1
|
Li HZ, Zhang JL, Yuan DL, Xie WQ, Ladel CH, Mobasheri A, Li YS. Role of signaling pathways in age-related orthopedic diseases: focus on the fibroblast growth factor family. Mil Med Res 2024; 11:40. [PMID: 38902808 PMCID: PMC11191355 DOI: 10.1186/s40779-024-00544-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Fibroblast growth factor (FGF) signaling encompasses a multitude of functions, including regulation of cell proliferation, differentiation, morphogenesis, and patterning. FGFs and their receptors (FGFR) are crucial for adult tissue repair processes. Aberrant FGF signal transduction is associated with various pathological conditions such as cartilage damage, bone loss, muscle reduction, and other core pathological changes observed in orthopedic degenerative diseases like osteoarthritis (OA), intervertebral disc degeneration (IVDD), osteoporosis (OP), and sarcopenia. In OA and IVDD pathologies specifically, FGF1, FGF2, FGF8, FGF9, FGF18, FGF21, and FGF23 regulate the synthesis, catabolism, and ossification of cartilage tissue. Additionally, the dysregulation of FGFR expression (FGFR1 and FGFR3) promotes the pathological process of cartilage degradation. In OP and sarcopenia, endocrine-derived FGFs (FGF19, FGF21, and FGF23) modulate bone mineral synthesis and decomposition as well as muscle tissues. FGF2 and other FGFs also exert regulatory roles. A growing body of research has focused on understanding the implications of FGF signaling in orthopedic degeneration. Moreover, an increasing number of potential targets within the FGF signaling have been identified, such as FGF9, FGF18, and FGF23. However, it should be noted that most of these discoveries are still in the experimental stage, and further studies are needed before clinical application can be considered. Presently, this review aims to document the association between the FGF signaling pathway and the development and progression of orthopedic diseases. Besides, current therapeutic strategies targeting the FGF signaling pathway to prevent and treat orthopedic degeneration will be evaluated.
Collapse
Affiliation(s)
- Heng-Zhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing-Lve Zhang
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Dong-Liang Yuan
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Medicine Central, South University, Changsha, 410083, China
| | - Wen-Qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | | | - Ali Mobasheri
- Faculty of Medicine, Research Unit of Health Sciences and Technology, University of Oulu, 90014, Oulu, Finland.
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, 08406, Vilnius, Lithuania.
- Department of Rheumatology and Clinical Immunology, Universitair Medisch Centrum Utrecht, Utrecht, 3508, GA, the Netherlands.
- Department of Joint Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, B-4000, Liège, Belgium.
| | - Yu-Sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
2
|
Li J, Hui Y, Xu Z, Tan J, Yin K, Kuang L, Tang Y, Wei J, Zhong Q, Zheng T. Non-canonical function of DPP4 promotes cognitive impairment through ERp29-associated mitochondrial calcium overload in diabetes. iScience 2023; 26:106271. [PMID: 36936785 PMCID: PMC10014273 DOI: 10.1016/j.isci.2023.106271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 01/15/2023] [Accepted: 02/19/2023] [Indexed: 03/02/2023] Open
Abstract
DPP4 has been shown to induce diabetes-associated mitochondrial dysfunction and cognitive impairment through its non-canonical function. Here, we report that enhanced DPP4 expression in diabetes contributes to IP3R2-mediated mitochondria-associated ER membrane (MAM) formation, mitochondria calcium overload, and cognitive impairment, and its knockdown showed opposite effects. Mechanistically, DPP4 binds to PAR2 in hippocampal neurons and activates ERK1/2/CEBPB signaling, which upregulates ERp29 expression and promotes its binding to IP3R2, thereby inhibiting IP3R2 degradation and promoting MAM formation, mitochondria calcium overload, and cognitive impairment. Meanwhile, targeting DPP4-mediated PAR2/ERK1/2/CEBPB/ERp29 signaling achieved satisfactory therapeutic effects on MAM formation, mitochondria calcium overload, and cognitive impairment. Notably, DPP4 activates this pathway in an enzymatic activity-independent manner, suggesting the non-canonical role of DPP4 in the pathogenesis of mitochondria calcium overload and cognitive impairment in diabetes. Together, these results identify DPP4-mediated PAR2/ERK1/2/CEBPB/ERp29 signaling as a promising therapeutic target for the treatment of cognitive impairment in type 2 diabetes.
