20 results on '"cafs"'
Search Results
2. Urokinase plasminogen activator secreted by cancer-associated fibroblasts induces tumor progression via PI3K/AKT and ERK signaling in esophageal squamous cell carcinoma
- Author
-
Hai-long Wang, An Hong, Wei Han, Xuan Tan, Xiao-yan Li, Li Fu, Xiaojia Chen, Changjun Nie, Yan Li, Baoqing Tian, Huihua Zhang, Jiakang Wang, Ying Zhao, Xin Yuan Guan, and Li Yi Zhang
- Subjects
0301 basic medicine ,Male ,Esophageal Neoplasms ,Gene Expression ,Kaplan-Meier Estimate ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Medicine ,uPA ,Extracellular Signal-Regulated MAP Kinases ,CAFs ,Antibodies, Monoclonal ,Middle Aged ,Prognosis ,ERK ,Oncology ,030220 oncology & carcinogenesis ,Carcinoma, Squamous Cell ,Disease Progression ,Immunohistochemistry ,Female ,Esophageal Squamous Cell Carcinoma ,Research Paper ,Signal Transduction ,Adult ,Stromal cell ,03 medical and health sciences ,Stroma ,Cell Line, Tumor ,Animals ,Humans ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Aged ,Cell Proliferation ,Neoplasm Staging ,PI3K/AKT ,Cell growth ,business.industry ,ESCC ,Fibroblasts ,Urokinase-Type Plasminogen Activator ,digestive system diseases ,030104 developmental biology ,Tumor progression ,Immunology ,Cancer research ,Stromal Cells ,business ,Proto-Oncogene Proteins c-akt - Abstract
Cancer-associated fibroblasts (CAFs) are believed to influence tumor behavior and clinical outcomes. We previously showed that conditioned medium (CM) from CAFs induces proliferation and motility of esophageal squamous cell carcinoma (ESCC) cells. Here, we investigated the molecular mechanisms by which the CAF-secreted proteins induce ESCC development and progression. Using antibody arrays, we identified urokinase plasminogen activator (uPA) as one of the main proteins whose release was increased in CAFs compared to normal fibroblasts (NFs). Immunohistochemical analysis of pathological sections showed that uPA-positive cells were localized at the boundaries of tumor and stroma tissues, in stroma between tumor nests, and within the tumors. Increased stromal uPA levels (132/146 cases) correlated with tumor invasion (p < 0.05) and overall survival of ESCC patients (p < 0.05). In vitro assays showed that uPA promotes ESCC cell proliferation, migration, and invasion via PI3K/AKT and ERK signaling pathways. In vivo, anti-uPA antibody suppressed tumor growth in ESCC xenografts. These results suggest that uPA released from stroma, and especially from CAFs, might be a predictive marker for ESCC diagnosis and prognosis, as well as an effective therapeutic target.
- Published
- 2017
3. miR-31 affects colorectal cancer cells by inhibiting autophagy in cancer-associated fibroblasts
- Author
-
Ming Li, Gang Zhou, Zhenyu Ye, Yong Wu, Yongyou Wu, Jianping Cao, Chungen Xing, Xiaohui Xu, Junjia Zhu, Hong Zhu, Kui Zhao, Xiaodong Yang, and Wei Peng
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,autophagy ,Colorectal cancer ,Cell ,Autophagy-Related Proteins ,colorectal cancer ,Apoptosis ,Cell Communication ,biological behaviors ,Radiation Tolerance ,03 medical and health sciences ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Cell Movement ,Internal medicine ,microRNA ,medicine ,Tumor Microenvironment ,Humans ,Neoplasm Invasiveness ,miR-31 ,Inhibitory effect ,Cell Proliferation ,business.industry ,CAFs ,Autophagy ,Middle Aged ,medicine.disease ,HCT116 Cells ,Coculture Techniques ,Surgery ,mir-31 ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business ,Colorectal Neoplasms ,Research Paper ,Signal Transduction - Abstract
// Xiaodong Yang 1, * , Xiaohui Xu 1, 2, * , Junjia Zhu 1, * , Shuyu Zhang 3 , Yong Wu 1 , Yongyou Wu 1 , Kui Zhao 1 , Chungen Xing 1 , Jianping Cao 3 , Hong Zhu 4 , Ming Li 3 , Zhenyu Ye 1 , Wei Peng 1 1 Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China 2 Department of General Surgery, The First People’s Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Suzhou 215400, China 3 School of Radiation Medicine and Protection, Medical College of Soochow University, Suzhou 215123, China 4 Oncology Department, The First Affiliated Hospital of Soochow University, Suzhou 215006, China * These authors have contributed equally to this work Correspondence to: Chungen Xing, email: xingcg@126.com Jianping Cao, email: jpcao@suda.edu.cn Keywords: miR-31, colorectal cancer, CAFs, autophagy, biological behaviors Received: October 06, 2015 Accepted: September 25, 2016 Published: October 25, 2016 ABSTRACT Autophagy is a double-edged sword in tumor development. Recent studies have found that miRNAs have an inhibitory effect on the regulation of autophagy. It has been reported that miR-31 plays an important role in the development of colorectal cancer. However, what role miR-31 plays in colorectal cancer-associated fibroblasts (CAFs) has not been determined. In this study, we confirmed that the expression of miR-31 in CAFs was higher than in normal colorectal fibroblasts (NFs). We also found that treatment of CAFs with miR-31 mimic inhibited the expression of the autophagy-related genes Beclin-1, ATG, DRAM and LC3. In addition, we found up-regulation of miR-31 significantly affected colorectal cancer cell behaviors, including proliferation, invasion and apoptosis. Also, up-regulation of miR-31 in CAF could increase the radiosensitivity of colorectal cancer cells co-cultured with CAF. In summary, miR-31 can inhibit autophagy in colorectal CAFs, affect colorectal cancer development, and increase the radiosensitivity of colorectal cancer cells co-cultured with CAF. We hypothesize that miR-31 may become a new target of treatments for colorectal cancer.
