1
|
Zhang Y, Hei F, Xiao Y, Liu Y, Han J, Hu D, Wang H. Acidic fibroblast growth factor inhibits reactive oxygen species-induced epithelial-mesenchymal transdifferentiation in vascular endothelial cells via the miR-155-5p/SIRT1/Nrf2/HO-1 pathway to promote wound healing in diabetic mice. BURNS & TRAUMA 2024; 12:tkae010. [PMID: 38803612 PMCID: PMC11129767 DOI: 10.1093/burnst/tkae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/22/2023] [Revised: 02/19/2024] [Accepted: 02/22/2024] [Indexed: 05/29/2024]
Abstract
Background Diabetic chronic wounds are among the most common and serious complications of diabetes and are associated with significant morbidity and mortality. Endothelial-to-mesenchymal transition (EndMT) is a specific pathological state in which endothelial cells are transformed into mesenchymal cells in response to various stimuli, such as high glucose levels and high oxidative stress. Acidic fibroblast growth factor (aFGF), which is a member of the fibroblast growth factor family, possesses strong antioxidant properties and can promote the differentiation of mesenchymal stem cells into angiogenic cells. Therefore, we investigated the role of aFGF in EndMT in diabetic wounds and analysed the underlying mechanisms. Methods A diabetic mouse model was used to verify the effect of aFGF on wound healing, and the effect of aFGF on vascular endothelial cells in a high-glucose environment was examined in vitro. We examined the expression of miR-155-5p in a high-glucose environment and the miR-155 downstream target gene SIRT1 by luciferase reporter assays. Results aFGF promoted wound closure and neovascularization in a mouse model of type 2 diabetes. In vitro, aFGF inhibited the production of total and mitochondrial reactive oxygen species (ROS) in vascular endothelial cells and alleviated epithelial-mesenchymal transdifferentiation in a high-glucose environment. Mechanistically, aFGF promoted the expression of SIRT1 and the downstream targets Nrf2 and HO-1 by negatively regulating miR-155-5p, thereby reducing ROS generation. Conclusions In conclusion, our results suggest that aFGF inhibits ROS-induced epithelial-mesenchymal transdifferentiation in diabetic vascular endothelial cells via the miR-155-5p/SIRT1/Nrf2/HO-1 axis, thereby promoting wound healing.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Fenghui Hei
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yujie Xiao
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Yang Liu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Juntao Han
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| | - Hongtao Wang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, Fourth Military Medical University, 127 Changle West Road, Xi'an, Shaanxi 710032, China
| |
Collapse
|
2
|
Kong T, Qu Y, Zhao T, Niu Z, Lv X, Wang Y, Ding Q, Wei P, Fu J, Wang L, Gao J, Zhou C, Wang S, Jiang J, Zheng J, Wang K, Wu K. Identification of novel protein biomarkers from the blood and urine for the early diagnosis of bladder cancer via proximity extension analysis. J Transl Med 2024; 22:314. [PMID: 38532419 PMCID: PMC10967215 DOI: 10.1186/s12967-024-04951-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/24/2023] [Accepted: 02/04/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Bladder cancer (BC) is a very common urinary tract malignancy that has a high incidence and lethality. In this study, we identified BC biomarkers and described a new noninvasive detection method using serum and urine samples for the early detection of BC. METHODS Serum and urine samples were retrospectively collected from patients with BC (n = 99) and healthy controls (HC) (n = 50), and the expression levels of 92 inflammation-related proteins were examined via the proximity extension analysis (PEA) technique. Differential protein expression was then evaluated by univariate analysis (p < 0.05). The expression of the selected potential marker was further verified in BC and adjacent tissues by immunohistochemistry (IHC) and single-cell sequencing. A model was constructed to differentiate BC from HC by LASSO regression and compared to the detection capability of FISH. RESULTS The univariate analysis revealed significant differences in the expression levels of 40 proteins in the serum (p < 0.05) and 17 proteins in the urine (p < 0.05) between BC patients and HC. Six proteins (AREG, RET, WFDC2, FGFBP1, ESM-1, and PVRL4) were selected as potential BC biomarkers, and their expression was evaluated at the protein and transcriptome levels by IHC and single-cell sequencing, respectively. A diagnostic model (a signature) consisting of 14 protein markers (11 in serum and three in urine) was also established using LASSO regression to distinguish between BC patients and HC (area under the curve = 0.91, PPV = 0.91, sensitivity = 0.87, and specificity = 0.82). Our model showed better diagnostic efficacy than FISH, especially for early-stage, small, and low-grade BC. CONCLUSION Using the PEA method, we identified a panel of potential protein markers in the serum and urine of BC patients. These proteins are associated with the development of BC. A total of 14 of these proteins can be used to detect early-stage, small, low-grade BC. Thus, these markers are promising for clinical translation to improve the prognosis of BC patients.
Collapse
Affiliation(s)
- Tong Kong
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Yang Qu
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Taowa Zhao
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Zitong Niu
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Xiuyi Lv
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Yiting Wang
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Qiaojiao Ding
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Pengyao Wei
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China
| | - Jun Fu
- LC-Bio Technology Co., Ltd., Hangzhou, China
| | | | - Jing Gao
- LC-Bio Technology Co., Ltd., Hangzhou, China
| | - Cheng Zhou
- Department of Urology, Key Laboratory of Translational Research for Urology of Ningbo City, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University (Ningbo First Hospital), Ningbo, Zhejiang, China
| | - Suying Wang
- Ningbo Clinical Pathology Diagnostic Centre, Ningbo, Zhejiang, China
| | - Junhui Jiang
- Department of Urology, Key Laboratory of Translational Research for Urology of Ningbo City, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University (Ningbo First Hospital), Ningbo, Zhejiang, China
| | - Jianping Zheng
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China.
| | - Kaizhe Wang
- Ningbo Cixi Institute of BioMedical Engineering, Ningbo Institute of Materials Technology & Engineering, Chinese Academy of Sciences (CAS), Ningbo, 315300, People's Republic of China.
| | - Kerong Wu
- Department of Urology, Key Laboratory of Translational Research for Urology of Ningbo City, Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, The First Affiliated Hospital of Ningbo University (Ningbo First Hospital), Ningbo, Zhejiang, China.
| |
Collapse
|
3
|
Dordoe C, Huang W, Bwalya C, Wang X, Shen B, Wang H, Wang J, Ye S, Wang P, Xiaoyan B, Li X, Lin L. The role of microglial activation on ischemic stroke: Modulation by fibroblast growth factors. Cytokine Growth Factor Rev 2023; 74:122-133. [PMID: 37573252 DOI: 10.1016/j.cytogfr.2023.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2023] [Accepted: 07/29/2023] [Indexed: 08/14/2023]
Abstract
Stroke is one of the devastating clinical conditions that causes death and permanent disability. Its occurrence causes the reduction of oxygen and glucose supply, resulting in events such as inflammatory response, oxidative stress, and apoptosis in the brain. Microglia are brain-resident immune cells in the central nervous system (CNS) that exert diverse roles and respond to pathological process after an ischemic insult. The discovery of fibroblast growth factors (FGFs) in mammals, resulted to the findings that they can treat experimental models of stroke in animals effectively. FGFs function as homeostatic factors that control cells and hormones involved in metabolism, and they also regulate the secretion of proinflammatory (M1) and anti-inflammatory (M2) cytokines after stroke. In this review, we outline current evidence of microglia activation in experimental models of stroke focusing on its ability to exacerbate damage or repair tissue. Also, our review sheds light on the pharmacological actions of FGFs on multiple targets to regulate microglial modulation and highlighted their theoretical molecular mechanisms to provide possible therapeutic targets, as well as their limitations for the treatment of stroke. DATA AVAILABILITY: Not applicable.
Collapse
Affiliation(s)
- Confidence Dordoe
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Wenting Huang
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Canol Bwalya
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xue Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bixin Shen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hao Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jing Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shasha Ye
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Peng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Bao Xiaoyan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaokun Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, Zhejiang 325035, China.
| | - Li Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Research Units of Clinical Translation of Cell Growth Factors and Diseases Research, Chinese Academy of Medical Science, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
4
|
Wang J, Li L, Jiang X, Wang B, Hu X, Liu W, Zhang Y. Silencing of long non-coding RNA TUC338 inhibits the malignant phenotype of nasopharyngeal cancer cells via modulating the miR-1226-3p/FGF2 axis. Discov Oncol 2022; 13:102. [PMID: 36224455 PMCID: PMC9556687 DOI: 10.1007/s12672-022-00544-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 03/29/2022] [Accepted: 07/08/2022] [Indexed: 11/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been suggested as essential regulators in the cancer progression. LncRNA TUC338 was found to promote the malignancy of various cancers, however, the involvement of TUC338 in nasopharyngeal cancer (NPC) has not been well characterized. Here, our results found the significant overexpression of TUC338 in NPC tissues. Higher level of TUC338 was also observed in NPC cells. Interestingly, NPC patients harboring overexpressed TUC338 have worse prognosis. Functional study indicated that down-regulated TUC338 remarkably suppressed the NPC cell proliferation and cell migration. Notably, depletion of TUC338 significantly inhibited the in vivo tumor growth. Mechanistically, TUC338 acted as molecular sponge of miR-1226-3p and attenuated the negative regulation of miR-1226-3p on the expression of fibroblast growth factor 2 (FGF2). Down-regulation of TUC338 inhibited FGF2 expression in NPC cells and tumor tissues. Overexpression of FGF2 attenuated the suppressed NPC proliferation upon the depletion of TUC338. Our results demonstrated the novel function of TUC338/miR-1226-3p/FGF2 axis in NPC progression, suggesting the potential diagnosis and therapeutics significance of TUC338 in NPC.
Collapse
Affiliation(s)
- Jian Wang
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Liping Li
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Xue Jiang
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Bin Wang
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Xiaodong Hu
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Weiwei Liu
- Department of Otolaryngology, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China
| | - Ying Zhang
- Infection Department, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061000, Hebei, China.
| |
Collapse
|
5
|
Bondaruk J, Jaksik R, Wang Z, Cogdell D, Lee S, Chen Y, Dinh KN, Majewski T, Zhang L, Cao S, Tian F, Yao H, Kuś P, Chen H, Weinstein JN, Navai N, Dinney C, Gao J, Theodorescu D, Logothetis C, Guo CC, Wang W, McConkey D, Wei P, Kimmel M, Czerniak B. The origin of bladder cancer from mucosal field effects. iScience 2022; 25:104551. [PMID: 35747385 PMCID: PMC9209726 DOI: 10.1016/j.isci.2022.104551] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/24/2021] [Revised: 11/19/2021] [Accepted: 06/02/2022] [Indexed: 12/30/2022] Open
Abstract
Whole-organ mapping was used to study molecular changes in the evolution of bladder cancer from field effects. We identified more than 100 dysregulated pathways, involving immunity, differentiation, and transformation, as initiators of carcinogenesis. Dysregulation of interleukins signified the involvement of inflammation in the incipient phases of the process. An aberrant methylation/expression of multiple HOX genes signified dysregulation of the differentiation program. We identified three types of mutations based on their geographic distribution. The most common were mutations restricted to individual mucosal samples that targeted uroprogenitor cells. Two types of mutations were associated with clonal expansion and involved large areas of mucosa. The α mutations occurred at low frequencies while the β mutations increased in frequency with disease progression. Modeling revealed that bladder carcinogenesis spans 10-15 years and can be divided into dormant and progressive phases. The progressive phase lasted 1-2 years and was driven by β mutations.