Collapse
Affiliation(s)
- Jiaxiu Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Ya Hui
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Zhiqiang Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Kai Yin
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Liuyu Kuang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Yunyun Tang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Junjie Wei
- Lingui Clinical Medical College, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Qiongsui Zhong
- Lingui Clinical Medical College, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
| | - Tianpeng Zheng
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541199, P. R. China
- Corresponding author
| |
Collapse
|
3
|
Li F, Zhang H, Wang Y, Yao Z, Xie K, Mo Q, Fan Q, Hou L, Deng F, Tan W. FGFBP1 as a potential biomarker predicting bacillus Calmette–Guérin response in bladder cancer. Front Immunol 2022; 13:954836. [PMID: 36119059 PMCID: PMC9478507 DOI: 10.3389/fimmu.2022.954836] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/11/2022] [Indexed: 11/30/2022] Open
Abstract
Accurate prediction of Bacillus Calmette–Guérin (BCG) response is essential to identify bladder cancer (BCa) patients most likely to respond sustainably, but no molecular marker predicting BCG response is available in clinical routine. Therefore, we first identified that fibroblast growth factor binding protein 1 (FGFBP1) was upregulated in failures of BCG therapy, and the increased FGFBP1 had a poor outcome for BCa patients in the E-MTAB-4321 and GSE19423 datasets. These different expression genes associated with FGFBP1 expression are mainly involved in neutrophil activation, neutrophil-mediated immunity, and tumor necrosis factor-mediated signal pathways in biological processes. A significant positive correlation was observed between FGFBP1 expression and regulatory T-cell (Treg) infiltration by the Spearman correlation test in the BCG cohort (r = 0.177) and The Cancer Genome Atlas (TCGA) cohort (r = 0.176), suggesting that FGFBP1 may influence the response of BCa patients to BCG immunotherapy through immune escape. Though FGFBP1 expression was positively correlated with the expressions of PD-L1, CTLA4, and PDCD1 in TCGA cohort, a strong association between FGFBP1 and PD-L1 expression was only detected in the BCG cohort (r = 0.750). Furthermore, elevated FGFBP1 was observed in BCa cell lines and tissues in comparison to corresponding normal controls by RT-qPCR, Western blotting, and immunohistochemical staining. Increased FGFBP1 was further detected in the failures than in the responders by immunohistochemical staining. Notably, FGFBP1 is positively associated with PD-L1 expression in BCa patients with BCG treatment. To sum up, FGFBP1 in BCa tissue could be identified as a promising biomarker for the accurate prediction of BCG response in BCa.
Collapse
Affiliation(s)
- Fei Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Henghui Zhang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yu Wang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhihao Yao
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kunfeng Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qixin Mo
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin Fan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Lina Hou
- Department of Healthy Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Wanlong Tan, ; Fan Deng, ; Lina Hou,
| | - Fan Deng
- Department of Cell Biology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
- *Correspondence: Wanlong Tan, ; Fan Deng, ; Lina Hou,
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Wanlong Tan, ; Fan Deng, ; Lina Hou,
| |
Collapse
|
4
|
Chen X, Miao M, Zhou M, Chen J, Li D, Zhang L, Sun A, Guan M, Wang Z, Liu P, Zhang S, Zha X, Fan X. Poly-L-arginine promotes asthma angiogenesis through induction of FGFBP1 in airway epithelial cells via activation of the mTORC1-STAT3 pathway. Cell Death Dis 2021; 12:761. [PMID: 34341336 PMCID: PMC8329163 DOI: 10.1038/s41419-021-04055-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 01/21/2023]
Abstract
Angiogenesis is a key characteristic of asthma airway remodeling. By releasing cationic granule proteins, such as major basic protein (MBP), activated eosinophils play a prominent role in asthma, but the underlying mechanisms are still not fully understood. In this study, we demonstrated that fibroblast growth factor-binding protein 1 (FGFBP1) was dramatically upregulated in airway epithelial cell lines treated by poly-L-arginine (PLA), a mimic of MBP. Elevated FGFBP1 expression was also detected in asthma clinical samples, as well as in ovalbumin (OVA)-induced chronic asthma mouse models. PLA enhanced FGFBP1 expression through activation of the mechanistic target of rapamycin complex 1-signal transducer and activator of transcription 3 (mTORC1-STAT3) signaling pathway. STAT3 transactivated FGFBP1 by directly binding to the promoter of the FGFBP1 gene. Furthermore, we identified that FGFBP1 secreted by PLA-treated airway epithelial cells served as a proangiogenesis factor. Lastly, we found the mTORC1-STAT3-FGFBP1 signaling pathway was activated in an OVA-induced chronic asthma model with airway remodeling features. Rapamycin treatment alleviated respiratory symptoms and reduced angiogenesis in asthmatic mice. Therefore, activation of the mTORC1-STAT3-FGFBP1 pathway in the airway epithelium contributes to the progress of angiogenesis and should be targeted for the treatment of asthma.