- Published
- 2016
4. PDCD4 is a CSL associated protein with a transcription repressive function in cancer associated fibroblast activation
- Author
-
Seung-Hee Jo, Andrea Clocchiatti, G. Paolo Dotto, and Dong-Eun Kim
- Subjects
0301 basic medicine ,Skin Neoplasms ,Stromal cell ,Transcription, Genetic ,Notch signaling pathway ,Down-Regulation ,Mice, SCID ,Biology ,medicine.disease_cause ,squamous cancer ,Dermal fibroblast ,Mice ,03 medical and health sciences ,Animals ,Apoptosis Regulatory Proteins/genetics ,Apoptosis Regulatory Proteins/metabolism ,Cancer-Associated Fibroblasts/immunology ,Cancer-Associated Fibroblasts/metabolism ,Carcinoma, Squamous Cell/genetics ,Carcinoma, Squamous Cell/metabolism ,Cell Line, Tumor ,Cellular Senescence ,HEK293 Cells ,HeLa Cells ,Humans ,Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism ,Keratosis, Actinic/genetics ,Keratosis, Actinic/metabolism ,Protein Binding ,RNA, Small Interfering/genetics ,RNA-Binding Proteins/genetics ,RNA-Binding Proteins/metabolism ,Signal Transduction ,Skin Neoplasms/genetics ,Skin Neoplasms/metabolism ,Xenograft Model Antitumor Assays ,CAFs ,Notch/CSL signaling ,PDCD4 ,transcription repression ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Transcription (biology) ,medicine ,RNA, Small Interfering ,Fibroblast ,Effector ,RNA-Binding Proteins ,3. Good health ,Keratosis, Actinic ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Immunoglobulin J Recombination Signal Sequence-Binding Protein ,030220 oncology & carcinogenesis ,Immunology ,Carcinoma, Squamous Cell ,Cancer research ,Signal transduction ,Apoptosis Regulatory Proteins ,Carcinogenesis ,Priority Research Paper - Abstract
// Seung-Hee Jo 1,2 , Dong Eun Kim 3 , Andrea Clocchiatti 1,2 and G. Paolo Dotto 1,3 1 Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA, USA 2 Department of Dermatology, Harvard Medical School, Boston, MA, USA 3 Department of Biochemistry, University of Lausanne, Epalinges, CH, Switzerland Correspondence to: G. Paolo Dotto, email: // Keywords : PDCD4, Notch/CSL signaling, transcription repression, squamous cancer, CAFs Received : May 12, 2016 Accepted : July 22, 2016 Published : August 11, 2016 Abstract The Notch/CSL pathway plays an important role in skin homeostasis and carcinogenesis. CSL, the key effector of canonical Notch signaling endowed with an intrinsic transcription repressive function, suppresses stromal fibroblast senescence and Cancer Associated Fibroblast (CAF) activation through direct down-modulation of key effector genes. Interacting proteins that participate with CSL in this context are as yet to be identified. We report here that Programmed Cell Death 4 (PDCD4), a nuclear/cytoplasmic shuttling protein with multiple functions, associates with CSL and plays a similar role in suppressing dermal fibroblast senescence and CAF activation. Like CSL, PDCD4 is down-regulated in stromal fibroblasts of premalignant skin actinic keratosis (AKs) lesions and squamous cell carcinoma (SCC). While devoid of intrinsic DNA binding capability, PDCD4 is present at CSL binding sites of CAF marker genes as well as canonical Notch/CSL targets and suppresses expression of these genes in a fibroblast-specific manner. Thus, we propose that PDCD4 is part of the CSL repressive complex involved in negative control of stromal fibroblasts conversion into CAFs.
- Published
- 2016
- Full Text
- View/download PDF
5. MARCKS contributes to stromal cancer-associated fibroblast activation and facilitates ovarian cancer metastasis
- Author
-
Dan Liu, Xin Yang, Qinglei Gao, Sen Xu, Pingbo Chen, Ding Ma, Gang Chen, Ping Jin, Dongyi Wan, Xiao Wei, Li Meng, Xiaoting Li, Sixiang Long, Taoran Zhang, Yong Fang, and Zongyuan Yang
- Subjects
0301 basic medicine ,Senescence ,MARCKS ,Stromal cell ,senescence ,endocrine system diseases ,Regulator ,Bioinformatics ,Disease-Free Survival ,Metastasis ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer-Associated Fibroblasts ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Metastasis ,Myristoylated Alanine-Rich C Kinase Substrate ,Protein kinase B ,Ovarian Neoplasms ,business.industry ,CAFs ,Mesenchymal stem cell ,Cell Differentiation ,medicine.disease ,030104 developmental biology ,ovarian cancer ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Female ,business ,Ovarian cancer ,Research Paper ,Twist1 ,Signal Transduction - Abstract
// Zongyuan Yang 1, * , Sen Xu 1, * , Ping Jin 1 , Xin Yang 1 , Xiaoting Li 1 , Dongyi Wan 1 , Taoran Zhang 1 , Sixiang Long 1 , Xiao Wei 1 , Gang Chen 1 , Li Meng 1 , Dan Liu 1 , Yong Fang 1 , Pingbo Chen 1 , Ding Ma 1 , Qinglei Gao 1 1 Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China * These authors have contributed equally to this work Correspondence to: Ding Ma, email: dma@tjh.tjmu.edu.cn Qinglei Gao, email: qlgao@tjh.tjmu.edu.cn Keywords: ovarian cancer, CAFs, MARCKS, senescence, Twist1 Received: January 07, 2016 Accepted: March 28, 2016 Published: April 13, 2016 ABSTRACT The Cancer Genome Atlas network has revealed that the ‘mesenchymal’ epithelial ovarian cancer (EOC) subtype represents the poorest outcome, indicating a crucial role of stromal cancer-associated fibroblasts (CAFs) in disease progression. The cooperative role of CAFs in EOC metastasis has long been recognized, but the mechanisms of stromal CAFs activation are still obscure. Therefore, we carried out an integrative analysis to identify the regulator genes that are responsible for CAFs activation in microdissected tumor stroma profiles. Here, we determined that myristoylated alanine-rich C-kinase substrate (MARCKS) was highly expressed in ovarian stroma, and was required for the differentiation and tumor promoting function of CAFs. Suppression of MARCKS resulted in the loss of CAF features, and diminished role of CAFs in supporting tumor cell growth in 3D organotypic cultures and in murine xenograft model. Mechanistically, we found that MARCKS maintained CAF activation through suppression of cellular senescence and activation of the AKT/Twist1 signaling. Moreover, high MARCKS expression was associated with poor patient survival in EOC. Collectively, our findings identify the potential of MARCKS inhibition as a novel stroma-oriented therapy in EOC.
