26. [PMID: 21718227 DOI: 10.1517/14728222.2011.594438] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
INTRODUCTION Cancer is an extremely complex disease and most cancer treatments are limited to chemotherapy, radiation and surgery. The progression of tumours towards malignancy requires the interaction of various cytokines, growth factors, transcription factors and effector molecules. Osteopontin is a cytokine-like, calcium-binding, extracelular-matrix- associated member of the small integrin-binding ligand, N-linked glycoprotein (SIBLING) family of proteins. It plays an important role in determining the oncogenic potential of various cancers. The role of osteopontin in various pathophysiological conditions suggests that the alteration in post-translational modification result in different functional forms that might change its normal physiological functions. AREAS COVERED Osteopontin -based anticancer therapy, which may provide a new insight for the effective management of cancer. EXPERT OPINION A better understanding of the signalling mechanism by which osteopontin promotes tumourigenesis may be useful in crafting novel osteopontin -based anticancer therapy. The role of osteopontin in promoting cancer progression is the subject of in depth investigation and thus targeting osteopontin might be a suitable therapeutic approach for the treatment of cancer.
Collapse
Affiliation(s)
- Mansoor Ahmed
- National Center for Cell Science , NCCS Complex, Ganeshkhind, Pune, India
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
233
|
Buckner CM, Calderon TM, Willams DW, Belbin TJ, Berman JW. Characterization of monocyte maturation/differentiation that facilitates their transmigration across the blood-brain barrier and infection by HIV: implications for NeuroAIDS. Cell Immunol 2010; 267:109-23. [PMID: 21292246 DOI: 10.1016/j.cellimm.2010.12.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 11/02/2010] [Accepted: 12/18/2010] [Indexed: 01/10/2023]
Abstract
The prevalence of human immunodeficiency virus 1 (HIV) associated neurocognitive disorders resulting from infection of the central nervous system (CNS) by HIV continues to increase despite the success of combination antiretroviral therapy. Although monocytes are known to transport HIV across the blood-brain barrier (BBB) into the CNS, there are few specific markers that identify monocyte subpopulations susceptible to HIV infection and/or capable of infiltrating the CNS. We cultured human peripheral blood monocytes and characterized the expression of the phenotypic markers CD14, CD16, CD11b, Mac387, CD163, CD44v6 and CD166 during monocyte/macrophage (Mo/Mac) maturation/differentiation. We determined that a CD14(+)CD16(+)CD11b(+)Mac387(+) Mo/Mac subpopulation preferentially transmigrates across our in vitro BBB model in response to CCL2. Genes associated with Mo/Mac subpopulations that transmigrate across the BBB and/or are infected by HIV were identified by cDNA microarray analyses. Our findings contribute to the understanding of monocyte maturation, infection and transmigration into the brain during the pathogenesis of NeuroAIDS.
Collapse
Affiliation(s)
- Clarisa M Buckner
- Departments of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | | | |
Collapse
|
234
|
Shibata T, Saito S, Kokubu A, Suzuki T, Yamamoto M, Hirohashi S. Global downstream pathway analysis reveals a dependence of oncogenic NF-E2-related factor 2 mutation on the mTOR growth signaling pathway. Cancer Res 2010; 70:9095-105. [PMID: 21062981 DOI: 10.1158/0008-5472.can-10-0384] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In multicellular organisms, adaptive responses to oxidative stress are regulated by NF-E2-related factor 2 (NRF2), a master transcription factor of antioxidant genes and phase II detoxifying enzymes. Aberrant activation of NRF2 by either loss-of-function mutations in the Keap1 gene or gain-of-function mutations in the Nrf2 gene occurs in a wide range of human cancers, but details of the biological consequences of NRF2 activation in the cancer cells remain unclear. Here, we report that mutant NRF2 induces epithelial cell proliferation, anchorage-independent growth, and tumorigenicity and metastasis in vivo. Genome-wide gene expression profiling revealed that mutant NRF2 affects diverse molecular pathways including the mammalian target of rapamycin (mTOR) pathway. Mutant NRF2 upregulates RagD, a small G-protein activator of the mTOR pathway, which was also overexpressed in primary lung cancer. Consistently, Nrf2-mutated lung cancer cells were sensitive to mTOR pathway inhibitors (rapamycin and NVP-BEZ235) in both in vitro and an in vivo xenograft model. The gene expression signature associated with mutant NRF2 was a marker of poor prognosis in patients with carcinoma of the head and neck region and lung. These results show that oncogenic Nrf2 mutation induces dependence on the mTOR pathway during carcinogenesis. Our findings offer a rationale to target NRF2 as an anticancer strategy, and they suggest NRF2 activation as a novel biomarker for personalized molecular therapies or prognostic assessment.