Collapse
Affiliation(s)
- Xu Chen
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Hefei, China.,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China.,Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Manli Miao
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Hefei, China.,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China
| | - Meng Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Jie Chen
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Hefei, China.,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China
| | - Dapeng Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Ling Zhang
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Hefei, China.,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China
| | - Anjiang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Minglong Guan
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Geriatric Institute, Hefei, China.,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China
| | - Zixi Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Ping Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Shengquan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China
| | - Xiaojun Zha
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, China.
| | - Xiaoyun Fan
- Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, China. .,Anhui Geriatric Institute, Hefei, China. .,Key Lab of Geriatric Molecular Medicine of Anhui Province, Hefei, China.
| |
Collapse
|
5
|
Muscle Fibers Secrete FGFBP1 to Slow Degeneration of Neuromuscular Synapses during Aging and Progression of ALS. J Neurosci 2017; 37:70-82. [PMID: 28053031 DOI: 10.1523/jneurosci.2992-16.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 01/13/2023] Open
Abstract
The identity of muscle secreted factors critical for the development and maintenance of neuromuscular junctions (NMJs) remains largely unknown. Here, we show that muscle fibers secrete and concentrate the fibroblast growth factor binding protein 1 (FGFBP1) at NMJs. Although FGFBP1 expression increases during development, its expression decreases before NMJ degeneration during aging and in SOD1G93A mice, a mouse model for amyotrophic lateral sclerosis (ALS). Based on these findings, we examined the impact of deleting FGFBP1 on NMJs. In the absence of FGFBP1, NMJs exhibit structural abnormalities in developing and middle age mice. Deletion of FGFBP1 from SOD1G93A mice also accelerates NMJ degeneration and death. Based on these findings, we sought to identify the mechanism responsible for decreased FGFBP1 in stressed skeletal muscles. We show that FGFBP1 expression is inhibited by increased accumulation of the transforming growth factor-β1 (TGF-β1) in skeletal muscles and at their NMJs. These findings suggest that targeting the FGFBP1 and TGF-β1 signaling axis holds promise for slowing age- and disease-related degeneration of NMJs. SIGNIFICANCE STATEMENT The neuromuscular junction (NMJ) is critical for all voluntary movement. Its malformation during development and degeneration in adulthood impairs motor function. Therefore, it is important to identify factors that function to maintain the structural integrity of NMJs. We show that muscle fibers secrete and concentrate the fibroblast growth factor binding protein 1 (FGFBP1) at NMJs. However, FGFBP1 expression decreases in skeletal muscles during aging and before NMJ degeneration in SOD1G93A mice, a mouse model for amyotrophic lateral sclerosis. We show that transforming growth factor-β1 is responsible for the decreased levels of FGFBP1. Importantly, we demonstrate critical roles for FGFBP1 at NMJs in developing, aging and SOD1G93A mice.