- Published
- 2016
6. Targeting chemotherapy-induced PTX3 in tumor stroma to prevent the progression of drug-resistant cancers
- Author
-
Jhih-Ying Chi, Shao Ming Wang, Yu Wei Hsiao, Ben Kuen Chen, Kelvin K C Tsai, Ju Ming Wang, Yu Chih Lo, Yang Ming Liu, Xiu Han, Chien-Feng Li, Zu Yau Lin, and Jhen Yi Hong
- Subjects
CCAAT-Enhancer-Binding Protein-delta ,Apoptosis ,medicine.disease_cause ,Metastasis ,Mice ,Cell Movement ,Neoplasms ,Enhancer binding ,Tumor Microenvironment ,Neoplasm Metastasis ,Myofibroblasts ,Mice, Inbred BALB C ,Reverse Transcriptase Polymerase Chain Reaction ,CAFs ,Gene Expression Regulation, Neoplastic ,Serum Amyloid P-Component ,C-Reactive Protein ,Cell Transformation, Neoplastic ,Oncology ,Disease Progression ,Female ,Fluorouracil ,Research Paper ,medicine.drug ,Cell Survival ,TAMs ,Antineoplastic Agents ,Biology ,In vivo ,Cell Line, Tumor ,CEBPD ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,PTX3 ,Cisplatin ,Tumor microenvironment ,Macrophages ,Cancer ,Fibroblasts ,medicine.disease ,Peptide Fragments ,Drug Resistance, Neoplasm ,Culture Media, Conditioned ,Cancer cell ,Cancer research ,Carcinogenesis ,Neoplasm Transplantation - Abstract
The tumor microenvironment has been suggested to participate in tumorigenesis, but the nature of the communication between cancer cells and the microenvironment, especially in response to anticancer drugs, remains obscure. We determined that activation of the CCAAT/enhancer binding protein delta (CEBPD) response to Cisplatin and 5-Fluorouracil in cancer-associated macrophages and fibroblasts contributed to the metastasis, invasion, acquired chemoresistance and stemness of cancer cells by in vitro and in vivo assays. Specifically, reporter and in vivo DNA binding assays were used to determine that Pentraxin 3 (PTX3) is a CEBPD responsive gene and serves a protumor role upon anticancer drug treatment. Finally, a PTX3 peptide inhibitor RI37 was developed and assessed the antitumor effects by in vivo assays. RI37 could function as a promising inhibitor for preventing cancer progression and the metastasis, invasion and progression of drug-resistant cancers. The identification of PTX3 provided a new insight in the interaction between host and tumor and the RI37 peptide showed a great opportunity to largely reduce the risk of invasion and metastasis of cancer and drug-resistant cancers.
- Published
- 2015
- Full Text
- View/download PDF
7. Estrogenic gper signaling regulates mir144 expression in cancer cells and cancer-associated fibroblasts (cafs)
- Author
-
Michele Pellegrino, Paola De Marco, Francesca Cirillo, Marcello Maggiolini, Adele Vivacqua, Sergio Abonante, Maria Francesca Santolla, and Maria Luisa Panno
- Subjects
Mice, Nude ,Estrogen receptor ,Biology ,Receptors, G-Protein-Coupled ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Line, Tumor ,Neoplasms ,Tumor Microenvironment ,medicine ,Animals ,Humans ,cancer ,Extracellular Signal-Regulated MAP Kinases ,PI3K/AKT/mTOR pathway ,ets-Domain Protein Elk-1 ,Tumor microenvironment ,Estradiol ,CAFs ,Estrogen Receptor alpha ,Cancer ,Hep G2 Cells ,Fibroblasts ,GPER ,medicine.disease ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,Receptors, Estrogen ,Oncology ,Core Binding Factor Alpha 2 Subunit ,miRNAs ,Cancer cell ,MCF-7 Cells ,Cancer research ,Heterografts ,Female ,Signal transduction ,Estrogen receptor alpha ,Neoplasm Transplantation ,Signal Transduction ,Research Paper ,estrogens - Abstract
MicroRNAs (miRNAs) are small non coding RNA molecules that play a crucial role in several pathophysiological conditions, including cancer. The stimulation of hormone-sensitive tumors by estrogens are mediated by estrogen receptor (ER)α and G protein estrogen receptor (GPER). Previous studies have reported that ERα regulates miRNA expression, while this ability of GPER remains to be elucidated. Here, we demonstrate that in SkBr3 breast cancer and HepG2 hepatocarcinoma cells, 17β-estradiol (E2) and the selective GPER ligand G-1 induce miR144 expression through GPER and the involvement of the PI3K/ERK1/2/Elk1 transduction pathway. Moreover, we show that E2 and G-1 down-regulate through miR144 the onco-suppressor Runx1 and increase cell cycle progression. The capability of E2 and G-1 in triggering the induction of miR144 and the down-regulation of Runx1 was also confirmed in cancer-associated fibroblasts (CAFs) that are main components of the tumor microenvironment driving cancer progression. Further confirming these results, Runx1 protein levels were found decreased in tumor xenografts upon G-1 treatment. On the basis of our findings miR144 and Runx1 may be included among the oncotargets of GPER action. Moreover, the present data provide new insights regarding the ability of estrogens to trigger the GPER/miR144/Runx1 transduction pathway toward the stimulation of cancer progression.