Collapse
Affiliation(s)
- Tatsuhiro Shibata
- Cancer Genomics Project and Pathology Division, National Cancer Center Research Institute, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
235
|
Prognostic significance of BRMS1 expression in human melanoma and its role in tumor angiogenesis. Oncogene 2010; 30:896-906. [PMID: 20935672 DOI: 10.1038/onc.2010.470] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer metastasis suppressor 1 (BRMS1) has been reported to suppress metastasis without significantly affecting tumorigenicity in breast cancer and ovarian cancer. To investigate the role of BRMS1 in human melanoma progression and prognosis, we established tissue microarray and BRMS1 expression was evaluated by immunohistochemistry in 41 dysplastic nevi, 90 primary melanomas and 47 melanoma metastases. We found that BRMS1 expression was significantly decreased in metastatic melanoma compared with primary melanoma or dysplastic nevi (P=0.021 and 0.001, respectively, χ(2) test). In addition, reduced BRMS1 staining was significantly correlated with American Joint Committee on Cancer stages (P=0.011, χ(2) test), but not associated with tumor thickness, tumor ulceration and other clinicopathological parameters. Furthermore, BRMS1 expression was significantly correlated with disease-specific 5-year survival of melanoma patients (P=0.007, log-rank test). Multivariate Cox regression analysis revealed that BRMS1 staining was an independent prognostic factor for melanoma patients (relative risk=0.51; confidence interval=0.29-0.91; P=0.022). Moreover, we demonstrated that BRMS1 overexpression inhibited endothelial cell growth and tube formation ability by suppressing NF-κB activity and IL-6 expression in vitro. We also showed that knockdown of BRMS1 increased IL-6 expression and promoted endothelial cell growth and tube formation. In addition, our data revealed that the BRMS1-mediated IL-6 expression is dependent on NF-κB. Strikingly, our in vivo studies using nude mice confirmed that BRMS1 inhibited blood vessel formation and the recruitment of CD31-positive cells in matrigel plugs. Taken together, BRMS1 expression was decreased in metastatic melanomas, which resulted in deficient suppression of angiogenesis and contributed to melanoma progression. BRMS1 may serve an important prognostic marker and therapeutic target for melanoma patients.