Collapse
|
6
|
Huang W, Chen Z, Shang X, Tian D, Wang D, Wu K, Fan D, Xia L. Sox12, a direct target of FoxQ1, promotes hepatocellular carcinoma metastasis through up-regulating Twist1 and FGFBP1. Hepatology 2015; 61:1920-33. [PMID: 25704764 DOI: 10.1002/hep.27756] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 02/14/2015] [Indexed: 12/29/2022]
Abstract
UNLABELLED Metastasis is the main reason for high recurrence and poor survival of hepatocellular carcinoma (HCC) after curative resection. However, the molecular mechanism underlying HCC metastasis remains unclear. Here, we report on a novel function of SRY (sex determining region Y)-box 12 (Sox12), a member of the SYR-related high mobility group box family proteins, in promoting HCC metastasis. Overexpression of Sox12 was significantly correlated with loss of tumor encapsulation, microvascular invasion, and a higher tumor-nodule-metastasis (TNM) stage and indicated poor prognosis in human HCC patients. Sox12 expression was an independent and significant risk factor for recurrence and reduced survival after curative resection. Overexpression of Sox12 induced epithelial-mesenchymal transition by transactivating Twist1 expression. Down-regulation of Twist1 decreased Sox12-enhanced HCC migration, invasion, and metastasis, whereas up-regulation of Twist1 rescued the decreased migration, invasion, and metastasis induced by Sox12 knockdown. Additionally, serial deletion, site-directed mutagenesis, and chromatin immunoprecipitation assays showed that fibroblast growth factor binding protein 1 (FGFBP1) was a direct transcriptional target of Sox12. Knockdown of FGFBP1 decreased Sox12-mediated HCC invasion and metastasis, whereas overexpression of FGFBP1 rescued the decreased invasion and metastasis induced by Sox12 knockdown. Furthermore, forkhead box Q1 (FoxQ1) directly bound to the Sox12 promoter and transactivated its expression, which contributed to Sox12 overexpression in human HCC. Knockdown of Sox12 dramatically decreased FoxQ1-mediated HCC metastasis. In two independent cohorts of human HCC tissues, Sox12 expression was positively correlated with Twist1, FGFBP1, and FoxQ1 expression, and patients with positive coexpression of Sox12/Twist1, Sox12/FGFBP1, or FoxQ1/Sox12 were associated with poorer prognosis. CONCLUSION Up-regulated Sox12 induced by FoxQ1 promotes HCC invasion and metastasis by transactivating Twist1 and FGFBP1 expression. Thus, our study implicates Sox12 as a potential prognostic biomarker and a novel therapeutic target for HCC.
Collapse
Affiliation(s)
- Wenjie Huang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China.,Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Zhangqian Chen
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xin Shang
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Daowen Wang
- Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Kaichun Wu
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Daiming Fan
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Limin Xia
- State Key Laboratory of Cancer Biology and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, People's Republic of China.,Department of Gastroenterology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| |
Collapse
|
7
|
Masha'our RS, Heinrich R, Garzozi HJ, Perlman I. Acetylcholinesterase (AChE) is an important link in the apoptotic pathway induced by hyperglycemia in Y79 retinoblastoma cell line. Front Mol Neurosci 2012; 5:69. [PMID: 22685426 PMCID: PMC3368359 DOI: 10.3389/fnmol.2012.00069] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 05/14/2012] [Indexed: 01/26/2023] Open
Abstract
Acetylcholinesterase (AChE) expression was found to be induced in the mammalian CNS, including the retina, by different types of stress leading to cellular apoptosis. Here, we tested possible involvement of AChE in hyperglycemia-induced apoptosis in a retinal cell line. Y79 retinoblastoma cells were incubated in starvation media (1% FBS and 1 mg/ml glucose) for 16–24 h, and then exposed to hyperglycemic environment by raising extracellular glucose concentrations to a final level of 3.5 mg/ml or 6 mg/ml. Similar levels of mannitol were used as control for hyperosmolarity. Cells were harvested at different time intervals for analysis of apoptosis and AChE protein expression. Apoptosis was detected by the cleavage of Poly ADP-ribose polymerase (PARP) using western blot, and by Terminal deoxynucleotidyl-transferase-mediated dUTP nick-end-labeling (TUNEL) assay. AChE protein expression and activity was detected by western blot and by the Karnovsky and Roots method, respectively. MissionTM shRNA for AChE was used to inhibit AChE protein expression. Treating Y79 cells with 3.5 mg/ml of glucose, but not with 3.5 mg/ml mannitol, induced apoptosis which was confirmed by TUNEL assay and by cleavage of PARP. A part of the signaling pathway accompanying the apoptotic process involved up-regulation of the AChE-R variant and an N-extended AChE variant as verified at the mRNA and protein level. Inhibition of AChE protein expression by shRNA protected Y79 cell from entering the apoptotic pathway. Our data suggest that expression of an N-extended AChE variant, most probably an R isoform, is involved in the apoptotic pathway caused by hyperglycemia in Y79 cells.