- Published
- 2015
- Full Text
- View/download PDF
8. TIMP-1 activated carcinoma-associated fibroblasts inhibit tumor apoptosis by activating SDF1/CXCR4 signaling in hepatocellular carcinoma
- Author
-
Xin Zheng, Kangsheng Tu, Changwei Dou, Yuli Jia, and Tao Song
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Receptors, CXCR4 ,Carcinoma, Hepatocellular ,Mice, Nude ,Matrix metalloproteinase ,Transfection ,Mice ,TIMP-1 ,Medicine ,tumor microenvironment ,Animals ,Humans ,HCC ,Protein kinase B ,PI3K/AKT/mTOR pathway ,TIMP1 ,Cell Proliferation ,Tumor microenvironment ,Tissue Inhibitor of Metalloproteinase-1 ,business.industry ,CAFs ,Liver Neoplasms ,apoptosis ,Tissue inhibitor of metalloproteinase ,Fibroblasts ,digestive system diseases ,Oncology ,Apoptosis ,Cancer research ,Female ,Signal transduction ,business ,Research Paper ,Signal Transduction - Abstract
Tissue inhibitor of metalloproteinase 1 (TIMP-1) is an endogenous inhibitor for MMPs that regulates the remodeling and turnover of the ECM during normal development and pathological conditions. Intriguingly, recent studies have shown that TIMP-1 plays a dual role in cancer progression. In this study, we found that TIMP-1 expression in HCC tissues is associated with advanced TNM stage, intrahepatic metastasis, portal vein invasion, and vasculature invasion. Notably, TIMP-1 expression in HCC tissue is significantly related to worse overall survival for patients with HCC after liver resection. Ectopic TIMP1 expression promoted the growth of HCC xenografts in nude mice. Both co-culture with Huh7 cells with a high level of TIMP-1 and TIMP1 treatment resulted in up-regulation of hallmarks of carcinoma-associated fibroblasts (CAFs) and accelerated cell proliferation, migration and invasion in immortalized liver fibroblasts (LFs) isolated from human normal liver tissue. By co-culture with CAFs, SDF-1/CXCR4/PI3K/AKT signaling was activated and apoptosis was markedly repressed with an increased Bcl-2/BAX ratio in Huh7 cells. Taken together, our observations suggest that TIMP-1 induces the trans-differentiation of LFs into CAFs, suppresses apoptosis via SDF-1/CXCR4/PI3K/AKT signaling and then promotes HCC progression. This protein may be a potential prognostic biomarker and therapeutic target for HCC.
- Published
- 2015
9. Cyclooxygenase-2 blockade can improve efficacy of VEGF-targeting drugs
- Author
-
Melanie Janning, Mark Wroblewski, Stefanie Sawall, Lin Zhao, Victoria Gensch, Boris Fehse, Miguel Cubas-Cordova, Kristoffer Riecken, Isabel Ben-Batalla, Florian Udonta, Klaus Pantel, Sonja Loges, Inigo Martinez-Zubiaurre, Jonas S. Waizenegger, and Carsten Bokemeyer
- Subjects
Vascular Endothelial Growth Factor A ,Oncology ,medicine.medical_specialty ,Basic fibroblast growth factor ,Pharmacology ,Mice ,Random Allocation ,chemistry.chemical_compound ,breast cancer ,Breast cancer ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Molecular Targeted Therapy ,Mice, Inbred BALB C ,Hematology ,Cyclooxygenase 2 Inhibitors ,Neovascularization, Pathologic ,Sunitinib ,business.industry ,CAFs ,Mammary Neoplasms, Experimental ,Cancer ,medicine.disease ,Blockade ,Transplantation ,Vascular endothelial growth factor A ,chemistry ,Cyclooxygenase 2 ,Female ,Cox-2 ,business ,anti-angiogenic therapies ,Signal Transduction ,Research Paper ,medicine.drug - Abstract
// Isabel Ben-Batalla 1,2 , Miguel Cubas-Cordova 1,2 , Florian Udonta 1,2 , Mark Wroblewski 1,2 , Jonas S. Waizenegger 1,2 , Melanie Janning 1,2 , Stefanie Sawall 1,2 , Victoria Gensch 1,2 , Lin Zhao 1,2 , Inigo Martinez-Zubiaurre 3 , Kristoffer Riecken 4 , Boris Fehse 4 , Klaus Pantel 2 , Carsten Bokemeyer 1 and Sonja Loges 1,2 1 Department of Hematology and Oncology, BMT with Section of Pneumology, Hubertus Wald Tumorzentrum, University Comprehensive Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 2 Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany 3 Department of Clinical Medicine, The Arctic University of Norway, Tromso, Norway 4 Research Department Cell and Gene Therapy, Clinic for Stem Cell Transplantation, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany Correspondence to: Sonja Loges, email: // Keywords : breast cancer, anti-angiogenic therapies, Cox-2, CAFs Received : September 28, 2014 Accepted : January 21, 2015 Published : January 31, 2015 Abstract Anti-angiogenic therapies were approved for different cancers. However, significant primary and secondary resistance hampers efficacy in several tumor types including breast cancer. Thus, we need to develop clinically applicable strategies to enhance efficacy of anti-angiogenic drugs. We report that anti-angiogenic therapies can induce upregulation of cyclooxygenase-2 (Cox-2) and of its product prostaglandin E2 (PGE 2 ) in breast cancer models. Upon Cox-2 inhibition PGE 2 levels were normalized and efficacy of anti-vascular endothelial growth factor receptor 2 (anti-VEGFR-2) antibodies and sunitinib was enhanced. Interestingly, both treatments exerted additive anti-angiogenic effects. Following Cox-2 inhibition, we observed reduced infiltration of tumors with cancer-associated fibroblasts (CAFs) and lower levels of pro-angiogenic factors active besides the VEGF axis including hepatocyte growth factor (HGF) and basic fibroblast growth factor (FGF2). Mechanistic studies indicated that Cox-2 inhibition reduced PGE 2 -induced migration and proliferation of CAFs via inhibiting phosphorylation of Akt. Hence, Cox-2 inhibition can increase efficacy of anti-angiogenic treatments and our findings might pave the road for clinical investigations of concomitant blockade of Cox-2 and VEGF-signaling.