Collapse
|
236
|
Lazar M, Sullivan J, Chipitsyna G, Gong Q, Ng CY, Salem AF, Aziz T, Witkiewicz A, Denhardt DT, Yeo CJ, Arafat HA. Involvement of osteopontin in the matrix-degrading and proangiogenic changes mediated by nicotine in pancreatic cancer cells. J Gastrointest Surg 2010; 14:1566-77. [PMID: 20824368 DOI: 10.1007/s11605-010-1338-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 08/18/2010] [Indexed: 01/31/2023]
Abstract
BACKGROUND Substantial evidence indicates that exposure to cigarette smoke is associated with an elevated risk of pancreatic ductal adenocarcinoma (PDA). However, the mechanisms underlying the effects of nicotine on the development or progression of PDA remain to be investigated. Previously, we showed that nicotine promotes the expression of osteopontin c (OPNc), an isoform of OPN protein that confers on cancer cells a migratory phenotype. In this study, we explored the potential prometastatic role of nicotine in PDA through studying its effect on the expression of matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF) and evaluated the role of OPN in mediating these effects. MATERIALS AND METHODS MMP-9 and VEGF mRNA and protein were analyzed in PDA cells treated with or without nicotine (3-300 nM). Transient transfection and luciferase-labeled promoter studies evaluated the effects of OPNc and OPN protein on the transcription and translation of MMP-9 and VEGF. Real-time PCR and immunohistochemistry were used to analyze the mRNA expression levels and localization of OPN, MMP-9, and VEGF proteins in matched invasive human PDA and surrounding nonmalignant tissues. RESULTS AND DISCUSSION Nicotine significantly enhanced the expression of MMP-9 and VEGF mRNA and protein in PDA cells. Blocking OPN with siRNA or OPN antibody prevented the nicotine-mediated increase of both MMP-9 and VEGF. Transient transfection of OPNc gene in PDA cells or their treatment with recombinant OPN protein significantly (p < 0.05) increased MMP-9 and VEGF mRNA expression levels and induced their promoter activities. In invasive PDA lesions, MMP-9 mRNA levels were significantly (p < 0.005) higher in smokers vs. nonsmokers. VEGF protein co-localized with MMP-9 and OPN in the malignant ducts and correlated well with their higher levels in invasive PDA lesions. CONCLUSIONS Our data show for the first time that cigarette smoking and nicotine may contribute to PDA metastasis through inducing MMP-9 and VEGF and suggest that OPN plays a central role in mediating these effects. The presence of OPN as a downstream effector of nicotine that is capable of mediating its prometastatic effects in PDA cells is novel and could provide a unique therapeutic target to control pancreatic cancer aggressiveness, especially in the cigarette-smoking population.
Collapse
Affiliation(s)
- Melissa Lazar
- Department of Surgery, Jefferson Pancreatic, Biliary and Related Cancer Center, Thomas Jefferson University, 1015 Walnut Street, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Kou G, Shi J, Chen L, Zhang D, Hou S, Zhao L, Fang C, Zheng L, Zhang X, Liang P, Zhang X, Li B, Guo Y. A bispecific antibody effectively inhibits tumor growth and metastasis by simultaneous blocking vascular endothelial growth factor A and osteopontin. Cancer Lett 2010; 299:130-6. [PMID: 20826049 DOI: 10.1016/j.canlet.2010.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 07/26/2010] [Accepted: 08/13/2010] [Indexed: 11/24/2022]
Abstract
Both vascular endothelial growth factor A (VEGF) and osteopontin (OPN) can directly induce tumor angiogenesis, which is essential for the growth and metastasis of solid tumors. Here we engineered a bispecific antibody (VEGF/OPN-BsAb) using the anti-VEGF-A antibody bevacizumab and the anti-OPN antibody hu1A12. Compared with hu1A12 alone and bevacizumab alone, VEGF/OPN-BsAb was significantly more effective in inhibiting tumor angiogenesis in a highly metastatic human hepatocellular carcinoma nude mouse model. Further study demonstrated that VEGF/OPN-BsAb could effectively suppress primary tumor growth and metastasis to lungs, suggesting that it might be a promising therapeutic agent for treatment of metastatic cancer.