Collapse
|
8
|
Zheng HQ, Zhou Z, Huang J, Chaudhury L, Dong JT, Chen C. Krüppel-like factor 5 promotes breast cell proliferation partially through upregulating the transcription of fibroblast growth factor binding protein 1. Oncogene 2009; 28:3702-13. [PMID: 19668233 DOI: 10.1038/onc.2009.235] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Krüppel-like factor 5 (KLF5) is a zinc-finger transcription factor promoting cell proliferation, cell-cycle progression and survival. A high expression level of KLF5 mRNA has been shown to be associated with shorter breast cancer patient survival. However, the mechanism of KLF5 action in breast cancer is still not clear. In this study, we found that both KLF5 and its downstream gene fibroblast growth factor binding protein 1 (FGF-BP) are co-expressed in breast cell lines and primary tumors. Manipulation of the KLF5 expression can positively regulate the FGF-BP mRNA and protein levels in multiple breast cell lines. In addition, the secreted FGF-BP protein in the conditional medium is also regulated by KLF5. Furthermore, we demonstrated that KLF5 binds and activates the FGF-BP promoter through a GC box by luciferase reporter, oligo pull down and chromatin immunoprecipitation (ChIP) assays. When FGF-BP is depleted by siRNA, KLF5 fails to promote cell proliferation in MCF10A, SW527 and TSU-Pr1. We further demonstrated that overexpression or addition of FGF-BP rescues the KLF5-knockdown-induced growth arrest in MCF10A cells. Finally, KLF5 significantly promotes MCF7 breast cancer cell xenograft growth in athymic nude mice. These findings suggest that KLF5 may promote breast cancer cell proliferation at least partially through directly activating the FGF-BP mRNA transcription. Understanding the mechanism of KLF5 action in breast cancer may result in useful diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- H-Q Zheng
- The Center for Cell Biology and Cancer Research, Albany Medical College, Albany, NY, USA
| | | | | | | | | | | |
Collapse
|
9
|
Lee JY, Han CY, Yang JW, Smith C, Kim SK, Lee EYHP, Kim SG, Kang KW. Induction of glutathione transferase in insulin-like growth factor type I receptor-overexpressed hepatoma cells. Mol Pharmacol 2007; 72:1082-93. [PMID: 17615245 DOI: 10.1124/mol.107.038174] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Insulin-like growth factor type I receptor (IGF-IR) is frequently overexpressed in human hepatocellular carcinoma cells (HCC), and this overexpression has been correlated with increased tumor growth. The protective response of HCC to reactive oxygen species (ROS) produced by chemotherapeutic agents is mediated with the induction of phase II detoxifying genes including glutathione transferase (GST). To understand the roles of IGF-IR overexpression in HCC in terms of its detoxifying effect on ROS and conferred resistance to chemotherapy, we analyzed whether IGF-IR overexpressions affect IGF-1-inducible GST expression. GSTalpha was induced by exposure to IGF-1 in IGF-IR cells but not in cells expressing normal levels of IGF-IR. Furthermore, IGF-IR-overexpressed HCCs (IR-HCC) are more resistant to doxorubicin than control HCC cells, which was associated with the increased GST induction by IGF-1. Molecular analyses using GSTA2 promoter supported the involvement of xenobiotic response element (XRE) in GSTalpha induction. IGF-1 caused the nuclear translocation of CCAAT/enhancer-binding protein beta (C/EBPbeta), which might be responsible for XRE activation. In addition, IGF-1 increased the activities of phosphatidylinositol 3-kinase (PI3-kinase) and extracellular signal-regulated kinase in IR-HCCs. Moreover, the inhibition of PI3-kinase completely abolished the nuclear translocation of C/EBPbeta and the up-regulation of GSTalpha protein in IR-HCC treated with IGF-1. However, specific inhibitors against extracellular signal-regulated kinase, c-Jun N-terminal kinase, or p38 kinase did not alter IGF-1-inducible GSTalpha expression. These results provide evidence that one of the pathological consequences of IGF-IR overexpression in HCCs is the potentiation of GSTalpha inducibility by IGF-1. Moreover, this potentiation of GST may be associated with decreased susceptibility to chemotherapeutic agents such as doxorubicin.