- Published
- 2015
- Full Text
- View/download PDF
10. Leptin as a mediator of tumor-stromal interactions promotes breast cancer stem cell activity
- Author
-
Ines Barone, Marilena Lanzino, Balázs Győrffy, Salvatore Panza, Bruno M Simões, Sebastiano Andò, Angela Cordella, Francesca Chemi, Adnan Hashim, Stefania Catalano, Daniela Bonofiglio, Robert Clarke, Cinzia Giordano, Antonella Leggio, Alessandro Weisz, Antonella Campana, and Pietro Rizza
- Subjects
Leptin ,breast cancer stem cells ,medicine.medical_specialty ,Cell signaling ,Stromal cell ,Microarrays ,Blotting, Western ,Breast Neoplasms ,Cell Communication ,Kaplan-Meier Estimate ,Metastasis ,Breast cancer ,breast cancer ,Internal medicine ,Cell Line, Tumor ,Spheroids, Cellular ,medicine ,Tumor Microenvironment ,Humans ,HSP90 Heat-Shock Proteins ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis ,Tumor microenvironment ,Leptin receptor ,business.industry ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,CAFs ,Fibroblasts ,medicine.disease ,microenvironment ,Gene Expression Regulation, Neoplastic ,Endocrinology ,Oncology ,Breast Cancer Stem Cells ,Culture Media, Conditioned ,MCF-7 Cells ,Neoplastic Stem Cells ,Receptors, Leptin ,Stem cell ,Stromal Cells ,business ,Research Paper - Abstract
Breast cancer stem cells (BCSCs) play crucial roles in tumor initiation, metastasis and therapeutic resistance. A strict dependency between BCSCs and stromal cell components of tumor microenvironment exists. Thus, novel therapeutic strategies aimed to target the crosstalk between activated microenvironment and BCSCs have the potential to improve clinical outcome. Here, we investigated how leptin, as a mediator of tumor-stromal interactions, may affect BCSC activity using patient-derived samples (n = 16) and breast cancer cell lines, and determined the potential benefit of targeting leptin signaling in these model systems. Conditioned media (CM) from cancer-associated fibroblasts and breast adipocytes significantly increased mammosphere formation in breast cancer cells and depletion of leptin from CM completely abrogated this effect. Mammosphere cultures exhibited increased leptin receptor (OBR) expression and leptin exposure enhanced mammosphere formation. Microarray analyses revealed a similar expression profile of genes involved in stem cell biology among mammospheres treated with CM and leptin. Interestingly, leptin increased mammosphere formation in metastatic breast cancers and expression of OBR as well as HSP90, a target of leptin signaling, were directly correlated with mammosphere formation in metastatic samples (r = 0.68/p = 0.05; r = 0.71/p = 0.036, respectively). Kaplan-Meier survival curves indicated that OBR and HSP90 expression were associated with reduced overall survival in breast cancer patients (HR = 1.9/p = 0.022; HR = 2.2/p = 0.00017, respectively). Furthermore, blocking leptin signaling by using a full leptin receptor antagonist significantly reduced mammosphere formation in breast cancer cell lines and patient-derived samples. Our results suggest that leptin/leptin receptor signaling may represent a potential therapeutic target that can block the stromal-tumor interactions driving BCSC-mediated disease progression.
- Published
- 2015
11. Stromal cyclin D1 promotes heterotypic immune signaling and breast cancer growth.
- Author
-
Pestell TG, Jiao X, Kumar M, Peck AR, Prisco M, Deng S, Li Z, Ertel A, Casimiro MC, Ju X, Di Rocco A, Di Sante G, Katiyar S, Shupp A, Lisanti MP, Jain P, Wu K, Rui H, Hooper DC, Yu Z, Goldman AR, Speicher DW, Laury-Kleintop L, and Pestell RG
- Abstract
The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that drives cell autonomous cell cycle progression and proliferation. Herein we show cyclin D1 abundance is increased >30-fold in the stromal fibroblasts of patients with invasive breast cancer, associated with poor outcome. Cyclin D1 transformed hTERT human fibroblast to a cancer-associated fibroblast phenotype. Stromal fibroblast expression of cyclin D1 (cyclin D1
Stroma ) in vivo , enhanced breast epithelial cancer tumor growth, restrained apoptosis, and increased autophagy. Cyclin D1Stroma had profound effects on the breast tumor microenvironment increasing the recruitment of F4/80+ and CD11b+ macrophages and increasing angiogenesis. Cyclin D1Stroma induced secretion of factors that promoted expansion of stem cells (breast stem-like cells, embryonic stem cells and bone marrow derived stem cells). Cyclin D1Stroma resulted in increased secretion of proinflammatory cytokines (CCL2, CCL7, CCL11, CXCL1, CXCL5, CXCL9, CXCL12), CSF (CSF1, GM-CSF1) and osteopontin (OPN) (30-fold). OPN was induced by cyclin D1 in fibroblasts, breast epithelial cells and in the murine transgenic mammary gland and OPN was sufficient to induce stem cell expansion. These results demonstrate that cyclin D1Stroma drives tumor microenvironment heterocellular signaling, promoting several key hallmarks of cancer., Competing Interests: CONFLICTS OF INTEREST The authors declare that they have no conflict of interest.- Published
- 2017
- Full Text
- View/download PDF
12. Urokinase plasminogen activator secreted by cancer-associated fibroblasts induces tumor progression via PI3K/AKT and ERK signaling in esophageal squamous cell carcinoma.
- Author
-
Tian B, Chen X, Zhang H, Li X, Wang J, Han W, Zhang LY, Fu L, Li Y, Nie C, Zhao Y, Tan X, Wang H, Guan XY, and Hong A
- Subjects
- Adult, Aged, Animals, Antibodies, Monoclonal pharmacology, Carcinoma, Squamous Cell genetics, Carcinoma, Squamous Cell mortality, Carcinoma, Squamous Cell pathology, Cell Line, Tumor, Cell Proliferation, Disease Progression, Esophageal Neoplasms genetics, Esophageal Neoplasms mortality, Esophageal Neoplasms pathology, Esophageal Squamous Cell Carcinoma, Female, Fibroblasts metabolism, Gene Expression, Humans, Kaplan-Meier Estimate, Male, Mice, Middle Aged, Neoplasm Staging, Prognosis, Stromal Cells metabolism, Urokinase-Type Plasminogen Activator antagonists & inhibitors, Urokinase-Type Plasminogen Activator genetics, Cancer-Associated Fibroblasts metabolism, Carcinoma, Squamous Cell metabolism, Esophageal Neoplasms metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Urokinase-Type Plasminogen Activator metabolism
- Abstract
Cancer-associated fibroblasts (CAFs) are believed to influence tumor behavior and clinical outcomes. We previously showed that conditioned medium (CM) from CAFs induces proliferation and motility of esophageal squamous cell carcinoma (ESCC) cells. Here, we investigated the molecular mechanisms by which the CAF-secreted proteins induce ESCC development and progression. Using antibody arrays, we identified urokinase plasminogen activator (uPA) as one of the main proteins whose release was increased in CAFs compared to normal fibroblasts (NFs). Immunohistochemical analysis of pathological sections showed that uPA-positive cells were localized at the boundaries of tumor and stroma tissues, in stroma between tumor nests, and within the tumors. Increased stromal uPA levels (132/146 cases) correlated with tumor invasion (p < 0.05) and overall survival of ESCC patients (p < 0.05). In vitro assays showed that uPA promotes ESCC cell proliferation, migration, and invasion via PI3K/AKT and ERK signaling pathways. In vivo, anti-uPA antibody suppressed tumor growth in ESCC xenografts. These results suggest that uPA released from stroma, and especially from CAFs, might be a predictive marker for ESCC diagnosis and prognosis, as well as an effective therapeutic target.