Collapse
Affiliation(s)
- Jolanta Bondaruk
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Roman Jaksik
- Department of Systems Biology and Engineering and Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Ziqiao Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Cogdell
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sangkyou Lee
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yujie Chen
- Systems, Synthetic and Physical Biology Program, Rice University, Houston, TX, USA
| | - Khanh Ngoc Dinh
- Department of Statistics and the Irving Institute for Cancer Dynamics, Columbia University, New York, NY, USA
| | - Tadeusz Majewski
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhang
- Department of Environmental Health, University of Cincinnati, Cincinnati, OH, USA
| | - Shaolong Cao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Feng Tian
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Yao
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paweł Kuś
- Department of Systems Biology and Engineering and Biotechnology Centre, Silesian University of Technology, Gliwice, Poland
| | - Huiqin Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John N. Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Neema Navai
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Colin Dinney
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Dan Theodorescu
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai, Los Angeles, CA, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, TX, USA
| | - Charles C. Guo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wenyi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marek Kimmel
- Department of Statistics, Rice University, Houston, TX, USA
| | - Bogdan Czerniak
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
6
|
Zou Y, Hu J, Huang W, Ye S, Han F, Du J, Shao M, Guo R, Lin J, Zhao Y, Xiong Y, Wang X. Non-Mitogenic Fibroblast Growth Factor 1 Enhanced Angiogenesis Following Ischemic Stroke by Regulating the Sphingosine-1-Phosphate 1 Pathway. Front Pharmacol 2020; 11:59. [PMID: 32194396 PMCID: PMC7063943 DOI: 10.3389/fphar.2020.00059] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/13/2019] [Accepted: 01/22/2020] [Indexed: 12/11/2022] Open
Abstract
Ischemic strokes account for about 80% of all strokes and are associated with a high risk of mortality. Angiogenesis of brain microvascular endothelial cells may contribute to functional restoration following ischemia. Fibroblast growth factor 1 (FGF1), a member of FGF superfamily, involved in embryonic development, angiogenesis, wound healing, and neuron survival. However, the mitogenic activity of FGF1 is known to contribute to several human pathologies, thereby questioning the safety of its clinical applications. Here, we explored the effects and mechanism of action of non-mitogenic FGF1 (nmFGF1) on angiogenesis in mice after ischemia stroke and an oxygen-glucose deprivation (OGD)-induced human brain microvascular endothelial cells (HBMECs) injury model. We found that intranasal administration nmFGF1 significantly promoted angiogenesis in mice after stroke, and significantly increased the formation of matrigel tube and promoted scratch migration in a dose-dependent manner in OGD-induced HBMECs in vitro. However, the co-administration of an FGF receptor 1 (FGFR1)-specific inhibitor PD173074 significantly reversed the effects of nmFGF1 in vitro, suggesting that nmFGF1 functions via FGFR1 activation. Moreover, nmFGF1 activated sphingosine-1-phosphate receptor 1 (S1PR1, S1P1) in mice after stroke in vivo. S1P1 protein antagonist VPC23019 and agonist FTY720 were used to confirm that nmFGF1 promotes angiogenesis in vitro partially through the S1P1 pathway. OGD induced downregulation of S1P1 expression. The S1P1 antagonist VPC23019 blocked the stimulatory effects of nmFGF1, whereas the S1P1 agonist FTY720 exerted effects comparable with those of nmFGF1. Furthermore, PD173074 reversed the effect of nmFGF1 on upregulating S1P1 signaling. In conclusion, nmFGF1 enhanced angiogenesis in mice following stroke and OGD-induced HBMECs through S1P1 pathway regulation mediated via FGFR1 activation. This new discovery suggests the potential therapeutic role of nmFGF1 for the treatment of ischemic strokes.
Collapse
Affiliation(s)
- Yuchi Zou
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Hu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenting Huang
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Shasha Ye
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Fanyi Han
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingting Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Mingjie Shao
- School of the First Clinical Medical Science, Wenzhou Medical University, Wenzhou, China
| | - Ruili Guo
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jingjing Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yeli Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou, China
| | - Ye Xiong
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xue Wang
- The Frist Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
7
|
Pecqueux C, Arslan A, Heller M, Falkenstein M, Kaczorowski A, Tolstov Y, Sültmann H, Grüllich C, Herpel E, Duensing A, Kristiansen G, Hohenfellner M, Navone NM, Duensing S. FGF-2 is a driving force for chromosomal instability and a stromal factor associated with adverse clinico-pathological features in prostate cancer. Urol Oncol 2018; 36:365.e15-365.e26. [PMID: 29887238 DOI: 10.1016/j.urolonc.2018.05.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2017] [Revised: 05/03/2018] [Accepted: 05/15/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND There is mounting evidence to suggest that stromal cells play an integral role in the progression of prostate cancer (PCa). One of the most frequently altered growth factors in PCa is fibroblast growth factor-2 (FGF-2). It has previously been proposed that early stages of PCa are characterized by a primarily exogenous, that is, stromal cell-derived FGF-2 production, whereas advanced tumors rely more on an autocrine FGF-2 production. Prostate cancer progression is characterized by an increase of genomic instability including aneuploidy and structural chromosomal alterations. Herein, we address 2 problems that have not been comprehensively answered. First, we ask whether exogenous FGF-2 can directly drive genomic instability to promote PCa progression. Second, we investigate whether and to what extent stromal FGF-2 expression is maintained in advanced PCa and whether this influences tumor progression and patient prognosis. METHODS In vitro experiments to investigate the role of FGF-2 in numerical and structural chromosomal instability were performed using immunofluorescence microscopy, fluorescence in situ hybridization and single cell electrophoresis. A human patient-derived xenograft mouse model recapitulating osteoblastic PCa bone metastasis was used for in vivo validation experiments. The prognostic role of stromal FGF-2 expression was analyzed using immunohistochemical staining of a tissue microarray with primary tumor specimens from 162 predominantly high-risk patients with PCa. RESULTS Our results show that FGF-2 not only rapidly induces mitotic defects and numerical chromosomal imbalances but also an enhanced DNA breakage to promote chromosomal instability. Using the patient-derived xenograft model, we show that a deregulation of the FGF axis results in an increase of mitotic aberrations as well as DNA damage checkpoint activation in vivo. The FGFR inhibitor dovitinib was found to reduce numerical chromosomal instability as well as DNA breakage, thus underscoring the relevance of the FGF axis in promoting genomic instability. An overexpression of tumor cell-associated FGF-2 was detected in 52 of 162 patients (32.1%), whereas a stromal overexpression was found in 27 of 165 patients (16%). Remarkably, a strong stromal FGF-2 expression was associated with a significantly higher clinical stage and higher biochemical recurrence rate. Patients with strong stromal FGF-2 expression also had a significantly worse biochemical recurrence-free survival. CONCLUSIONS Our results underscore that exogenous FGF-2 can shape PCa cell genomes and that stromal FGF-2 expression is detectable in a sizeable proportion of advanced PCa where it is associated with adverse clinico-pathological features. Our results highlight the impact of the tumor stroma on malignant progression and provide a rationale for a further exploration of components of the tumor stroma as therapeutic targets in PCa.
Collapse
Affiliation(s)
- Carine Pecqueux
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany; Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Aysenur Arslan
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Martina Heller
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Michael Falkenstein
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Adam Kaczorowski
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Yanis Tolstov
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| | - Holger Sültmann
- Division of Cancer Genome Research, National Center for Tumor Diseases (NCT), German Cancer Research Center, and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Carsten Grüllich
- Department of Medical Oncology, University of Heidelberg School of Medicine, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University of Heidelberg School of Medicine, Heidelberg, Germany; Tissue Bank of the National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anette Duensing
- Cancer Therapeutics Program, University of Pittsburgh Cancer Institute, Hillman Cancer Center, Pittsburgh, PA
| | - Glen Kristiansen
- Institute of Pathology, University of Bonn School of Medicine, Bonn, Germany
| | - Markus Hohenfellner
- Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany
| | - Nora M Navone
- Division of Genitourinary Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX
| | - Stefan Duensing
- Molecular Urooncology, Department of Urology, Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany; Department of Urology, University of Heidelberg School of Medicine, Heidelberg, Germany.
| |
Collapse
|
8
|
Hui Q, Zhang L, Yang X, Yu B, Huang Z, Pang S, Zhou Q, Yang R, Li W, Hu L, Li X, Cao G, Wang X. Higher Biostability of rh-aFGF-Carbomer 940 Hydrogel and Its Effect on Wound Healing in a Diabetic Rat Model. ACS Biomater Sci Eng 2018; 4:1661-1668. [PMID: 33445322 DOI: 10.1021/acsbiomaterials.8b00011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022]
Abstract
Hydrogels are excellent drug delivery carriers with excellent ductility. Here, we report the design of a higher biostability of a recombinant human acidic fibroblast growth factor (rh-aFGF) carbomer hydrogel formulation. To verify the optimality of this formula, we prepared various prescriptions and tested the resulting physical properties including micromorphology, long-term stability, accelerated stability, and destructive test. Furthermore, the efficacy for promoting wound healing in full-thickness injury and scald wound diabetic rat models was explored. We found that rh-aFGF-carbomer hydrogel had good physical properties. It was stable for 24 months at 5 ± 3 °C, and for 6 months at 25 ± 3 °C. In vivo, the rh-aFGF-carbomer 940 hydrogel achieved a remarkable promotion of skin wound healing in diabetic rats with full-thickness injuries or scald wounds. Our data suggest that rh-aFGF-carbomer hydrogel may have applications for the treatment of diabetic ulcers combined with other wounds.
Collapse
Affiliation(s)
- Qi Hui
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | | | - Xuanxin Yang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Bingjie Yu
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Zhen Huang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Shucai Pang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Qingde Zhou
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Rongshuai Yang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Wenqing Li
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Lufeng Hu
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Xiaokun Li
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Gaozhong Cao
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| | - Xiaojie Wang
- School of Pharmacy, Wenzhou Medical University, Chashan University Park, Wenzhou 325035, China
| |
Collapse
|
9
|
Chen L, Lei Z, Ma X, Huang Q, Zhang X, Zhang Y, Hao P, Yang M, Zhao X, Chen J, Liu G, Zheng T. Prognostic significance of fibroblast growth factor receptor 4 polymorphisms on biochemical recurrence after radical prostatectomy in a Chinese population. Sci Rep 2016; 6:33604. [PMID: 27640814 PMCID: PMC5027536 DOI: 10.1038/srep33604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/18/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is a transmembrane receptor with ligand-induced tyrosine kinase activity and is involved in various biological and pathological processes. Several polymorphisms of FGFR4 are associated with the incidence and mortality of numerous cancers, including prostate cancer. In this study, we investigated whether the polymorphisms of FGFR4 influence the biochemical recurrence of prostate cancer in Chinese men after radical prostatectomy. Three common polymorphisms (rs1966265, rs2011077, and rs351855) of FGFR4 were genotyped from 346 patients with prostate cancer by using the Sequenom MassARRAY system. Kaplan–Meier curves and Cox proportional hazard models were used for survival analysis. Results showed biochemical recurrence (BCR) free survival was significantly affected by the genotypes of rs351855 but not influenced by rs1966265 and rs2011077. After adjusting for other variables in multivariable analysis, patients with rs351855 AA/AG genotypes showed significantly worse BCR-free survival than those with the GG genotype (HR = 1.873; 95% CI, 1.209–2.901; P = 0.005). Hence, FGFR4 rs351855 could be a novel independent prognostic factor of BCR after radical prostatectomy in the Chinese population. This functional polymorphism may also provide a basis for surveillance programs. Additional large-scale studies must be performed to validate the significance of this polymorphism in prostate cancer.