Collapse
Affiliation(s)
- Jeong Yong Lee
- College of Pharmacy, Chosun University, 375 Seosuk-dong, Dong-gu, Gwangju 501-759, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Khanna H, Akimoto M, Siffroi-Fernandez S, Friedman JS, Hicks D, Swaroop A. Retinoic acid regulates the expression of photoreceptor transcription factor NRL. J Biol Chem 2006; 281:27327-34. [PMID: 16854989 PMCID: PMC1592579 DOI: 10.1074/jbc.m605500200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
NRL (neural retina leucine zipper) is a key basic motif-leucine zipper (bZIP) transcription factor, which orchestrates rod photoreceptor differentiation by activating the expression of rod-specific genes. The deletion of Nrl in mice results in functional cones that are derived from rod precursors. However, signaling pathways modulating the expression or activity of NRL have not been elucidated. Here, we show that retinoic acid (RA), a diffusible factor implicated in rod development, activates the expression of NRL in serum-deprived Y79 human retinoblastoma cells and in primary cultures of rat and porcine photoreceptors. The effect of RA is mimicked by TTNPB, a RA receptor agonist, and requires new protein synthesis. DNaseI footprinting and electrophoretic mobility shift assays (EMSA) using bovine retinal nuclear extract demonstrate that RA response elements (RAREs) identified within the Nrl promoter bind to RA receptors. Furthermore, in transiently transfected Y79 and HEK293 cells the activity of Nrl-promoter driving a luciferase reporter gene is induced by RA, and this activation is mediated by RAREs. Our data suggest that signaling by RA via RA receptors regulates the expression of NRL, providing a framework for delineating early steps in photoreceptor cell fate determination.
Collapse
Affiliation(s)
- Hemant Khanna
- From the Departments of Ophthalmology and Visual Sciences and
| | - Masayuki Akimoto
- From the Departments of Ophthalmology and Visual Sciences and
- Translational Research Center, Kyoto University Hospital, Sakyo-ku, Kyoto 606-8507, Japan, the
| | | | | | - David Hicks
- Laboratory of Neurobiological Rhythms, UMR CNRS 7518, Centre de Neurochimie, 67084 Strasbourg, France
| | - Anand Swaroop
- From the Departments of Ophthalmology and Visual Sciences and
- Human Genetics, University of Michigan, Ann Arbor, Michigan 48105, the
- Harold F. Falls Collegiate Professor and a recipient of RPB Senior Scientific Investigator award. To whom correspondence should be addressed: Dept. of Ophthalmology and Visual Sciences, W. K. Kellogg Eye Center, University of Michigan, 1000 Wall St., Ann Arbor, MI 48105. Tel.: 734-763-3731; Fax: 734-647-0228; E-mail:
| |
Collapse
|
11
|
Briones VR, Chen S, Riegel AT, Lechleider RJ. Mechanism of fibroblast growth factor-binding protein 1 repression by TGF-beta. Biochem Biophys Res Commun 2006; 345:595-601. [PMID: 16690027 DOI: 10.1016/j.bbrc.2006.04.052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Accepted: 04/11/2006] [Indexed: 01/04/2023]
Abstract
Transforming growth factor-beta (TGF-beta) is the prototypical member of a family of growth factors that play important roles in normal development and human diseases. We identified the gene for fibroblast growth factor-binding protein 1 (FGF-BP1) as being significantly repressed following TGF-beta treatment. FGF-BP1 is an extracellular matrix bound protein that enhances fibroblast growth factor (FGF) signaling. We demonstrate here that TGF-beta signaling significantly represses FGF-BP1 expression in mesenchymal and neural crest cells undergoing in vitro smooth muscle differentiation. Analysis of the downstream signaling pathways shows that Smad2/3 are crucial for efficient FGF-BP1 repression by TGF-beta. Furthermore, we identified a novel element in the region from -785 to -782 bp of the FGF-BP1 promoter, which represents a known binding site for Hypermethylation in Cancer-1 (Hic-1), necessary for repression of FGF-BP1 by TGF-beta. These data define the molecular mechanism of transcriptional repression of an important target of TGF-beta signaling during angiogenesis.
Collapse
Affiliation(s)
- Victorino R Briones
- Department of Cell Biology, Georgetown University Medical Center, Washington, DC 20057, USA
| | | | | | | |
Collapse
|
12
|
Abuharbeid S, Czubayko F, Aigner A. The fibroblast growth factor-binding protein FGF-BP. Int J Biochem Cell Biol 2005; 38:1463-8. [PMID: 16324873 DOI: 10.1016/j.biocel.2005.10.017] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2005] [Revised: 10/21/2005] [Accepted: 10/26/2005] [Indexed: 12/25/2022]
Abstract
Fibroblast growth factors (FGFs) are important regulators of cell migration, proliferation and differentiation, e.g., during embryogenesis and wound healing, and under several pathological conditions including tumor growth and tumor angiogenesis. Since heparin-binding FGFs are tightly bound to heparansulfate proteoglycans, and therefore, trapped in the extracellular matrix, their release through the action of an FGF-binding protein (FGF-BP) is one of the critical steps in FGF bioactivation. FGF-BP expression is highly tissue specific and strictly regulated through different promoter elements. Besides its role in embryogenesis and wound healing, FGF-BP is upregulated in several tumors and it is associated especially with early stages of tumor formation, where angiogenesis plays a critical role. Concomitantly, in several mouse tumor models, targeting of FGF-BP by ribozymes or RNA interference (RNAi) abolishes or reduces tumor growth and tumor angiogenesis. This indicates that FGF-BP can be rate-limiting for tumor growth and serves as an angiogenic switch molecule, and that it represents an increasingly promising target molecule in anti-tumor therapy.