- Published
- 2017
- Full Text
- View/download PDF
13. miR-31 affects colorectal cancer cells by inhibiting autophagy in cancer-associated fibroblasts.
- Author
-
Yang X, Xu X, Zhu J, Zhang S, Wu Y, Wu Y, Zhao K, Xing C, Cao J, Zhu H, Li M, Ye Z, and Peng W
- Subjects
- Apoptosis, Autophagy-Related Proteins genetics, Autophagy-Related Proteins metabolism, Cancer-Associated Fibroblasts pathology, Cell Movement, Cell Proliferation, Coculture Techniques, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms radiotherapy, Gene Expression Regulation, Neoplastic, HCT116 Cells, Humans, MicroRNAs genetics, Middle Aged, Neoplasm Invasiveness, Radiation Tolerance, Signal Transduction, Tumor Microenvironment, Autophagy, Cancer-Associated Fibroblasts metabolism, Cell Communication, Colorectal Neoplasms metabolism, MicroRNAs metabolism
- Abstract
Autophagy is a double-edged sword in tumor development. Recent studies have found that miRNAs have an inhibitory effect on the regulation of autophagy. It has been reported that miR-31 plays an important role in the development of colorectal cancer. However, what role miR-31 plays in colorectal cancer-associated fibroblasts (CAFs) has not been determined. In this study, we confirmed that the expression of miR-31 in CAFs was higher than in normal colorectal fibroblasts (NFs). We also found that treatment of CAFs with miR-31 mimic inhibited the expression of the autophagy-related genes Beclin-1, ATG, DRAM and LC3. In addition, we found up-regulation of miR-31 significantly affected colorectal cancer cell behaviors, including proliferation, invasion and apoptosis. Also, up-regulation of miR-31 in CAF could increase the radiosensitivity of colorectal cancer cells co-cultured with CAF. In summary, miR-31 can inhibit autophagy in colorectal CAFs, affect colorectal cancer development, and increase the radiosensitivity of colorectal cancer cells co-cultured with CAF. We hypothesize that miR-31 may become a new target of treatments for colorectal cancer.
- Published
- 2016
- Full Text
- View/download PDF
14. PDCD4 is a CSL associated protein with a transcription repressive function in cancer associated fibroblast activation.
- Author
-
Jo SH, Kim DE, Clocchiatti A, and Dotto GP
- Subjects
- Animals, Apoptosis Regulatory Proteins genetics, Cancer-Associated Fibroblasts metabolism, Carcinoma, Squamous Cell genetics, Cell Line, Tumor, Cellular Senescence, Down-Regulation, HEK293 Cells, HeLa Cells, Humans, Keratosis, Actinic genetics, Mice, Mice, SCID, Protein Binding, RNA, Small Interfering genetics, RNA-Binding Proteins genetics, Signal Transduction, Skin Neoplasms genetics, Transcription, Genetic, Xenograft Model Antitumor Assays, Apoptosis Regulatory Proteins metabolism, Cancer-Associated Fibroblasts immunology, Carcinoma, Squamous Cell metabolism, Immunoglobulin J Recombination Signal Sequence-Binding Protein metabolism, Keratosis, Actinic metabolism, RNA-Binding Proteins metabolism, Skin Neoplasms metabolism
- Abstract
The Notch/CSL pathway plays an important role in skin homeostasis and carcinogenesis. CSL, the key effector of canonical Notch signaling endowed with an intrinsic transcription repressive function, suppresses stromal fibroblast senescence and Cancer Associated Fibroblast (CAF) activation through direct down-modulation of key effector genes. Interacting proteins that participate with CSL in this context are as yet to be identified. We report here that Programmed Cell Death 4 (PDCD4), a nuclear/cytoplasmic shuttling protein with multiple functions, associates with CSL and plays a similar role in suppressing dermal fibroblast senescence and CAF activation. Like CSL, PDCD4 is down-regulated in stromal fibroblasts of premalignant skin actinic keratosis (AKs) lesions and squamous cell carcinoma (SCC). While devoid of intrinsic DNA binding capability, PDCD4 is present at CSL binding sites of CAF marker genes as well as canonical Notch/CSL targets and suppresses expression of these genes in a fibroblast-specific manner. Thus, we propose that PDCD4 is part of the CSL repressive complex involved in negative control of stromal fibroblasts conversion into CAFs.
- Published
- 2016
- Full Text
- View/download PDF
15. MARCKS contributes to stromal cancer-associated fibroblast activation and facilitates ovarian cancer metastasis.
- Author
-
Yang Z, Xu S, Jin P, Yang X, Li X, Wan D, Zhang T, Long S, Wei X, Chen G, Meng L, Liu D, Fang Y, Chen P, Ma D, and Gao Q
- Subjects
- Animals, Cell Differentiation physiology, Cell Line, Tumor, Disease-Free Survival, Female, Humans, Mice, Myristoylated Alanine-Rich C Kinase Substrate genetics, Neoplasm Metastasis, Ovarian Neoplasms genetics, Signal Transduction, Cancer-Associated Fibroblasts metabolism, Cancer-Associated Fibroblasts pathology, Myristoylated Alanine-Rich C Kinase Substrate metabolism, Ovarian Neoplasms metabolism, Ovarian Neoplasms pathology
- Abstract
The Cancer Genome Atlas network has revealed that the 'mesenchymal' epithelial ovarian cancer (EOC) subtype represents the poorest outcome, indicating a crucial role of stromal cancer-associated fibroblasts (CAFs) in disease progression. The cooperative role of CAFs in EOC metastasis has long been recognized, but the mechanisms of stromal CAFs activation are still obscure. Therefore, we carried out an integrative analysis to identify the regulator genes that are responsible for CAFs activation in microdissected tumor stroma profiles. Here, we determined that myristoylated alanine-rich C-kinase substrate (MARCKS) was highly expressed in ovarian stroma, and was required for the differentiation and tumor promoting function of CAFs. Suppression of MARCKS resulted in the loss of CAF features, and diminished role of CAFs in supporting tumor cell growth in 3D organotypic cultures and in murine xenograft model. Mechanistically, we found that MARCKS maintained CAF activation through suppression of cellular senescence and activation of the AKT/Twist1 signaling. Moreover, high MARCKS expression was associated with poor patient survival in EOC. Collectively, our findings identify the potential of MARCKS inhibition as a novel stroma-oriented therapy in EOC., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2016