Collapse
Affiliation(s)
- Luyao Chen
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Zhengwei Lei
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xin Ma
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Qingbo Huang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Xu Zhang
- Department of Urology, Chinese PLA General Hospital, Beijing, China
| | - Yong Zhang
- Department of Urology, Puai Hospital, Wuhan, China
| | - Peng Hao
- Department of Urology, Puai Hospital, Wuhan, China
| | | | - Xuetao Zhao
- Department of Urology, Puai Hospital, Wuhan, China
| | - Jun Chen
- Department of Urology, Puai Hospital, Wuhan, China
| | - Gongxue Liu
- Department of Urology, Puai Hospital, Wuhan, China
| | - Tao Zheng
- Department of Urology, Puai Hospital, Wuhan, China
| |
Collapse
|
10
|
Liu C, Guan H, Wang Y, Chen M, Xu B, Zhang L, Lu K, Tao T, Zhang X, Huang Y. miR-195 Inhibits EMT by Targeting FGF2 in Prostate Cancer Cells. PLoS One 2015; 10:e0144073. [PMID: 26650737 PMCID: PMC4674136 DOI: 10.1371/journal.pone.0144073] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/21/2015] [Accepted: 10/14/2015] [Indexed: 11/21/2022] Open
Abstract
Prostate cancer (PCa) is one of the leading causes of deaths in America. The major cause of mortality can be attributed to metastasis. Cancer metastasis involves sequential and interrelated events. miRNAs and epithelial-mesenchymal transition (EMT) are implicated in this process. miR-195 is downregulated in many human cancers. However, the roles of miR-195 in PCa metastasis and EMT remain unclear. In this study, data from Memorial Sloan Kettering Cancer Center (MSKCC) prostate cancer database were re-analysed to detect miR-195 expression and its roles in PCa. miR-195 was then overexpressed in castration-resistant PCa cell lines, DU-145 and PC-3. The role of miR-195 in migration and invasion in vitro was also investigated, and common markers in EMT were evaluated through Western blot analysis. A luciferase reporter assay was conducted to confirm the target gene of miR-195; were validated in PCa cells. In MSKCC data re-analyses, miR-195 was poorly expressed in metastatic PCa; miR-195 could be used to diagnose metastatic PCa by measuring the corresponding expression. Area under the receiver operating characteristic curve (AUC-ROC) was 0.705 (P = 0.017). Low miR-195 expression was characterised with a shorter relapse-free survival (RFS) time. miR-195 overexpression suppressed cell migration, invasion and EMT. Fibroblast growth factor 2 (FGF2) was confirmed as a direct target of miR-195. FGF2 knockdown also suppressed migration, invasion and EMT; by contrast, increased FGF2 partially reversed the suppressive effect of miR-195. And data from ONCOMINE prostate cancer database showed that PCa patients with high FGF2 expression showed shorter RFS time (P = 0.046). Overall, this study demonstrated that miR-195 suppressed PCa cell metastasis by downregulating FGF2. miR-195 restoration may be considered as a new therapeutic method to treat metastatic PCa.
Collapse
Affiliation(s)
- Chunhui Liu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Han Guan
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Yiduo Wang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- * E-mail:
| | - Bin Xu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Lei Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Kai Lu
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Tao Tao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Xiaowen Zhang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| | - Yeqing Huang
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
- Surgical Research Center, Institute of Urology, Southeast University Medical School, 87 Dingjia Bridge Hunan Road, Nanjing, 210009, China
| |
Collapse
|
11
|
Shi S, Li X, You B, Shan Y, Cao X, You Y. High Expression of FGFR4 Enhances Tumor Growth and Metastasis in Nasopharyngeal Carcinoma. J Cancer 2015; 6:1245-54. [PMID: 26535066 PMCID: PMC4622855 DOI: 10.7150/jca.12825] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2015] [Accepted: 08/28/2015] [Indexed: 01/02/2023] Open
Abstract
Background: FGF receptor (FGFR) family can be activated by FGFs and play important roles in regulating cell growth, differentiation, migration and angiogenesis. Recent studies suggested that FGFR4 could regulate several processes including tumor progression. Nasopharyngeal carcinoma (NPC) is a malignancy with a high occurrence in Southeast Asia and Southern China. However, the molecule mechanism and the potential roles of FGFR4 in NPC remain unknown Methods: Immunohistochemistry and western blot were used to investigate the expression of FGFR4 in NPC samples. Then we used statistical analysis to evaluate the diagnostic value and the associations of FGFR4 expression with clinical parameters. In vitro studies, the effects of FGFR4 on proliferation and migration of NPC cell line CNE2 were measured by the starvation-refeeding experiment, CCK8 assay, wounding healing assay and transwell migration assay. The changes of the epithelial-mesenchymal transition (EMT) markers in CNE2 cells after knocking down the expression of FGFR4 were measured by Western blot and immunofluorescence analysis. Results: FGFR4 was overexpressed in NPC as compared with the inflammatory tissues. High expression of FGFR4 was correlated with Ki67 expression, clinical stages and prognosis in NPC patients (P<0.05).While in vitro, the upregulation of FGFR4 was accompanied with CNE2 cells released from serum starvation. Moreover, it could increase cell proliferation and migration by regulating EMT markers in CNE2 cells. Conclusion: Our data suggested that FGFR4 might induce NPC progression and act as a potential therapeutic target in NPC.
Collapse
Affiliation(s)
- Si Shi
- 1. Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xingyu Li
- 2. Department of Pathology, Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Bo You
- 1. Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ying Shan
- 1. Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xiaolei Cao
- 2. Department of Pathology, Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Yiwen You
- 1. Department of Otorhinolaryngology Head and Neck surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
12
|
Xiao L, Yang S, Hao J, Yuan X, Luo W, Jiang L, Hu Y, Fu Z, Zhang Y, Zou C. Endostar attenuates melanoma tumor growth via its interruption of b-FGF mediated angiogenesis. Cancer Lett 2015; 359:148-54. [PMID: 25597785 DOI: 10.1016/j.canlet.2015.01.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/23/2014] [Revised: 11/25/2014] [Accepted: 01/08/2015] [Indexed: 12/31/2022]
Abstract
To develop optimal therapeutics is one of the hotspots in both clinical and basic melanoma studies. Previous studies indicate that fibroblast growth factors (b-FGF/FGF-2), an angiogenesis inducer beyond VEGF, might be a potential drug target in melanoma. As a novel anti-angiogenesis peptide drug, Endostar has shown promising therapeutic efficacy in non-small cell lung cancer. However, the effect of Endostar on b-FGF-induced angiogenesis in melanoma is unraveled. To this end, both in vivo and in vitro experiments were conducted and it was found that treatment of Endostar could inhibit tumor growth, which was accompanied by decreased micro-vessel density and serum b-FGF levels in a mouse melanoma model. In addition, treatment with Endostar in blood vessel endothelial cells could reduce their proliferation, cell migration and tube formation capacity in a dosage-dependent manner. Moreover, treatment of Endostar could also attenuate b-FGF-activated phosphorylation of p38 and ERK1/2 in HUVECs. These findings indicate that Endostar might exert its anti-tumor effect via suppressing b-FGF-induced angiogenesis and b-FGF-activated MAPK signaling pathway, suggesting that Endostar might be a potential choice for clinical melanoma treatment.
Collapse
Affiliation(s)
- Lijia Xiao
- Department of Clinical Laboratory, Nanshan affiliated Hospital of Guangdong Medical College, Shenzhen 518052, China
| | - ShuCai Yang
- Department of Surgery, School of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Department of Laboratory Medicine, Yijishan Hospital, Wannan Medical College, Wuhu 241001, Anhui, China
| | - Jianhua Hao
- Department of Clinical Laboratory, Nanshan affiliated Hospital of Guangdong Medical College, Shenzhen 518052, China
| | - Xue Yuan
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Wei Luo
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Liping Jiang
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Yang Hu
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Zhongping Fu
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Yun Zhang
- Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China
| | - Chang Zou
- Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Department of Pharmacology, Jiangsu Simcere Pharmaceutical Research Institute, 699-18 Xuan Wu Avenue, Nanjing 210042, China.
| |
Collapse
|
13
|
miRNA-646 suppresses osteosarcoma cell metastasis by downregulating fibroblast growth factor 2 (FGF2). Tumour Biol 2014; 36:2127-34. [PMID: 25403884 DOI: 10.1007/s13277-014-2822-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/05/2014] [Accepted: 11/06/2014] [Indexed: 10/24/2022] Open
Abstract
MicroRNAs are short regulatory RNAs that play crucial roles in cancer development and progression. MicroRNA-646 (miR-646) is downregulated in many human cancers, and increasing evidence indicates that it functions as a tumor suppressor. However, the role of miR-646 in osteosarcoma remains unclear. Expression levels of miR-646 in osteosarcoma cell lines and patient tissues were evaluated by quantitative real-time PCR (qRT-PCR), and the clinicopathological significance of the resultant data was later analyzed. Next, we investigated the role of miR-646 to determine its potential roles on osteosarcoma cell proliferation, migration, and invasion in vitro. A luciferase reporter assay was conducted to confirm the target gene of miR-646, and the results were validated in the osteosarcoma cell line. In this study, we found that miR-646 was downregulated in osteosarcoma cell lines and osteosarcoma tissues compared with normal osteoblast cell line NHOst and paired adjacent nontumor tissue. We found that a lower expression of miR-646 was associated with metastasis. In osteosarcoma cells, overexpression of miR-646 inhibited cell proliferation, migration, and invasion. In contrast, downregulation of miR-646 expression promoted osteosarcoma cell proliferation, migration, and invasion. Next, we identified that the FGF2 gene is a novel direct target of miR-646 in osteosarcoma cells. Moreover, enforced expression of FGF2 partially reversed the inhibition of cell proliferation, migration, and invasion that was caused by miR-646. Our study demonstrated that miR-646 might be a tumor suppressor in osteosarcoma via the regulation of FGF2, which provided a potential prognostic biomarker and therapeutic target.
Collapse
|
14
|
Wang Y, Wang H, Ren L, Weng Q, Bao Y, Tian H, Yang YG, Li X. Non-mitogenic form of acidic fibroblast growth factor protects against graft-versus-host disease without accelerating leukemia. Int Immunopharmacol 2014; 23:395-9. [PMID: 25239811 DOI: 10.1016/j.intimp.2014.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/25/2014] [Revised: 08/13/2014] [Accepted: 09/05/2014] [Indexed: 10/24/2022]
Abstract
Acid fibroblast growth factor (aFGF) has been shown to prevent epithelial damage under various conditions, suggesting its potential to inhibit GVHD. However, because aFGF receptors are expressed on tumor cells, it may possibly offset the graft-vs.-tumor (GVT) effects of allogeneic bone marrow transplantation (allo-BMT). Here, we addressed these questions in a B6→B6D2F1 allo-BMT model. Although aFGF administration attenuated GVHD in non-leukemic recipients, aFGF treatment markedly accelerated death in mice that received recipient-type tumor (P815) cells along with allo- or syngeneic-BMT. Similar protection against GVHD was achieved by administration of a non-mitogenic form of aFGF (naFGF). Importantly, GVT effects were fully preserved in naFGF-treated recipients. Furthermore, aFGF, but not naFGF, significantly enhanced P815 cell proliferation both in vitro and in vivo. Our data indicate that the tumor-promoting, but not GVHD-protecting, effect of aFGF largely depends on its mitogenic activity, and suggest that naFGF may provide a safer approach to inhibiting GVHD in patients with malignancies.