Collapse
Affiliation(s)
- Geng Kou
- International Joint Cancer Institute, The Second Military Medical University, Shanghai 200433, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
238
|
Dianzani C, Minelli R, Mesturini R, Chiocchetti A, Barrera G, Boscolo S, Sarasso C, Gigliotti CL, Sblattero D, Yagi J, Rojo JM, Fantozzi R, Dianzani U. B7h triggering inhibits umbilical vascular endothelial cell adhesiveness to tumor cell lines and polymorphonuclear cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:3970-9. [PMID: 20817864 DOI: 10.4049/jimmunol.0903269] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular endothelial cells (ECs) are key players in leukocyte recruitment into tissues and metastatic dissemination of tumor cells. ECs express B7h, which is the ligand of the ICOS T cell costimulatory molecule. The aim of this work was to assess the effect of B7h triggering by a soluble form of ICOS (ICOS-Fc) on the adhesion of colon carcinoma cell lines to HUVECs. We found that B7h triggering inhibited HUVEC adhesiveness to HT29 and DLD1 cells (by 50 and 35%, respectively) but not to HCT116 cells. The effect was dependent on the ICOS-Fc dose and was detectable as early as 30 min after treatment and was still present after 24 h. It was inhibited by soluble anti-ICOS reagents (mAb and B7h-Fc) and silencing of B7h on HUVECs, and it was not displayed by an F119S mutated form of ICOS-Fc that does not bind B7h. HUVEC treatment with ICOS-Fc did not modulate expression of adhesion molecules and cytokines, but it substantially downmodulated ERK phosphorylation induced by E-selectin triggering or osteopontin, which may influence HUVEC adhesiveness. Moreover, HUVEC treatment with ICOS-Fc also inhibited adhesion of polymorphonuclear cells and several tumor cell lines from different origins. Therefore, the B7h-ICOS interaction may modulate spreading of cancer metastases and recruitment of polymorphonuclear cells in inflammatory sites, which opens a view on the use of ICOS-Fc as an immunomodulatory drug.
Collapse
Affiliation(s)
- Chiara Dianzani
- Department of Anatomy, Pharmacology and Experimental Medicine, University of Torino, Torino, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Abstract
The family of matricellular proteins comprises molecules with disparate biology. The main characteristic of matricellular proteins is to be expressed during tissue renewal and repair in order to "normalize" the tissue. Tumors are wound that do not heal, and tumor growth and metastasis can be viewed as a consequence of aberrant homeostasis, during which matricellular proteins are often upregulated. In the tumor microenvironment, they can be produced by both tumor cells and surrounding stromal cells, such as fibroblasts and macrophages. In this context, matricellular proteins can exert several functions that actively contribute to tumor progression. They may (a) regulate cellular adhesion and migration and extracellular matrix deposition, (b) control tumor infiltration by macrophages or other leukocytes, (c) affect tumor angiogenesis, (d) regulate TGFbeta and other growth factor receptor signals, (e) directly stimulate integrin receptors to transduce pro-survival or pro-migratory signals, and (f) regulate the wnt/beta-catenin pathways. Most of these functions contribute to settle a chronic low inflammatory state, whose involvement in tissue transformation and tumor progression is now established.
Collapse
|
240
|
Anborgh PH, Mutrie JC, Tuck AB, Chambers AF. Role of the metastasis-promoting protein osteopontin in the tumour microenvironment. J Cell Mol Med 2010; 14:2037-44. [PMID: 20597997 PMCID: PMC3822994 DOI: 10.1111/j.1582-4934.2010.01115.x] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Osteopontin (OPN) is a secreted protein present in bodily fluids and tissues. It is subject to multiple post-translational modifications, including phosphorylation, glycosylation, proteolytic cleavage and crosslinking by transglutamination. Binding of OPN to integrin and CD44 receptors regulates signalling cascades that affect processes such as adhesion, migration, invasion, chemotaxis and cell survival. A variety of cells and tissues express OPN, including bone, vasculature, kidney, inflammatory cells and numerous secretory epithelia. Normal physiological roles include regulation of immune functions, vascular remodelling, wound repair and developmental processes. OPN also is expressed in many cancers, and elevated levels in patients’ tumour tissue and blood are associated with poor prognosis. Tumour growth is regulated by interactions between tumour cells and their tissue microenvironment. Within a tumour mass, OPN can be expressed by both tumour cells and cellular components of the tumour microenvironment, and both tumour and normal cells may have receptors able to bind to OPN. OPN can also be found as a component of the extracellular matrix. The functional roles of OPN in a tumour are thus complex, with OPN secreted by both tumour cells and cells in the tumour microenvironment, both of which can in turn respond to OPN. Much remains to be learned about the cross-talk between normal and tumour cells within a tumour, and the role of multiple forms of OPN in these interactions. Understanding OPN-mediated interactions within a tumour will be important for the development of therapeutic strategies to target OPN.