- Full Text
- View/download PDF
16. Leptin as a mediator of tumor-stromal interactions promotes breast cancer stem cell activity.
- Author
-
Giordano C, Chemi F, Panza S, Barone I, Bonofiglio D, Lanzino M, Cordella A, Campana A, Hashim A, Rizza P, Leggio A, Győrffy B, Simões BM, Clarke RB, Weisz A, Catalano S, and Andò S
- Subjects
- Blotting, Western, Breast Neoplasms metabolism, Breast Neoplasms pathology, Cell Communication drug effects, Cell Communication genetics, Cell Line, Tumor, Cells, Cultured, Culture Media, Conditioned pharmacology, Fibroblasts metabolism, Gene Expression Profiling methods, Gene Expression Regulation, Neoplastic drug effects, HSP90 Heat-Shock Proteins genetics, HSP90 Heat-Shock Proteins metabolism, Humans, Kaplan-Meier Estimate, Leptin metabolism, Leptin pharmacology, MCF-7 Cells, Oligonucleotide Array Sequence Analysis, Receptors, Leptin genetics, Receptors, Leptin metabolism, Reverse Transcriptase Polymerase Chain Reaction, Spheroids, Cellular drug effects, Spheroids, Cellular metabolism, Tumor Microenvironment genetics, Breast Neoplasms genetics, Leptin genetics, Neoplastic Stem Cells metabolism, Stromal Cells metabolism
- Abstract
Breast cancer stem cells (BCSCs) play crucial roles in tumor initiation, metastasis and therapeutic resistance. A strict dependency between BCSCs and stromal cell components of tumor microenvironment exists. Thus, novel therapeutic strategies aimed to target the crosstalk between activated microenvironment and BCSCs have the potential to improve clinical outcome. Here, we investigated how leptin, as a mediator of tumor-stromal interactions, may affect BCSC activity using patient-derived samples (n = 16) and breast cancer cell lines, and determined the potential benefit of targeting leptin signaling in these model systems. Conditioned media (CM) from cancer-associated fibroblasts and breast adipocytes significantly increased mammosphere formation in breast cancer cells and depletion of leptin from CM completely abrogated this effect. Mammosphere cultures exhibited increased leptin receptor (OBR) expression and leptin exposure enhanced mammosphere formation. Microarray analyses revealed a similar expression profile of genes involved in stem cell biology among mammospheres treated with CM and leptin. Interestingly, leptin increased mammosphere formation in metastatic breast cancers and expression of OBR as well as HSP90, a target of leptin signaling, were directly correlated with mammosphere formation in metastatic samples (r = 0.68/p = 0.05; r = 0.71/p = 0.036, respectively). Kaplan-Meier survival curves indicated that OBR and HSP90 expression were associated with reduced overall survival in breast cancer patients (HR = 1.9/p = 0.022; HR = 2.2/p = 0.00017, respectively). Furthermore, blocking leptin signaling by using a full leptin receptor antagonist significantly reduced mammosphere formation in breast cancer cell lines and patient-derived samples. Our results suggest that leptin/leptin receptor signaling may represent a potential therapeutic target that can block the stromal-tumor interactions driving BCSC-mediated disease progression.
- Published
- 2016
- Full Text
- View/download PDF
17. Targeting chemotherapy-induced PTX3 in tumor stroma to prevent the progression of drug-resistant cancers.
- Author
-
Chi JY, Hsiao YW, Li CF, Lo YC, Lin ZY, Hong JY, Liu YM, Han X, Wang SM, Chen BK, Tsai KK, and Wang JM
- Subjects
- Animals, Apoptosis drug effects, C-Reactive Protein antagonists & inhibitors, Cell Line, Tumor, Cell Movement, Cell Survival, Cell Transformation, Neoplastic genetics, Cisplatin chemistry, Culture Media, Conditioned chemistry, Disease Progression, Female, Fibroblasts drug effects, Fibroblasts metabolism, Fluorouracil chemistry, Gene Expression Regulation, Neoplastic, Humans, Macrophages drug effects, Macrophages metabolism, Mice, Mice, Inbred BALB C, Myofibroblasts drug effects, Neoplasm Invasiveness, Neoplasm Metastasis, Neoplasm Transplantation, Neoplasms metabolism, Reverse Transcriptase Polymerase Chain Reaction, Serum Amyloid P-Component antagonists & inhibitors, Tumor Microenvironment drug effects, Antineoplastic Agents chemistry, C-Reactive Protein chemistry, C-Reactive Protein metabolism, CCAAT-Enhancer-Binding Protein-delta metabolism, Drug Resistance, Neoplasm, Peptide Fragments chemistry, Serum Amyloid P-Component chemistry, Serum Amyloid P-Component metabolism
- Abstract
The tumor microenvironment has been suggested to participate in tumorigenesis, but the nature of the communication between cancer cells and the microenvironment, especially in response to anticancer drugs, remains obscure. We determined that activation of the CCAAT/enhancer binding protein delta (CEBPD) response to Cisplatin and 5-Fluorouracil in cancer-associated macrophages and fibroblasts contributed to the metastasis, invasion, acquired chemoresistance and stemness of cancer cells by in vitro and in vivo assays. Specifically, reporter and in vivo DNA binding assays were used to determine that Pentraxin 3 (PTX3) is a CEBPD responsive gene and serves a protumor role upon anticancer drug treatment. Finally, a PTX3 peptide inhibitor RI37 was developed and assessed the antitumor effects by in vivo assays. RI37 could function as a promising inhibitor for preventing cancer progression and the metastasis, invasion and progression of drug-resistant cancers. The identification of PTX3 provided a new insight in the interaction between host and tumor and the RI37 peptide showed a great opportunity to largely reduce the risk of invasion and metastasis of cancer and drug-resistant cancers.