Collapse
Affiliation(s)
- Yi Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China; Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | - Hui Wang
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA
| | - Luqing Ren
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Qiaoyou Weng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yuyan Bao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Haishan Tian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China
| | - Yong-Guang Yang
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA; Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, USA; First Hospital of Jilin University, Changchun, PR China.
| | - Xiaokun Li
- Key Laboratory of Biotechnology Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, PR China.
| |
Collapse
|
15
|
Wu X, Huang H, Wang C, Lin S, Huang Y, Wang Y, Liang G, Yan Q, Xiao J, Wu J, Yang Y, Li X. Identification of a novel peptide that blocks basic fibroblast growth factor-mediated cell proliferation. Oncotarget 2014; 4:1819-28. [PMID: 24142482 PMCID: PMC3858566 DOI: 10.18632/oncotarget.1312] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/25/2022] Open
Abstract
Basic fibroblast growth factor (bFGF) has been implicated in tumor growth via interactions with its receptors (FGFRs) on the cell surface and therefore, bFGF/FGFRs are considered essential targets for cancer therapy. Herein, a consensus heptapeptide (LSPPRYP) was identified for the first time from a phage display heptapeptide library after three sequential rounds of biopanning against FGFR-expressing cells with competitive displacement of phage by bFGF, followed by subtraction of non-specific binding by FGFR-deficient cells. Phage bearing LSPPRYP showed high levels of binding to Balb/c 3T3 cells expressing high-affinity bFGF-binding FGFR (bFGFR), but not to the cells that do not express bFGFR (Cos-7), or express a very low affinity bFGFR (HaCat). The selected-phage-derived peptide synthesized by solid phase method using a rapid and practical Fmoc strategy was found to specifically compete with bFGF for binding to its receptors, inhibit bFGF-stimulated cell proliferation by inducing cell cycle arrest, and block bFGF-induced activation of Erk1 and Erk2 kinase in B16-F10 melanoma cells. Importantly, treatment of melanoma-bearing mice with the synthetic peptide significantly suppressed tumor growth. The results demonstrate a strong anticancer activity of the isolated bFGFR-binding peptide (and its future derivatives), which may have great potential for cancer therapy.
Collapse
Affiliation(s)
- Xiaoping Wu
- School of Pharmaceutical Science, Key Laboratory of Biotechnology and Pharmaceutical Engineering of Zhejiang Province, Wenzhou Medical College, Wenzhou, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li D, Wei X, Xie K, Chen K, Li J, Fang J. A novel decoy receptor fusion protein for FGF-2 potently inhibits tumour growth. Br J Cancer 2014; 111:68-77. [PMID: 24874473 PMCID: PMC4090743 DOI: 10.1038/bjc.2014.282] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/17/2014] [Revised: 04/29/2014] [Accepted: 04/30/2014] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Antiangiogenic therapies have been proven effective in cancer treatment. Fibroblast growth factor-2 (FGF-2) has been functionally implicated in tumour angiogenesis and is an important target of antiangiogenic therapies. The aim of this work was to develop a novel FGF-2 inhibitor for cancer therapy. METHODS Eleven fusion proteins were developed by fusing various truncated extracellular regions of FGFR1 with the Fc region of IgG1. The optimal decoy receptor fusion protein with the highest binding affinity for FGF-2 was identified by an FGF-2-binding assay and its potential antitumour effects were investigated. RESULTS We obtained a soluble decoy receptor fusion protein with the highest binding activity for FGF-2, named FGF-Trap. Fibroblast growth factor-Trap significantly abolished FGF-2-stimulated activation of FGF signalling as demonstrated by its suppression of FGF-2-mediated phosphorylation of Erk1/2 and Akt, upregulation of cyclins D1 and E and the increase in mRNA levels of vascular endothelial growth factor R1 and R2 (VEGFR1 and VEGFR2). Furthermore, FGF-Trap effectively suppressed FGF-2-induced proliferation and migration of human umbilical vein endothelial cells (HUVECs) in vitro. Most importantly, FGF-Trap potently inhibited tumour growth and angiogenesis in Caki-1 and A549 xenograft models in vivo. CONCLUSIONS Fibroblast growth factor-Trap potently inhibits tumour growth by blocking FGF-2 signalling pathways and could be an effective therapeutic agent for cancer patients.
Collapse
Affiliation(s)
- D Li
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - X Wei
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - K Xie
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - K Chen
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - J Li
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - J Fang
- School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| |
Collapse
|
17
|
Tan X, Chen X, Huang T, Luo W, He D, Lin X, Song L, Wu X. Mechanism of bFGF-binding Peptide Reversing Adriamycin Resistance in Human Gastric Cancer Cells. Int J Pept Res Ther 2014. [DOI: 10.1007/s10989-014-9402-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/04/2023]
|
18
|
Deng SQ, Xu H, He Q, Jiang HX, Su BJ, Zhang QH. Detecting the developmental toxicity of bFGF in the embryonic stem cell test using differential gene expression of differentiation-related genes. Toxicol Mech Methods 2014; 24:323-31. [PMID: 24559483 DOI: 10.3109/15376516.2014.894165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Abstract
Basic fibroblast growth factor (bFGF) is a mitogenic cytokine that can stimulate mesoderm-and neuroectoderm-originated cell proliferation. This study was performed to investigate the effects of bFGF on cell differentiation and the expression of specific markers at different embryonic developmental stages. We firstly evaluated the embryotoxic potential of bFGF in vitro using a modified EST protocol. Sequentially, we further investigated how bFGF impact the different tissue-special genes and proteins expressions during the differentiation of murine ES cells in vitro and attempt to reveal the effects of bFGF on differentiation processes. This analysis was focused on key tissue- and stage-specific genes involved in ectodermal, mesodermal, and endodermal differentiation, including ectodermal-specific gene Nestin, Oligo2 and Syn, mesodermal-specific gene MHC and MyoD, and endodermal-specific gene GATA6, TTR and ALB, as well as undifferentiated gene Sox-2 and Oct-4. The results demonstrate that bFGF could promote expression of ectodermal-specific genes and protein, but suppress the expressions of endoderm-specific and some mesoderm-specific gene and protein. A conclusion can be drawn that bFGF exhibits weak embryotoxicity and mainly promotes ES cell differentiation towards the ectodermal lineages but suppress differentiation into endoderm lineages. These opposing effects of bFGF on the embryonic development of the three germ layers may be related to its weak embryotoxic potential. More specifically, inhibition of expression of the endodermal-specific markers transthyretin (TTR), and albumin (ALB) by bFGF may be of more value in detecting the embryotoxic potential of bFGF.
Collapse
Affiliation(s)
- Shu-Qin Deng
- Pharmacy College, Jinan University , Guangzhou , China
| | | | | | | | | | | |
Collapse
|
19
|
Carstens JL, Shahi P, Van Tsang S, Smith B, Creighton CJ, Zhang Y, Seamans A, Seethammagari M, Vedula I, Levitt JM, Ittmann MM, Rowley DR, Spencer DM. FGFR1-WNT-TGF-β signaling in prostate cancer mouse models recapitulates human reactive stroma. Cancer Res 2013; 74:609-20. [PMID: 24305876 DOI: 10.1158/0008-5472.can-13-1093] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
Abstract
The reactive stroma surrounding tumor lesions performs critical roles ranging from supporting tumor cell proliferation to inducing tumorigenesis and metastasis. Therefore, it is critical to understand the cellular components and signaling control mechanisms that underlie the etiology of reactive stroma. Previous studies have individually implicated fibroblast growth factor receptor 1 (FGFR1) and canonical WNT/β-catenin signaling in prostate cancer progression and the initiation and maintenance of a reactive stroma; however, both pathways are frequently found to be coactivated in cancer tissue. Using autochthonous transgenic mouse models for inducible FGFR1 (JOCK1) and prostate-specific and ubiquitously expressed inducible β-catenin (Pro-Cat and Ubi-Cat, respectively) and bigenic crosses between these lines (Pro-Cat × JOCK1 and Ubi-Cat × JOCK1), we describe WNT-induced synergistic acceleration of FGFR1-driven adenocarcinoma, associated with a pronounced fibroblastic reactive stroma activation surrounding prostatic intraepithelial neoplasia (mPIN) lesions found both in in situ and reconstitution assays. Both mouse and human reactive stroma exhibited increased transforming growth factor-β (TGF-β) signaling adjacent to pathologic lesions likely contributing to invasion. Furthermore, elevated stromal TGF-β signaling was associated with higher Gleason scores in archived human biopsies, mirroring murine patterns. Our findings establish the importance of the FGFR1-WNT-TGF-β signaling axes as driving forces behind reactive stroma in aggressive prostate adenocarcinomas, deepening their relevance as therapeutic targets.
Collapse
Affiliation(s)
- Julienne L Carstens
- Authors' Affiliations: Departments of Pathology and Immunology and Molecular and Cellular Biology; and Dan L Duncan Cancer Center, Baylor College of Medicine, Houston; M.E. DeBakey, Department of Veterans Affairs Medical Center, Houston, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Luo W, Yu Y, Wang R, He D, Wang C, Zeng X, Chen X, Tan X, Huang T, Wu X. P7 peptides targeting bFGF sensitize colorectal cancer cells to CPT-11. Int J Mol Med 2013; 33:194-200. [PMID: 24190390 DOI: 10.3892/ijmm.2013.1547] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2013] [Accepted: 10/04/2013] [Indexed: 11/06/2022] Open
Abstract
The low survival rate of patients with colorectal cancer (CRC) is mainly due to the drug resistance of tumor cells to chemotherapeutic agents. It has been reported that basic fibroblast growth factor (bFGF) is an essential factor involved in the epigenetic mechanisms of drug resistance, which provides a novel potential target for improving the sensitivity of tumor cells to chemotherapeutic agents. In this study, we first demonstrate that a novel bFGF antagonist, peptide P7, previously isolated by phage display technology, reversed bFGF-induced resistance to irinotecan hydrochloride (CPT-11), and counteracted the anti-apoptotic effects of bFGF on CPT-11-treated HT-29 cells. Further experiments indicated that the inhibition of Akt activation, the suppression of bFGF internalization, the increase in the Bax to Bcl-2 ratio and the downregulation of cytokeratin 8 (CK8) by P7 may contribute to the counteracting of the anti-apoptotic effects of bFGF, and further reversal of bFGF-induced resistance to CPT-11. Our results suggest that peptide P7 may have therapeutic potential in CRC as a sensitizer to chemotherapeutic agents by targeting bFGF.
Collapse
Affiliation(s)
- Wu Luo
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, Guangdong, P.R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Wang R, Luo W, Zeng X, Di J, Yu Y, Wang C, Wang W, Chen X, He D, Li T, Wu X. A bFGF Antagonist Peptide with Anti-angiogenesis Properties in Non-small Cell Lung Cancer Cells. Int J Pept Res Ther 2013. [DOI: 10.1007/s10989-013-9352-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/24/2022]
|
23
|
Peláez-García A, Barderas R, Torres S, Hernández-Varas P, Teixidó J, Bonilla F, de Herreros AG, Casal JI. FGFR4 role in epithelial-mesenchymal transition and its therapeutic value in colorectal cancer. PLoS One 2013; 8:e63695. [PMID: 23696849 PMCID: PMC3655941 DOI: 10.1371/journal.pone.0063695] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/09/2012] [Accepted: 04/06/2013] [Indexed: 12/16/2022] Open
Abstract
Fibroblast growth factor receptor 4 (FGFR4) is vital in early development and tissue repair. FGFR4 expression levels are very restricted in adult tissues, except in several solid tumors including colorectal cancer, which showed overexpression of FGFR4. Here, FGFR4 mutation analysis discarded the presence of activating mutations, other than Arg(388), in different colorectal cancer cell lines and tumoral samples. Stable shRNA FGFR4-silencing in SW480 and SW48 cell lines resulted in a significant decrease in cell proliferation, adhesion, cell migration and invasion. This decrease in the tumorigenic and invasive capabilities of colorectal cancer cells was accompanied by a decrease of Snail, Twist and TGFβ gene expression levels and an increase of E-cadherin, causing a reversion to a more epithelial phenotype, in three different cell lines. In addition, FGFR4-signaling activated the oncogenic SRC, ERK1/2 and AKT pathways in colon cancer cells and promoted an increase in cell survival. The relevance of FGFR4 in tumor growth was supported by two different strategies. Kinase inhibitors abrogated FGFR4-related cell growth and signaling pathways at the same extent than FGFR4-silenced cells. Specific FGFR4-targeting using antibodies provoked a similar reduction in cell growth. Moreover, FGFR4 knock-down cells displayed a reduced capacity for in vivo tumor formation and angiogenesis in nude mice. Collectively, our data support a crucial role for FGFR4 in tumorigenesis, invasion and survival in colorectal cancer. In addition, FGFR4 targeting demonstrated its applicability for colorectal cancer therapy.