Collapse
Affiliation(s)
- Pieter H Anborgh
- London Regional Cancer Program, London, Ontario, Canada Department of Pathology, University of Western Ontario, London, Ontario, Canada
| | | | | | | |
Collapse
|
241
|
Bhattacharya SD, Garrison J, Guo H, Mi Z, Markovic J, Kim VM, Kuo PC. Micro-RNA-181a regulates osteopontin-dependent metastatic function in hepatocellular cancer cell lines. Surgery 2010; 148:291-7. [PMID: 20576283 DOI: 10.1016/j.surg.2010.05.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2010] [Accepted: 05/14/2010] [Indexed: 12/16/2022]
Abstract
BACKGROUND Osteopontin (OPN) is a variably expressed, secreted glycophosphoprotein that mediates the growth and metastases of hepatocellular cancer (HCC). MicroRNAs (miRNAs) may be responsible for variant OPN expression, interrupting translation by binding OPN messenger RNA (mRNA) in 3'-untranslated regions (UTRs). METHODS A microarray analysis identified miRNAs of interest. Plasmid constructs using a luciferase reporter with variable OPN 3'UTR mutations were transfected into 2 HCC cell lines to determine miRNA regulation of OPN expression. Western blot analyses confirmed variable OPN expression in both cell lines. Invasion, adhesion, and migration evaluated metastatic behavior in Hep G2 and Hep 3B with modified miRNA and OPN expression. RESULTS Hep 3B produces 36 x miRNA 181a compared with Hep G2. Luciferase activity after transfection with miRNA 181a precursor was decreased in both cell lines (P < .01); luciferase activity increased with miRNA 181a inhibitor transfection in both cell lines (P < .01). Hep 3B transfected with mutated OPN 3'UTR increased luciferase activity 108% (P < .01). Hep G2 transfected with miRNA precursor decreased OPN expression 5 x (P < .01) in Western blot analyses. Hep 3B transfection with miRNA precursor increased OPN expression 3 x (P < .01) in Western blot analyses. In vitro metastatic correlates increased in Hep 3B lines after transfection with siOPN and/or miRNA 181a inhibitor (P < .01). CONCLUSION MiRNA 181a decreases OPN expression in HCC cell lines. This previously undescribed mechanism may confer metastatic characteristics to HCC.
Collapse
|
242
|
Imano M, Okuno K, Itoh T, Ishimaru E, Satou T, Shiozaki H. Increased Osteopontin-Positive Macrophage Expression in Colorectal Cancer Stroma with Synchronous Liver Metastasis. World J Surg 2010; 34:1930-6. [DOI: 10.1007/s00268-010-0582-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
243
|
Shih IM, Davidson B. Pathogenesis of ovarian cancer: clues from selected overexpressed genes. Future Oncol 2010; 5:1641-57. [PMID: 20001801 DOI: 10.2217/fon.09.126] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Ovarian cancer is the most malignant gynecologic neoplasm. Although new chemotherapeutic agents have improved patients' 5-year survival rate, the overall mortality of ovarian cancer has remained largely unchanged in the past several decades. The main reason for the lack of success in effectively treating ovarian cancer is our limited understanding of its etiology and the very few molecular diagnostic markers and therapeutic targets known so far. Identification and characterization of ovarian cancer-associated genes are fundamental for unveiling the pathogenesis of its initiation and progression, especially the development of recurrent diseases. As there are a vast number of genes for which molecular genetic changes and aberrant gene expression have been reported in ovarian cancer, this review will only focus on summarizing those exemplified genes that have been demonstrated to have biological functions in promoting ovarian cancer development and potential clinical significance. The genes to be discussed include nuclear proteins (Notch3, HBXAP [Rsf-1], NAC1 and NFkappaB), cytoplasmic proteins (fatty acid synthase and apolipoprotein E) and cell surface/secretory proteins (mucin-4, mesothelin, claudin, HLA-G, kallikrein and folate receptor and osteopontin). Since the study of ovarian cancer-associated genes is complicated by several factors unique to ovarian cancer, we will also present our views on the limitations and challenges of current ovarian cancer research.