Collapse
Affiliation(s)
- Shaker Abuharbeid
- Department of Pharmacology and Toxicology, Philipps-University School of Medicine, Marburg, Germany
| | | | | |
Collapse
|
13
|
Berg T, Didon L, Barton J, Andersson O, Nord M. Glucocorticoids increase C/EBPbeta activity in the lung epithelium via phosphorylation. Biochem Biophys Res Commun 2005; 334:638-45. [PMID: 16009338 DOI: 10.1016/j.bbrc.2005.06.146] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2005] [Accepted: 06/24/2005] [Indexed: 11/29/2022]
Abstract
Glucocorticoids are widely prescribed anti-inflammatory drugs used for the treatment of many inflammatory lung disorders. However, much still remains unknown about their molecular mechanisms of action. We have previously shown that glucocorticoid-induced transcription in the lung epithelial cell line NCI-H441 is mediated via C/EBP sites in the promoters of target genes, and is likely to involve the transcription factors C/EBPbeta and C/EBPdelta. Here, we report that C/EBPbeta is the most active C/EBP-factor in both human and mouse lung epithelium and that glucocorticoids induce DNA binding of C/EBPbeta in cultured primary mouse lung epithelial cells. Mechanistic studies in H441 cells revealed that glucocorticoids, acting via the glucocorticoid receptor, increase C/EBPbeta binding starting 10 min after stimulation. The mechanism is independent of de novo protein synthesis and involves phosphorylation of C/EBPbeta at Thr(235). Together this shows that glucocorticoids increase DNA-binding activity of C/EBPbeta via post-translational mechanism(s) involving phosphorylation.
Collapse
Affiliation(s)
- Tove Berg
- Department of Medicine, Division for Respiratory Medicine, Karolinska Institute, Lung Research Laboratory, Karolinska University Hospital, Solna, Sweden
| | | | | | | | | |
Collapse
|
14
|
Kurtz A, Aigner A, Cabal-Manzano RH, Butler RE, Hood DR, Sessions RB, Czubayko F, Wellstein A. Differential regulation of a fibroblast growth factor-binding protein during skin carcinogenesis and wound healing. Neoplasia 2005; 6:595-602. [PMID: 15548369 PMCID: PMC1531664 DOI: 10.1593/neo.04214] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The initiation of premalignant lesions is associated with subtle cellular and gene expression changes. Here we describe a severe combined immunodeficiency mouse xenograft model with human adult skin and compare chemical carcinogenesis and wound healing. We focus on a secreted binding protein for fibroblast growth factors (FGF-BP) that enhances the activity of locally stored FGFs and is expressed at high levels in human epithelial cancers. Carcinogen treatment of murine skin induced papilloma within 6 weeks, whereas the human skin grafts displayed no obvious macroscopic alterations. Microscopic studies of the human skin, however, showed p53-positive keratinocytes in the epidermis, increased angiogenesis in the dermis of the treated skin, enhanced proliferation of keratinocytes in the basal layer, and an increase of FGF-BP protein and mRNA expression. In contrast, after surgical wounding of human skin grafts or of mouse skin, FGF-BP expression was upregulated within a few hours and returned to control levels after 2 days with wound closure. Enhanced motility of cultured keratinocytes and dermal fibroblasts by FGF-BP supports a role in wound healing. We conclude that adult human skin xenografts can be used to identify early molecular events during malignant transformation as well as transient changes during wound healing.