Collapse
Affiliation(s)
- Alberto Peláez-García
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Rodrigo Barderas
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | - Sofía Torres
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
| | | | - Joaquín Teixidó
- Chemokines and Cell Migration Laboratory, CIB-CSIC, Madrid, Spain
| | - Félix Bonilla
- Hospital Puerta de Hierro Majadahonda, Madrid, Spain
| | | | - J. Ignacio Casal
- Department of Celular and Molecular Medicine, Centro de Investigaciones Biológicas (CIB-CSIC), Madrid, Spain
- * E-mail:
| |
Collapse
|
24
|
Yang Z, Yang Z, Zou Q, Yuan Y, Li J, Li D, Liang L, Zeng G, Chen S. A comparative study of clinicopathological significance, FGFBP1, and WISP-2 expression between squamous cell/adenosquamous carcinomas and adenocarcinoma of the gallbladder. Int J Clin Oncol 2013; 19:325-35. [PMID: 23592278 DOI: 10.1007/s10147-013-0550-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/20/2012] [Accepted: 03/09/2013] [Indexed: 12/29/2022]
Abstract
BACKGROUND The differences in clinical, pathological, and biological characteristics between adenocarcinoma (AC) and squamous cell/adenosquamous carcinoma (SC/ASC) of gallbladder cancer have not been well documented. This study is to compare the clinicopathological characteristics and FGFBP1 and WISP-2 expression between AC and SC/ASC patients. METHODS We examined FGFBP1 and WISP-2 expression in 46 SC/ASC and 80 AC samples using immunohistochemistry and analyzed their correlations with clinicopathological characteristics. RESULTS SC/ASCs occur more frequently in older patients and often correspond to larger tumor masses than ACs. Positive FGFBP1 and negative WISP-2 expression were significantly associated with lymph node metastasis and invasion of SC/ASCs and ACs. In addition, positive FGFBP1 and negative WISP-2 expression were significantly associated with differentiation and TMN stage in ACs. Univariate Kaplan-Meier analysis showed that either elevated FGFBP1 (p < 0.001) or lowered WISP-2 (p < 0.001) expression was closely associated with decreased overall survival in both SC/ASC and AC patients. Multivariate Cox regression analysis showed that positive FGFBP1 expression (p = 0.001) or negative WISP-2 expression (p = 0.035 for SC/ASC and p = 0.009 for AC) is an independent predictor of poor prognosis in both SC/ASC and AC patients. We also revealed that differentiation, tumor size, TNM stage, lymph node metastasis, invasion, and surgical procedure were associated with survival of both SC/ASC and AC patients. CONCLUSION Our study suggested that the overexpression of FGFBP1 or loss of WISP-2 expression is closely related to the metastasis, invasion and poor prognosis of gallbladder cancer.
Collapse
Affiliation(s)
- Zhulin Yang
- Research Laboratory of Hepatobiliary Diseases, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, People's Republic of China,
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Jiang X, Skibba M, Zhang C, Tan Y, Xin Y, Qu Y. The roles of fibroblast growth factors in the testicular development and tumor. J Diabetes Res 2013; 2013:489095. [PMID: 24159602 PMCID: PMC3789391 DOI: 10.1155/2013/489095] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/04/2013] [Accepted: 08/19/2013] [Indexed: 01/07/2023] Open
Abstract
Fibroblast growth factors (FGFs) are classically known as hormonal factors and recent studies have revealed that FGFs have a key role in regulating growth and development of several reproductive organs, including the testis. The testis is mainly consisted of germ cells, Sertoli cells and Leydig cells to develop and maintain the male phenotype and reproduction. This review summarizes the structure and fuctions of testis, the roles of FGFs on testicular development and potential involvement in testicular tumor and its regulatory mechanism. Among 23 members of FGFs, the FGF-1, FGF-2, FGF-4, FGF-8, FGF-9, and FGF-21 were involved and describe in details. Understanding the roles and mechanism of FGFs is the foundation to modeling testicular development and treatments in testicular disease. Therefore, in the last part, the potential therapy with FGFs for the testis of cancer and diabetes was also discussed.
Collapse
Affiliation(s)
- Xin Jiang
- The First Hospital of Jilin University, Changchun 130021, China
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville 40202, USA
| | - Melissa Skibba
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville 40202, USA
| | - Chi Zhang
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville 40202, USA
- The Chinese-American Research Institute for Diabetic Complications, Wenzhou 325200, China
| | - Yi Tan
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville 40202, USA
- The Chinese-American Research Institute for Diabetic Complications, Wenzhou 325200, China
| | - Ying Xin
- KCHRI at the Department of Pediatrics, The University of Louisville, Louisville 40202, USA
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun 130021, China
- *Ying Xin: and
| | - Yaqin Qu
- The First Hospital of Jilin University, Changchun 130021, China
- *Yaqin Qu:
| |
Collapse
|
26
|
Cuevas R, Korzeniewski N, Tolstov Y, Hohenfellner M, Duensing S. FGF-2 Disrupts Mitotic Stability in Prostate Cancer through the Intracellular Trafficking Protein CEP57. Cancer Res 2012; 73:1400-10. [DOI: 10.1158/0008-5472.can-12-1857] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
|
27
|
Yu Y, Gao S, Li Q, Wang C, Lai X, Chen X, Wang R, Di J, Li T, Wang W, Wu X. The FGF2-binding peptide P7 inhibits melanoma growth in vitro and in vivo. J Cancer Res Clin Oncol 2012; 138:1321-8. [PMID: 22481251 DOI: 10.1007/s00432-012-1201-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/09/2012] [Accepted: 03/12/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Melanoma is a malignant tumor and causes majority of deaths related to skin cancer. Fibroblast growth factor 2 (FGF2) greatly contributes to melanoma growth and progress. In this paper, we attempt to evaluate the therapeutic potential of FGF2-binding peptide (named P7) using as a potent FGF2 antagonist via exploration of its antitumor effect on melanoma in vitro and in vivo. METHODS Cell viability was measured by WST-1. Cell cycle progression was determined by propidium iodide staining and flow cytometry. Western blotting was carried out to detect the activation of Erk1/2, P38, Akt, and MEK, and the expression of apoptosis-associated proteins. The influence of P7 on FGF2 internalization was assessed by separation of nuclear and cytoplasmic protein fractions followed by Western blotting. Female C57BL/6 mice bearing xenograft melanoma were established and used to evaluate the antitumor effect of P7 in vivo. RESULTS In this study, we first proved that P7 peptides significantly inhibited proliferation of FGF2-induced melanoma cell line B16-F10. Further investigations revealed that the mechanisms of P7 peptides inhibiting cell proliferation of melanoma cells stimulated with FGF2 in vitro involved cell cycle arrest at the G0/G1 phase, blockade of the activation of Erk1/2, P38, and Akt cascades, and inhibition of FGF2 internalization. Finally, treatment of P7 peptides in a murine melanoma model resulted in significant inhibition of tumor growth and angiogenesis in vivo, which was associated with blockade of mitogen-activated protein kinase signal activation, and suppression of the expressions of anti-apoptotic Bcl-2 protein and angiogenic factor in the melanoma tumors. CONCLUSIONS The FGF2-binding peptide with potent antiproliferation and anti-angiogenic activity may have therapeutic potential in melanoma.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Western
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Endocytosis/drug effects
- Female
- Fibroblast Growth Factor 2/antagonists & inhibitors
- Fibroblast Growth Factor 2/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Flow Cytometry
- G1 Phase/genetics
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mitogen-Activated Protein Kinases/metabolism
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- Phosphorylation/drug effects
- Protein Binding
- Proto-Oncogene Proteins c-akt/metabolism
- Resting Phase, Cell Cycle/drug effects
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Yonglin Yu
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Ribeiro RJT, Monteiro CPD, Azevedo ASM, Cunha VFM, Ramanakumar AV, Fraga AM, Pina FM, Lopes CMS, Medeiros RM, Franco EL. Performance of an adipokine pathway-based multilocus genetic risk score for prostate cancer risk prediction. PLoS One 2012; 7:e39236. [PMID: 22792137 PMCID: PMC3387135 DOI: 10.1371/journal.pone.0039236] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/15/2012] [Accepted: 05/17/2012] [Indexed: 12/25/2022] Open
Abstract
Few biomarkers are available to predict prostate cancer risk. Single nucleotide polymorphisms (SNPs) tend to have weak individual effects but, in combination, they have stronger predictive value. Adipokine pathways have been implicated in the pathogenesis. We used a candidate pathway approach to investigate 29 functional SNPs in key genes from relevant adipokine pathways in a sample of 1006 men eligible for prostate biopsy. We used stepwise multivariate logistic regression and bootstrapping to develop a multilocus genetic risk score by weighting each risk SNP empirically based on its association with disease. Seven common functional polymorphisms were associated with overall and high-grade prostate cancer (Gleason≥7), whereas three variants were associated with high metastatic-risk prostate cancer (PSA≥20 ng/mL and/or Gleason≥8). The addition of genetic variants to age and PSA improved the predictive accuracy for overall and high-grade prostate cancer, using either the area under the receiver-operating characteristics curves (P<0.02), the net reclassification improvement (P<0.001) and integrated discrimination improvement (P<0.001) measures. These results suggest that functional polymorphisms in adipokine pathways may act individually and cumulatively to affect risk and severity of prostate cancer, supporting the influence of adipokine pathways in the pathogenesis of prostate cancer. Use of such adipokine multilocus genetic risk score can enhance the predictive value of PSA and age in estimating absolute risk, which supports further evaluation of its clinical significance.
Collapse
Affiliation(s)
- Ricardo J T Ribeiro
- Molecular Oncology Group-CI, Portuguese Institute of Oncology, Porto, Portugal.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Gao S, Yu Y, Lai X, Li T, Li Q, Chen X, Wang R, Wang W, Wu X. Anti-Proliferation Effect of a Short Peptide on bFGF-Induced Human Gastric Cancer Cells. Int J Pept Res Ther 2012. [DOI: 10.1007/s10989-012-9301-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/04/2023]
|
30
|
Stoehr CG, Stoehr R, Hartmann A, Hofstaedter F, Junker K, Blaszyk H, Wieland WF, Otto W, Denzinger S, Walter B. Mutational activation of FGFR3: no involvement in the development of renal cell carcinoma. J Cancer Res Clin Oncol 2012; 138:359-61. [PMID: 22203473 DOI: 10.1007/s00432-011-1130-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/19/2011] [Accepted: 12/19/2011] [Indexed: 12/20/2022]
Abstract
BACKGROUND Somatic point mutations in the fibroblast growth factor receptor 3 (FGFR3) gene have been identified in certain types of urological cancers, especially urothelial carcinoma of the bladder and the renal pelvis, and could be correlated with a favourable outcome. However, comprehensive data on the FGFR3 mutation status in renal cell carcinoma (RCC) are still missing. METHODS In order to investigate a possible role for FGFR3 mutations in renal cell carcinogenesis, we performed a sequence-based mutational analysis of FGFR3 in 238 primary RCC. The cohort obtained the common RCC subtypes including 101 clear cell, 50 papillary and 68 chromophobe RCC specimens. The analysed regions encompassed all FGFR3 point mutations previously described in epithelial tumours and other noncutaneous epithelial malignancies. RESULTS No mutations were detected in any renal tumour type examined, and all cases showed wild-type sequence. CONCLUSION Our results argue against an involvement of mutational activation of FGFR3 in the development of RCC. A recently described cystic renal dysplasia in a patient with thanatophoric dysplasia type 1 due to a germ line FGFR3 mutation might portend to an involvement of mutational FGFR3 activation in renal cyst formation, but this speculation needs further evaluation.