Collapse
Affiliation(s)
- Ie-Ming Shih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21212, USA.
| | | |
Collapse
|
244
|
Chang ST, Zahn JM, Horecka J, Kunz PL, Ford JM, Fisher GA, Le QT, Chang DT, Ji H, Koong AC. Identification of a biomarker panel using a multiplex proximity ligation assay improves accuracy of pancreatic cancer diagnosis. J Transl Med 2009; 7:105. [PMID: 20003342 PMCID: PMC2796647 DOI: 10.1186/1479-5876-7-105] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 12/11/2009] [Indexed: 02/08/2023] Open
Abstract
Background Pancreatic cancer continues to prove difficult to clinically diagnose. Multiple simultaneous measurements of plasma biomarkers can increase sensitivity and selectivity of diagnosis. Proximity ligation assay (PLA) is a highly sensitive technique for multiplex detection of biomarkers in plasma with little or no interfering background signal. Methods We examined the plasma levels of 21 biomarkers in a clinically defined cohort of 52 locally advanced (Stage II/III) pancreatic ductal adenocarcinoma cases and 43 age-matched controls using a multiplex proximity ligation assay. The optimal biomarker panel for diagnosis was computed using a combination of the PAM algorithm and logistic regression modeling. Biomarkers that were significantly prognostic for survival in combination were determined using univariate and multivariate Cox survival models. Results Three markers, CA19-9, OPN and CHI3L1, measured in multiplex were found to have superior sensitivity for pancreatic cancer vs. CA19-9 alone (93% vs. 80%). In addition, we identified two markers, CEA and CA125, that when measured simultaneously have prognostic significance for survival for this clinical stage of pancreatic cancer (p < 0.003). Conclusions A multiplex panel assaying CA19-9, OPN and CHI3L1 in plasma improves accuracy of pancreatic cancer diagnosis. A panel assaying CEA and CA125 in plasma can predict survival for this clinical cohort of pancreatic cancer patients.
Collapse
Affiliation(s)
- Stephanie T Chang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Jepson MA, Davis MJ, Horton AA, Walker DG. Histochemical and biochemical observations on the cytotoxicity of paracetamol and its effects on glycogen metabolism in rat liver. Toxicology 1988; 54:926-34. [PMID: 3424388 DOI: 10.1002/mc.22153] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 01/20/2014] [Accepted: 03/06/2014] [Indexed: 12/14/2022]
Abstract
The effects of paracetamol overdose on glycogen metabolism in rat liver have been investigated and related to its cytotoxicity. Paracetamol was administered to male rats by gavaging after a 24-h fast and refeeding was not permitted. An early (9-12-h) increase in histochemically demonstrable glycogen phosphorylase alpha activity in perivenous hepatocytes preceded major loss of membrane integrity as assessed by serum glutamate-pyruvate transaminase (SGPT) activity and uptake of trypan blue during perfusion. These changes occurred only after a decrease in the concentration of reduced glutathione, which is generally observed about 4 h after paracetamol treatment. The activation of glycogen phosphorylase in perivenous hepatocytes occurred concurrently with an increase in glycogen content of periportal hepatocytes, indicating a clear heterogeneity in the response of the two-cell populations to the hepatotoxin. The use of trypan blue perfusion together with histochemical techniques allowed changes in glycogen content and phosphorylase alpha activity of individual hepatocytes to be assessed with reference to the extent of membrane damage evident. The relevance of the results to possible mechanisms of hepatotoxicity is discussed.
Collapse
Affiliation(s)
- M A Jepson
- Department of Biochemistry, University of Birmingham, U.K
| | | | | | | |
Collapse
|