- Published
- 2015
- Full Text
- View/download PDF
18. Estrogenic gper signaling regulates mir144 expression in cancer cells and cancer-associated fibroblasts (cafs).
- Author
-
Vivacqua A, De Marco P, Santolla MF, Cirillo F, Pellegrino M, Panno ML, Abonante S, and Maggiolini M
- Subjects
- Animals, Cell Line, Tumor, Core Binding Factor Alpha 2 Subunit biosynthesis, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Fibroblasts pathology, Gene Expression Regulation, Neoplastic, Hep G2 Cells, Heterografts, Humans, MCF-7 Cells, Mice, Mice, Nude, MicroRNAs biosynthesis, Neoplasm Transplantation, Neoplasms genetics, Phosphatidylinositol 3-Kinases metabolism, Signal Transduction genetics, Tumor Microenvironment physiology, ets-Domain Protein Elk-1 metabolism, Estradiol metabolism, Estrogen Receptor alpha metabolism, MicroRNAs genetics, Neoplasms pathology, Receptors, Estrogen metabolism, Receptors, G-Protein-Coupled metabolism
- Abstract
MicroRNAs (miRNAs) are small non coding RNA molecules that play a crucial role in several pathophysiological conditions, including cancer. The stimulation of hormone-sensitive tumors by estrogens are mediated by estrogen receptor (ER)α and G protein estrogen receptor (GPER). Previous studies have reported that ERα regulates miRNA expression, while this ability of GPER remains to be elucidated. Here, we demonstrate that in SkBr3 breast cancer and HepG2 hepatocarcinoma cells, 17β-estradiol (E2) and the selective GPER ligand G-1 induce miR144 expression through GPER and the involvement of the PI3K/ERK1/2/Elk1 transduction pathway. Moreover, we show that E2 and G-1 down-regulate through miR144 the onco-suppressor Runx1 and increase cell cycle progression. The capability of E2 and G-1 in triggering the induction of miR144 and the down-regulation of Runx1 was also confirmed in cancer-associated fibroblasts (CAFs) that are main components of the tumor microenvironment driving cancer progression. Further confirming these results, Runx1 protein levels were found decreased in tumor xenografts upon G-1 treatment. On the basis of our findings miR144 and Runx1 may be included among the oncotargets of GPER action. Moreover, the present data provide new insights regarding the ability of estrogens to trigger the GPER/miR144/Runx1 transduction pathway toward the stimulation of cancer progression.
- Published
- 2015
- Full Text
- View/download PDF
19. TIMP-1 activated carcinoma-associated fibroblasts inhibit tumor apoptosis by activating SDF1/CXCR4 signaling in hepatocellular carcinoma.
- Author
-
Song T, Dou C, Jia Y, Tu K, and Zheng X
- Subjects
- Animals, Apoptosis, Cell Proliferation, Female, Humans, Male, Mice, Mice, Nude, Signal Transduction, Transfection, Carcinoma, Hepatocellular genetics, Fibroblasts metabolism, Liver Neoplasms genetics, Receptors, CXCR4 metabolism, Tissue Inhibitor of Metalloproteinase-1 genetics, Tissue Inhibitor of Metalloproteinase-1 metabolism
- Abstract
Tissue inhibitor of metalloproteinase 1 (TIMP-1) is an endogenous inhibitor for MMPs that regulates the remodeling and turnover of the ECM during normal development and pathological conditions. Intriguingly, recent studies have shown that TIMP-1 plays a dual role in cancer progression. In this study, we found that TIMP-1 expression in HCC tissues is associated with advanced TNM stage, intrahepatic metastasis, portal vein invasion, and vasculature invasion. Notably, TIMP-1 expression in HCC tissue is significantly related to worse overall survival for patients with HCC after liver resection. Ectopic TIMP1 expression promoted the growth of HCC xenografts in nude mice. Both co-culture with Huh7 cells with a high level of TIMP-1 and TIMP1 treatment resulted in up-regulation of hallmarks of carcinoma-associated fibroblasts (CAFs) and accelerated cell proliferation, migration and invasion in immortalized liver fibroblasts (LFs) isolated from human normal liver tissue. By co-culture with CAFs, SDF-1/CXCR4/PI3K/AKT signaling was activated and apoptosis was markedly repressed with an increased Bcl-2/BAX ratio in Huh7 cells. Taken together, our observations suggest that TIMP-1 induces the trans-differentiation of LFs into CAFs, suppresses apoptosis via SDF-1/CXCR4/PI3K/AKT signaling and then promotes HCC progression. This protein may be a potential prognostic biomarker and therapeutic target for HCC.
- Published
- 2015
- Full Text
- View/download PDF
20. Cyclooxygenase-2 blockade can improve efficacy of VEGF-targeting drugs.
- Author
-
Ben-Batalla I, Cubas-Cordova M, Udonta F, Wroblewski M, Waizenegger JS, Janning M, Sawall S, Gensch V, Zhao L, Martinez-Zubiaurre I, Riecken K, Fehse B, Pantel K, Bokemeyer C, and Loges S
- Subjects
- Animals, Cell Line, Tumor, Female, Mammary Neoplasms, Experimental metabolism, Mice, Mice, Inbred BALB C, Molecular Targeted Therapy, Neovascularization, Pathologic drug therapy, Random Allocation, Signal Transduction, Cyclooxygenase 2 metabolism, Cyclooxygenase 2 Inhibitors pharmacology, Mammary Neoplasms, Experimental blood supply, Mammary Neoplasms, Experimental drug therapy, Vascular Endothelial Growth Factor A metabolism
- Abstract
Anti-angiogenic therapies were approved for different cancers. However, significant primary and secondary resistance hampers efficacy in several tumor types including breast cancer. Thus, we need to develop clinically applicable strategies to enhance efficacy of anti-angiogenic drugs.We report that anti-angiogenic therapies can induce upregulation of cyclooxygenase-2 (Cox-2) and of its product prostaglandin E2 (PGE2) in breast cancer models. Upon Cox-2 inhibition PGE2 levels were normalized and efficacy of anti-vascular endothelial growth factor receptor 2 (anti-VEGFR-2) antibodies and sunitinib was enhanced. Interestingly, both treatments exerted additive anti-angiogenic effects. Following Cox-2 inhibition, we observed reduced infiltration of tumors with cancer-associated fibroblasts (CAFs) and lower levels of pro-angiogenic factors active besides the VEGF axis including hepatocyte growth factor (HGF) and basic fibroblast growth factor (FGF2). Mechanistic studies indicated that Cox-2 inhibition reduced PGE2-induced migration and proliferation of CAFs via inhibiting phosphorylation of Akt.Hence, Cox-2 inhibition can increase efficacy of anti-angiogenic treatments and our findings might pave the road for clinical investigations of concomitant blockade of Cox-2 and VEGF-signaling.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.