Collapse
Affiliation(s)
- C G Stoehr
- Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Wang J, Liu XT, Huang H, Xiao G, Zhou ZY, Chen Y, Yu ZH, He SL, Chen AA, Wang DD, He Y, Zhang ZC, Hong A. Antitumor activity of a recombinant soluble ectodomain of mutant human fibroblast growth factor receptor-2 IIIc. Mol Cancer Ther 2011; 10:1656-66. [PMID: 21750221 DOI: 10.1158/1535-7163.mct-11-0163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
The fibroblast growth factor (FGF) signaling pathway is a recognized target of cancer therapy. We have developed a strong inhibitor (S252W mutant soluble ectodomain of FGF recptor-2 IIIc, msFGFR2) that binds FGFs and blocks the activation of FGFRs. Thermodynamic binding studies indicated that msFGFR2 bound FGF-2 16.9 times as strongly as wild-type soluble FGFR2IIIc ectodomain (wsFGFR2). It successfully suppressed the growth, angiogenesis, and metastasis of two tumor cell lines in vitro and in vivo, and it potently inhibited cancer cell proliferation but not normal cell proliferation. Therefore, msFGFR2 is a useful probe for FGF-dependent signaling pathways and a potential broad-spectrum antitumor agent.
Collapse
Affiliation(s)
- Ju Wang
- Guangdong Provincial Key Laboratory of Bio-engineering Medicine, National Engineering Research Centre of Genetic Medicine, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong 510632, China.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wu X, Yan Q, Huang Y, Huang H, Su Z, Xiao J, Zeng Y, Wang Y, Nie C, Yang Y, Li X. Isolation of a novel basic FGF-binding peptide with potent antiangiogenetic activity. J Cell Mol Med 2011; 14:351-6. [PMID: 20414975 PMCID: PMC3837584 DOI: 10.1111/j.1582-4934.2008.00506.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/27/2023] Open
Abstract
Basic fibroblast growth factor (bFGF), which plays an important role in tumour angiogenesis and progression, provides a potential target for cancer therapy. Here we screened a phage display heptapeptide library with bFGF and identified 11 specific bFGF-binding phage clones. Two of these clones had identical sequence and the corresponding peptide (referred to as P7) showed high homology to the immunoglobulin-like (Ig-like) domain III (D3) of high-affinity bFGF receptors, FGFR1 (IIIc) and FGFR2 (IIIc). The P7 peptide and its corresponding motif in D3 of FGFRs both carried negative charges and shared similar hydrophobic profiles. Functional analysis demonstrated that synthetic P7 peptides mediate strong inhibition of bFGF-induced cell proliferation and neovascularization. Our results demonstrate that the P7 peptide is a potent bFGF antagonist with strong antiangiogenetic activity, and might have therapeutic potential in cancer therapy.
Collapse
Affiliation(s)
- Xiaoping Wu
- School of Pharmaceutical Science, Key Laboratory of Biotechnology and Pharmaceutical Engineering of Zhejiang Province, Wenzhou Medical College, Wenzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Zhang H, Zhao X, Yan L, Li M. Similar expression to FGF (Sef) reduces endometrial adenocarcinoma cells proliferation via inhibiting fibroblast growth factor 2-mediated MAPK/ERK signaling pathway. Gynecol Oncol 2011; 122:669-74. [PMID: 21663947 DOI: 10.1016/j.ygyno.2011.05.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/26/2011] [Revised: 05/12/2011] [Accepted: 05/14/2011] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Fibroblast growth factors (FGF) axis, and in particular FGF2 axis, is an important mitogenic stimulus in endometrial carcinogenesis. hSef is a key inhibitory regulator of FGF signaling and aberrant hSef expression is reported to be present in various human carcinomas. The objective of this study was to investigate the role of hSef in the growth and proliferation of endometrial adenocarcinoma cells and to explore the mechanism that may be involved. METHODS Using western blot analysis, we determined the expression of hSef in Ishikawa cells under different conditions. Using luciferase reporter assays and western blot analysis, we detected the effect of hSef on MAPK/ERK-mediated FGF2 signaling. Using MTT, cell counting and colony formation assays, we analyzed the growth and proliferation of Ishikawa cells under different conditions. RESULTS We found that the hSef expression was positively regulated by FGF2-induced MAPK/ERK signaling and inversely, hSef expression efficiently inhibited the activity of FGF2-induced MAPK/ERK signaling, indicating the presence of hSef-mediated negative feedback mechanism for FGF signaling in endometrial cancer cells. In addition, we found that MAPK/ERK signaling was essential for the growth and proliferation of endometrial cancer cells in vitro, and hSef expression significantly reduced the cell proliferation. CONCLUSIONS hSef expression can inhibit the growth and proliferation of endometrial cancer cells via acting on the FGF2/MAPK/ERK signaling.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Obstetrics & Gynecology, Provincial Hospital Affiliated to Shandong University, 324 Jingwu Road, Jinan, Shandong 250021, People's Republic of China
| | | | | | | |
Collapse
|
34
|
Tsimafeyeu I, Demidov L, Stepanova E, Wynn N, Ta H. Overexpression of fibroblast growth factor receptors FGFR1 and FGFR2 in renal cell carcinoma. ACTA ACUST UNITED AC 2011; 45:190-5. [DOI: 10.3109/00365599.2011.552436] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/13/2022]
Affiliation(s)
- Ilya Tsimafeyeu
- Kidney Cancer Research Bureau, Moscow, Russia, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia, University of Toronto, Toronto, Canada
| | - Lev Demidov
- Kidney Cancer Research Bureau, Moscow, Russia, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia, University of Toronto, Toronto, Canada
| | - Eugenia Stepanova
- Kidney Cancer Research Bureau, Moscow, Russia, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia, University of Toronto, Toronto, Canada
| | - Nigel Wynn
- Kidney Cancer Research Bureau, Moscow, Russia, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia, University of Toronto, Toronto, Canada
| | - Hung Ta
- Kidney Cancer Research Bureau, Moscow, Russia, N.N. Blokhin Russian Cancer Research Center, Moscow, Russia, University of Toronto, Toronto, Canada
| |
Collapse
|
35
|
|
36
|
Abstract
Solid tumors can be thought of as multicellular 'organs' that consist of a variety of cells as well as a scaffold of noncellular matrix. Stromal-epithelial crosstalk is integral to prostate cancer progression and metastasis, and androgen signaling is an important component of this crosstalk at both the primary and metastatic sites. Intratumoral production of androgen is an important mechanism of castration resistance and has been the focus of novel therapeutic approaches with promising results. Various other pathways are important for stromal-epithelial crosstalk and represent attractive candidate therapeutic targets. Hedgehog signaling has been associated with tumor progression, growth and survival, while Src family kinases have been implicated in tumor progression and in regulation of cancer cell migration. Fibroblast growth factors and transforming growth factor beta signaling regulate cell proliferation, apoptosis and angiogenesis in the prostate cancer microenvironment. Integrins mediate communication between the cell and the extracellular matrix, enhancing growth, migration, invasion and metastasis of cancer cells. The contribution of stromal-epithelial crosstalk to prostate cancer initiation and progression provides the impetus for combinatorial microenvironment-targeting strategies.
Collapse
Affiliation(s)
- Maria Karlou
- Department of Genitourinary Medical Oncology, David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77230-1439, USA
| | | | | |
Collapse
|
37
|
Boxler S, Djonov V, Kessler TM, Hlushchuk R, Bachmann LM, Held U, Markwalder R, Thalmann GN. Matrix metalloproteinases and angiogenic factors: predictors of survival after radical prostatectomy for clinically organ-confined prostate cancer? THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:2216-24. [PMID: 20889560 PMCID: PMC2966781 DOI: 10.2353/ajpath.2010.091190] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Accepted: 07/21/2010] [Indexed: 12/27/2022]
Abstract
The aim of the present study was to investigate whether biomarkers improve the prediction of recurrence-free, disease-specific, and overall survival in patients with clinically localized prostate cancer. A tissue microarray was constructed from prostate specimens of 278 patients who underwent open radical retropubic prostatectomy for clinically localized prostate cancer. For immunohistochemical studies, antibodies were used against matrix metalloproteinase (MMP)-2, MMP-3, MMP-7, MMP-9, MMP-13, and MMP-19, as well as against vascular endothelial growth factor, hypoxia-induced factor 1α, basic fibroblast growth factor, and cluster of differentiation 31. Univariate and multivariable analyses were performed to evaluate the potential predictors of overall, disease-specific, and recurrence-free survival. In univariate analysis of patients with clinically organ-confined prostate cancer, only higher expression levels of MMP-9 (hazard ratio [0.6], 95% CI 0.45-0.8) had a protective effect in terms of overall survival. This positive effect of high MMP-9 expression was also observed for recurrence-free (HR 0.88, 95% CI 0.78-0.99) and disease-specific survival (HR 0.5, 95% CI 0.36-0.73). In multivariable analysis, none of these potential markers was found to be an independent prognostic factor of survival. Of all MMPs and angiogenic factors tested, MMP-9 expression has the potential as a prognostic marker in patients undergoing radical prostatectomy for clinically organ-confined cases of prostate cancer.
Collapse
Affiliation(s)
- Silvan Boxler
- Institute of Anatomy, Department of Urology, University of Bern, 3010 Bern, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Wang C, Lin S, Nie Y, Jia X, Wang J, Xiao J, Wu J, Li X, Wu X. Mechanism of antitumor effect of a novel bFGF binding peptide on human colon cancer cells. Cancer Sci 2010; 101:1212-8. [PMID: 20331620 PMCID: PMC11159635 DOI: 10.1111/j.1349-7006.2010.01501.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022] Open
Abstract
Colon cancer is a leading cause of morbidity and mortality in Western countries. Basic fibroblast growth factor (bFGF) was up-regulated in patients with colon cancer and was considered as a potential therapeutic target. In this study, we first demonstrated that a novel bFGF-binding peptide (named P7) inhibited proliferation of several colon cancer cell lines including HT-29, LoVo, and Caco2 cells stimulated by bFGF. Further investigations with HT-29 cells indicated that P7 arrested the cell cycle at the G0/G1 phase of bFGF-stimulated cells, reduced the levels of phospho-Erk1/Erk2 induced by bFGF, and caused significant changes in the expression of proteins related to proliferation, cell cycle, and cancer. Our results suggested that the bFGF-binding peptide has a potential antitumor effect on colon cancer.
Collapse
Affiliation(s)
- Cong Wang
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fletcher AM, Bregman CL, Woicke J, Salcedo TW, Zidell RH, Janke HE, Fang H, Janusz WJ, Schulze GE, Mense MG. Incisor degeneration in rats induced by vascular endothelial growth factor/fibroblast growth factor receptor tyrosine kinase inhibition. Toxicol Pathol 2010; 38:267-79. [PMID: 20100840 DOI: 10.1177/0192623309357950] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
BMS-645737, an inhibitor of vascular endothelial growth factor (VEGF) receptor-2 and fibroblast growth factor (FGF) receptor-1, has anti-angiogenic activity and was evaluated in nonclinical studies as a treatment for cancer. This article characterizes the BMS-645737-induced clinical, gross, and histologic lesions of incisor teeth in Sprague-Dawley (SD) rats. Rats received 0 800 mg/kg BMS-645737 in a single-dose study or consecutive daily doses of 0 20 mg/kg/day in a 1-month study. The reversibility of these effects was assessed in the 1-month study. White discoloration and fracture of incisors were observed clinically and grossly in the 1-month study. In both studies, dose-dependent histopathologic lesions of incisors were degeneration and/or necrosis of odontoblasts and ameloblasts; decreased mineralization of dentin; inflammation and necrosis of the dental pulp; and edema, congestion, and hemorrhage in the pulp and periodontal tissue adjacent to the enamel organ. Partial recovery was observed at lower doses after a two-week dose-free period in the one-month study. Drug-induced incisor lesions were considered to be related to the pharmacologic inhibitory effects on VEGF and FGF signaling, that is, inhibition of growth and maintenance of small-diameter vessels that support the formation of dentin and enamel in growing teeth and/or to perturbances of function of odontoblasts and ameloblasts or their precursors.
Collapse
Affiliation(s)
- Anthony M Fletcher
- Drug Safety Evaluation, Bristol-Myers Squibb Company, Syracuse, New York, NY 13221-4755, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Li L, Ren C, Yang G, Goltsov AA, Tabata KI, Thompson TC. Caveolin-1 promotes autoregulatory, Akt-mediated induction of cancer-promoting growth factors in prostate cancer cells. Mol Cancer Res 2009; 7:1781-91. [PMID: 19903767 DOI: 10.1158/1541-7786.mcr-09-0255] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2023]
Abstract
Caveolin-1 (cav-1) and the cancer-promoting growth factors vascular endothelial growth factor (VEGF), transforming growth factor beta1 (TGF-beta1), and fibroblast growth factor 2 (FGF2) are often found to be upregulated in advanced prostate cancer and other malignancies. However, the relationship between cav-1 overexpression and growth factor upregulation remains unclear. This report presents, to our knowledge, the first evidence that in prostate cancer cells, a positive autoregulatory feedback loop is established in which VEGF, TGF-beta1, and FGF2 upregulate cav-1, and cav-1 expression, in turn, leads to increased levels of VEGF, TGF-beta1, and FGF2 mRNA and protein, resulting in enhanced invasive activities of prostate cancer cells, i.e., migration and motility. Our results further show that cav-1-enhanced mRNA stability is a major mechanism underlying the upregulation of these cancer-promoting growth factors, and that PI3-K-Akt signaling is required for forming this positive autoregulatory feedback loop.
Collapse
Affiliation(s)
- Likun Li
- Department of Genitourinary Medical Oncology, University of Texas M D Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | | | | | | | | | |
Collapse
|
41
|
Wang Y, Lin H, Lin S, Qu J, Xiao J, Huang Y, Xiao Y, Fu X, Yang Y, Li X. Cell-penetrating peptide TAT-mediated delivery of acidic FGF to retina and protection against ischemia-reperfusion injury in rats. J Cell Mol Med 2009; 14:1998-2005. [PMID: 19432810 PMCID: PMC3823281 DOI: 10.1111/j.1582-4934.2009.00786.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/30/2022] Open
Abstract
The development of non-invasive ocular drug delivery systems is of practical importance in the treatment of retinal disease. In this study, we evaluated the efficacy of transactivator of transcription protein transduction domain (TAT-PTD, TAT(49-57)) as a vehicle to deliver acidic FGF (aFGF) to retina in rats. TAT-conjugated aFGF-His (TAT-aFGF-His) exhibited efficient penetration into the retina following topical administration to the ocular surface. Immunochemical staining with anti-His revealed that TAT-aFGF-His proteins were readily found in the retina (mainly in the ganglion cell layer) at 30 min. and remained detectable for at least 8 hrs after administration. In contrast, His(+) proteins were undetectable in the retina after topical administration of aFGF-His, indicating that aFGF-His cannot penetrate the ocular barrier. Furthermore, TAT-aFGF-His, but not aFGF-His, mediated significant protection against retinal ischemia-reperfusion (IR) injury. After IR injury, retina from TAT-aFGF-His-treated rats showed better-maintained inner retinal layer structure, reduced apoptosis of retinal ganglion cells and improved retinal function compared to those treated with aFGF-His or PBS. These results indicate that conjugation of TAT to aFGF-His can markedly improve the ability of aFGF-His to penetrate the ocular barrier without impairing its biological function. Thus, TAT(49-57) provides a potential vehicle for efficient drug delivery in the treatment of retinal disease.
Collapse
Affiliation(s)
- Yi Wang
- Key Laboratory of Biotechnology Pharmaceutical Engineering of Zhejiang Province, School of Pharmaceutical Science, Wenzhou Medical College, Wenzhou, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Wang L, Zhang H, Zhang S, Yu M, Yang X. Construction and characterization of a novel superantigen fusion protein: bFGF/SEB. Cancer Invest 2009; 27:376-83. [PMID: 19160106 DOI: 10.1080/07357900802487228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND As one of the bacterial superantigens, Staphylococcal enterotoxins (SEs) are potent activators of T cells, especially for those expressing T cell receptor V(beta) chains, and can induce the production of cytokines such as IFN-gamma, TNF-alpha, IL-1, IL-2, IL-6, IL-12, etc. Thus, SEs could be used in tumor-targeting therapy when cooperated with the vectors that can specifically recognize the tumor cells. MATERIALS AND METHODS The coding sequences of Staphylococcal enterotxin B (SEB) was amplified and fused with human basic fibroblast growth factor (bFGF). Recombinant protein SEB and fusion protein bFGF/SEB were expressed and purified. The biological activity was detected, including splenocytes proliferation, cytokine production, and cytotoxicity in tumor cells in vitro. In addition, the binding of bFGF/SEB with tumor cells and the tumor cell apoptosis were also tested by immunofluorescent technique. RESULTS The fusion protein bFGF/SEB had similar biological activities compared with natural SEA and recombinant SEB, including tumor-inhibition ratio. CONCLUSION The recombinant bFGF/SEB-fusion protein was shown to retain the superantigenic activity of SEB, and might be a novel promising immunotherapeutic agent for the treatment of some carcinomas.
Collapse
Affiliation(s)
- Lichan Wang
- Serum Division, National Institute for Control of Pharmaceutical and Biological Products, Beijing, China
| | | | | | | | | |
Collapse
|
43
|
Sirintrapun SJ, Parwani AV. Molecular Pathology of the Genitourinary Tract: Prostate and Bladder. Surg Pathol Clin 2008; 1:211-36. [PMID: 26837907 DOI: 10.1016/j.path.2008.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
The knowledge of cellular mechanisms in tumors of the prostate and bladder has grown exponentially. Molecular technologies have led to the discovery of TMPRSS2 in prostate cancer and the molecular pathways distinguishing low- and high-grade urothelial neoplasms. UroVysion with fluorescence in situ hybridization is already commonplace as an adjunct to cytologic diagnosis of urothelial neoplasms. This trend portends the future in which classification and diagnosis of tumors of the prostate and bladder through morphologic analysis will be supplemented by molecular information correlating with prognosis and targeted therapy. This article outlines tumor molecular pathology of the prostate and bladder encompassing current genomic, epigenomic, and proteonomic findings.
Collapse
Affiliation(s)
- S Joseph Sirintrapun
- Pathology Informatics, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Anil V Parwani
- Department of Pathology, University of Pittsburgh Medical Center Shadyside Hospital, Room WG 07, 5230 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
44
|
Fischer H, Taylor N, Allerstorfer S, Grusch M, Sonvilla G, Holzmann K, Setinek U, Elbling L, Cantonati H, Grasl-Kraupp B, Gauglhofer C, Marian B, Micksche M, Berger W. Fibroblast growth factor receptor-mediated signals contribute to the malignant phenotype of non-small cell lung cancer cells: therapeutic implications and synergism with epidermal growth factor receptor inhibition. Mol Cancer Ther 2008; 7:3408-19. [PMID: 18852144 PMCID: PMC2879863 DOI: 10.1158/1535-7163.mct-08-0444] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/22/2023]
Abstract
Fibroblast growth factors (FGF) and their high-affinity receptors (FGFR) represent an extensive cellular growth and survival system. Aim of this study was to evaluate the contribution of FGF/FGFR-mediated signals to the malignant growth of non-small cell lung cancer (NSCLC) and to assess their potential as targets for therapeutic interventions. Multiple FGFR mRNA splice variants were coexpressed in NSCLC cells (n = 16) with predominance of FGFR1. Accordingly, both expression of a dominant-negative FGFR1 (dnFGFR1) IIIc-green fluorescent protein fusion protein and application of FGFR small-molecule inhibitors (SU5402 and PD166866) significantly reduced growth, survival, clonogenicity, and migratory potential of the majority of NSCLC cell lines. Moreover, dnFGFR1 expression completely blocked or at least significantly attenuated s.c. tumor formation of NSCLC cells in severe combined immunodeficient mice. Xenograft tumors expressing dnFGFR1 exhibited significantly reduced size and mitosis rate, enhanced cell death, and decreased tissue invasion. When FGFR inhibitors were combined with chemotherapy, antagonistic to synergistic in vitro anticancer activities were obtained depending on the application schedule. In contrast, simultaneous blockage of FGFR- and epidermal growth factor receptor-mediated signals exerted synergistic effects. In summary, FGFR-mediated signals in cooperation with those transmitted by epidermal growth factor receptor are involved in growth and survival of human NSCLC cells and should be considered as targets for combined therapeutic approaches.
Collapse
Affiliation(s)
- Hendrik Fischer
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Ninon Taylor
- Third Medical Department of the Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Sigrid Allerstorfer
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Michael Grusch
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Gudrun Sonvilla
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Klaus Holzmann
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Ulrike Setinek
- Institute of Pathology and Bacteriology, Hospital Baumgartner Höhe, Vienna, Austria and
| | - Leonilla Elbling
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Heidelinde Cantonati
- Institute of Pathology and Bacteriology, Hospital Baumgartner Höhe, Vienna, Austria and
| | - Bettina Grasl-Kraupp
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Christine Gauglhofer
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Brigitte Marian
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Michael Micksche
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| | - Walter Berger
- Institute of Cancer Research, Department of Medicine I, Medical University Vienna
| |
Collapse
|
45
|
Cotton LM, O'Bryan MK, Hinton BT. Cellular signaling by fibroblast growth factors (FGFs) and their receptors (FGFRs) in male reproduction. Endocr Rev 2008; 29:193-216. [PMID: 18216218 PMCID: PMC2528845 DOI: 10.1210/er.2007-0028] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/22/2007] [Accepted: 11/29/2007] [Indexed: 12/25/2022]
Abstract
The major function of the reproductive system is to ensure the survival of the species by passing on hereditary traits from one generation to the next. This is accomplished through the production of gametes and the generation of hormones that function in the maturation and regulation of the reproductive system. It is well established that normal development and function of the male reproductive system is mediated by endocrine and paracrine signaling pathways. Fibroblast growth factors (FGFs), their receptors (FGFRs), and signaling cascades have been implicated in a diverse range of cellular processes including: proliferation, apoptosis, cell survival, chemotaxis, cell adhesion, motility, and differentiation. The maintenance and regulation of correct FGF signaling is evident from human and mouse genetic studies which demonstrate that mutations leading to disruption of FGF signaling cause a variety of developmental disorders including dominant skeletal diseases, infertility, and cancer. Over the course of this review, we will provide evidence for differential expression of FGFs/FGFRs in the testis, male germ cells, the epididymis, the seminal vesicle, and the prostate. We will show that this signaling cascade has an important role in sperm development and maturation. Furthermore, we will demonstrate that FGF/FGFR signaling is essential for normal epididymal function and prostate development. To this end, we will provide evidence for the involvement of the FGF signaling system in the regulation and maintenance of the male reproductive system.
Collapse
Affiliation(s)
- Leanne M Cotton
- Department of Cell Biology, University of Virginia Health Sciences Center, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
46
|
Valta MP, Tuomela J, Bjartell A, Valve E, Väänänen HK, Härkönen P. FGF-8 is involved in bone metastasis of prostate cancer. Int J Cancer 2008; 123:22-31. [DOI: 10.1002/ijc.23422] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/29/2022]
|
47
|
Sahadevan K, Darby S, Leung HY, Mathers ME, Robson CN, Gnanapragasam VJ. Selective over-expression of fibroblast growth factor receptors 1 and 4 in clinical prostate cancer. J Pathol 2007; 213:82-90. [PMID: 17607666 DOI: 10.1002/path.2205] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/09/2022]
Abstract
Fibroblast growth factor receptors (FGFRs) mediate the tumourigenic effects of FGFs in prostate cancer. These receptors are therefore potential therapeutic targets in the development of inhibitors to this pathway. To identify the most relevant targets, we simultaneously investigated FGFR1-4 expression using a prostate cancer tissue microarray (TMA) and in laser capture microdissected (LCM) prostate epithelial cells. In malignant prostates (n = 138) we observed significant FGFR1 and FGFR4 protein over-expression in comparison with benign prostates (n = 58; p < 0.0001). FGFR1 was expressed at high levels in the majority of tumours (69% of grade 3 or less, 74% of grade 4 and 70% of grade 5), while FGFR4 was strongly expressed in 83% of grade 5 cancers but in only 25% of grade 1-3 cancers (p < 0.0001). At the transcript level we observed a similar pattern, with FGFR1 and FGFR4 mRNA over-expressed in malignant epithelial cells compared to benign cells (p < 0.0005 and p < 0.05, respectively). While total FGFR2 was increased in some cancers, there was no association between expression and tumour grade or stage. Transcript analysis, however, revealed a switch in the predominant isoform expressed from FGFR2IIIb to FGFR2IIIc among malignant epithelial cells. In contrast, protein and transcript expression of FGFR3 was very similar between benign and cancer biopsies. The functional effect of targeting FGFR4 in prostate cancer cells has not previously been investigated. In in vitro experiments, suppression of FGFR4 by RNA interference effectively blocked prostate cancer cell proliferation (p < 0.0001) and invasion (p < 0.001) in response to exogenous stimulation. This effect was evident regardless of whether the cells expressed the FGFR4 Arg388 or Gly388 allele. In parallel experiments, FGFR3 suppression had no discernible effect on cancer cell behaviour. These results suggest evidence of selective over-expression of FGFR1 and FGFR4 in clinical prostate cancer and support the notion of targeted inhibition of these receptors to disrupt FGF signalling.
Collapse
MESH Headings
- Case-Control Studies
- Cell Line, Tumor
- Cell Proliferation
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic
- Humans
- Immunohistochemistry
- Male
- Microdissection
- Microscopy, Confocal
- Oligonucleotide Array Sequence Analysis
- Polymorphism, Single Nucleotide
- Prostatic Neoplasms/genetics
- Prostatic Neoplasms/metabolism
- Protein Isoforms/genetics
- RNA Interference
- RNA, Small Interfering/pharmacology
- Receptor, Fibroblast Growth Factor, Type 1/analysis
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/analysis
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptor, Fibroblast Growth Factor, Type 3/analysis
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/metabolism
- Receptor, Fibroblast Growth Factor, Type 4/analysis
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Receptors, Fibroblast Growth Factor/analysis
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Transcription, Genetic
Collapse
Affiliation(s)
- K Sahadevan
- Urology Research Group, Northern Institute for Cancer Research, Medical School, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | | | | | | | | | | |
Collapse
|
48
|
Xu H, Hai GF, Xiang JZ, Yao CC, Zheng Q, Zhang QH, Hong H. Protective effect of non-mitogenic human acidic fibroblast growth factor on hepatocyte injury. Hepatol Res 2007; 37:836-44. [PMID: 17573954 DOI: 10.1111/j.1872-034x.2007.00131.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 02/08/2023]
Abstract
AIM To study whether non-mitogenic human acidic fibroblast growth factor (nm-haFGF) has protective effects on H(2)O(2)-induced hepatocyte injury in vitro and CCl(4)-induced hepatocyte injury in vivo. METHODS (i) HL-7702 hepatocytes were incubated with different concentrations of nm-haFGF for 12 h, and then the activity of lactate dehydrogenase (LDH) in culture medium was detected, and genomic DNA electrophoresis analysis was observed after being exposed to H(2)O(2) (8 mmol/L) for 4 h. Proximately, apoptotic rates and protein expressions of Bcl-2 and Bax of HL-7702 cell were detected after being exposed to H(2)O(2) (0.2 mmol/L) for 20 h. (ii) Being injected intraperitoneally with nm-haFGF, mice were treated with CCl(4) intraperitoneally to induce hepatic injury. Twenty-four hours later, serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were measured and histopathologic changes were evaluated. RESULTS (i) In vitro tests: LDH activities and apoptotic rates decreased, the protein expression of Bcl-2 increased and Baxdecreased in nm-haFGF-treated groups at the concentrations of 100 150 and 200 ng/mL, compared with that in the model control group, which was treated with H(2)O(2) alone. The genomic DNA remained nearly intact at the concentrations of 150 and 200 ng/mL. (ii) In vivo tests: serum ALT and AST in nm-haFGF-treated groups (10 mug/kg and 20 mug/kg) were much lower as compared to the model control group, which was treated with CCl(4) alone. Histological examination showed that nm-haFGF markedly ameliorated hepatocytes vacuolation, cloudy swelling and inflammatory cells infiltration induced by CCl(4). CONCLUSION nm-haFGF had protective effects against H(2)O(2)-induced hepatocyte injury in vitro and CCl(4)-induced acute liver injury in vivo.
Collapse
Affiliation(s)
- Hua Xu
- Tongji Medical College, Huazhong University of Science and Technology, Wuhan, and Pharmacy College, Ji-nan University, Guangzhou, China
| | | | | | | | | | | | | |
Collapse
|
49
|
Sales KJ, Boddy SC, Williams ARW, Anderson RA, Jabbour HN. F-prostanoid receptor regulation of fibroblast growth factor 2 signaling in endometrial adenocarcinoma cells. Endocrinology 2007; 148:3635-44. [PMID: 17478553 DOI: 10.1210/en.2006-1517] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
Prostaglandin (PG) F(2alpha) is a potent bioactive lipid in the female reproductive tract, and exerts its function after coupling with its heptahelical G-protein-coupled receptor [F-series-prostanoid (FP) receptor] to initiate cell signaling and target gene transcription. In the present study, we found elevated expression of fibroblast growth factor (FGF) 2, FGF receptor 1 (FGFR1), and FP receptor, colocalized within the neoplastic epithelial cells of endometrial adenocarcinomas. We investigated a role for PGF(2alpha)-FP receptor interaction in modulating FGF2 expression and signaling using an endometrial adenocarcinoma cell line stably expressing the FP receptor to the levels detected in endometrial adenocarcinomas (FPS cells) and endometrial adenocarcinoma tissue explants. PGF(2alpha)-FP receptor activation rapidly induced FGF2 mRNA expression, and elevated FGF2 protein expression and secretion into the culture medium in FPS cells and endometrial adenocarcinoma explants. The effect of PGF(2alpha) on the expression and secretion of FGF2 could be abolished by treatment of FPS cells and endometrial tissues with an FP receptor antagonist (AL8810) and inhibitor of ERK (PD98059). Furthermore, we have shown that FGF2 can promote the expression of FGF2 and cyclooxygenase-2, and enhance proliferation of endometrial adenocarcinoma cells via the FGFR1 and ERK pathways, thereby establishing a positive feedback loop to regulate neoplastic epithelial cell function in endometrial adenocarcinomas.
Collapse
MESH Headings
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adult
- Aged
- Autocrine Communication/physiology
- Cell Line, Tumor
- Cells, Cultured
- Cyclooxygenase 2/metabolism
- Dinoprost/metabolism
- Dinoprost/pharmacology
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Endometrium/cytology
- Endometrium/physiology
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Extracellular Signal-Regulated MAP Kinases/metabolism
- Feedback, Physiological/physiology
- Female
- Fibroblast Growth Factor 2/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/physiology
- Humans
- MAP Kinase Signaling System/physiology
- Middle Aged
- Paracrine Communication/physiology
- RNA, Messenger/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptors, Prostaglandin/genetics
- Receptors, Prostaglandin/metabolism
Collapse
Affiliation(s)
- Kurt J Sales
- Medical Research Council Human Reproductive Sciences Unit, The Queen's Medical Research Institute, 47 Little France Crescent, Edinburgh, Scotland EH16 4TJ, United Kingdom
| | | | | | | | | |
Collapse
|
50
|
Park SJ, Kim SJ, Rhee Y, Byun JH, Kim SH, Kim MH, Lee EJ, Lim SK. Fidgetin-like 1 gene inhibited by basic fibroblast growth factor regulates the proliferation and differentiation of osteoblasts. J Bone Miner Res 2007; 22:889-96. [PMID: 17352653 DOI: 10.1359/jbmr.070311] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED The FIGNL1 gene was proven to be a new subfamily member of ATPases associated with diverse cellular activities (AAA proteins). In this in vitro study, the AAA proteins inhibited osteoblast proliferation and stimulated osteoblast differentiation. We showed that FIGNL1 may play some regulatory role in osteoblastogenesis. INTRODUCTION The fidgetin-like 1 (FIGNL1) gene encodes a new subfamily member of ATPases associated with diverse cellular activities (AAA proteins). Although the FIGNL1 protein localizes to both the nucleus and cytoplasm, the function of FIGNL1 remains unknown. In a previous study, we identified several genes that mediate the anabolic effects of basic fibroblast growth factor (bFGF) on bone by using microarray data. FIGNL1 was one of the genes that downregulated >2-fold in MC3T3-E1 cells after treatment with bFGF. Therefore, this study was aimed to identify and confirm the function of FIGNL1 on osteoblastogenesis. MATERIALS AND METHODS We examined the effect of the FIGNL1 gene on proliferation, differentiation, and apoptosis in mouse osteoblast cells (MC3T3-E1 and mouse primary calvarial cells) using flow cytometry, RT-PCR, cell proliferation assay, and cell death assay. MC3T3-E1 cells and mouse calvarial cells were transfected with small interfering RNA (siRNA) directed against the FIGNL1 or nontargeting control siRNA and examined by cell proliferation and cell death assays. Also, FIGNL1 was fused to enhance green fluorescent protein (EGFP), and the EGFP-fused protein was transiently expressed in MC3T3-E1 cells. RESULTS Reduced expression of FIGNL1 by bFGF and TGF-beta1 treatment was verified by RT-PCR analysis. Overexpression of FIGNL1 reduced the proliferation of MC3T3-E1 and calvarial cells, more than the mock transfected control cells did. In contrast, siFIGNL1 transfection significantly increased the proliferation of osteoblasts, whereas overexpression of FIGNL1 did not seem to alter apoptosis in osteoblasts. Meanwhile, overexpression of FIGNL1 enhanced the mRNA expression of alkaline phosphatase (ALP) and osteocalcin (OCN) in osteoblasts. In contrast, siFIGNL1 decreased the expression of ALP and OCN. A pEGFP-FIGNL1 transfected into MCT3-E1 cells had an initially ubiquitous distribution and rapidly translocated to the nucleus 1 h after bFGF treatment. CONCLUSIONS From these results, we proposed that FIGNL1, a subfamily member of the AAA family of proteins, might play some regulatory role in osteoblast proliferation and differentiation. Further analyses of FIGNL1 will be needed to better delineate the mechanisms contributing to the inhibition of proliferation and stimulation of osteoblast differentiation.
Collapse
Affiliation(s)
- Su Jin Park
- Brain Korea 21 Project for Medical Science, College of Medicine, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|