Collapse
Affiliation(s)
- Andreas Kurtz
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA
| | - Achim Aigner
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA
| | - Rafael H Cabal-Manzano
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA
| | - Robert E Butler
- Lombardi Cancer Center, Department of Otolaryngology/Head and Neck Surgery, Georgetown University, Washington, DC 20057, USA
| | - Dozier R Hood
- Lombardi Cancer Center, Department of Otolaryngology/Head and Neck Surgery, Georgetown University, Washington, DC 20057, USA
| | - Roy B Sessions
- Lombardi Cancer Center, Department of Otolaryngology/Head and Neck Surgery, Georgetown University, Washington, DC 20057, USA
| | - Frank Czubayko
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA
| | - Anton Wellstein
- Lombardi Cancer Center, Department of Oncology, Georgetown University, Washington, DC 20057, USA
| |
Collapse
|
15
|
Ziesche R, Petkov V, Lambers C, Erne P, Block LH. The calcium channel blocker amlodipine exerts its anti¬proliferative action via p21
(Waf1/Cip1)
gene activation. FASEB J 2004; 18:1516-23. [PMID: 15466360 DOI: 10.1096/fj.04-1662com] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Proliferation of vascular smooth muscle cells (VSMC) contributes to the progression of atherosclerotic plaques. Calcium channel blockers have been shown to reduce VSMC proliferation, but the underlying molecular mechanism remains unclear. p21(Waf1/Cip1) is a potent inhibitor of cell cycle progression. Here, we demonstrate that amlodipine (10(-6) to 10(-8) M) activates de novo synthesis of p21(Waf1/Cip1) in vitro. We show that amlodipine-dependent activation of p21(Waf1/Cip1) involves the action of the glucocorticoid receptor (GR) and C/EBP-alpha. The underlying pathway apparently involves the action of mitogen-activated protein kinase or protein kinase C, but not of extracellular signal-related kinase or changes of intracellular calcium. Amlodipine-induced p21(Waf1/Cip1) promoter activity and expression were abrogated by C/EBP-alpha antisense oligonucleotide or by the GR antagonist RU486. Amlodipine-dependent inhibition of cell proliferation was partially reversed by RU486 at 10(-8) M (58%+/-29%), antisense oligonucleotides targeting C/EBP-alpha (91%+/-26%), or antisense mRNAs targeting p21(Waf1/Cip1) (96%+/-32%, n=6); scrambled antisense oligonucleotides or those directed against C/EBP-beta were ineffective. The data suggest that the anti-proliferative action of amlodipine is achieved by induction of the p21 (Waf1/Cip1) gene, which may explain beneficial covert effects of this widely used cardiovascular therapeutic drug beyond a more limited role as a vascular relaxant.
Collapse
Affiliation(s)
- Rolf Ziesche
- Department of Internal Medicine IV, University of Vienna, Austria
| | | | | | | | | |
Collapse
|
16
|
Orlev LN, Ehud B, Tamar BG, Orit SA, Yoel K, Witz IP. Does the dual-specificity MAPK phosphatase Pyst2-L lead a monogamous relationship with the Erk2 protein? Immunol Lett 2004; 92:149-56. [PMID: 15081539 DOI: 10.1016/j.imlet.2003.11.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2003] [Accepted: 11/03/2003] [Indexed: 02/02/2023]
Abstract
The dual-specificity phosphatase Pyst2-L was found to be highly expressed in leukocytes derived from AML and ALL patients as well as in certain other solid tumors and lymphoblastoid cell lines. Recently, by use of the 5'-RNA ligation-mediated rapid amplification of cDNA ends (5'-RLM-RACE) technique, we sequenced and cloned the entire open reading frame (ORF) of Pyst2-L. In the present study we determined the effect of exogenous overexpression on Erk1/2 phosphorylation. It was demonstrated that overexpression of this phosphatase in HEK293 cells reduced the basal levels of phospho-Erk1/2 as compared to the same cells transfected with the wild-type vector. This reduction was concomitant with a growth retardation of the Pyst2-L-transfected cells. Treating Pyst2-L transfected cells with known activators of the MAPK signaling cascade such as TPA or stimulating them by serum, it was demonstrated that the up regulation of phospho-Erk1/2, caused by these activators, was only partially suppressed by the over expression of the Pyst2-L phosphatase in these cells. These results together with our previous ones showing that the TPA-induced up regulation of Pyst2-L mRNA was only partially inhibited by the use of a specific Mek1/2 inhibitor, lead us to ask whether the Pyst2-L phosphatase has a monogamous relationship with the Erk2 protein. To answer this question, we employed the pull-down method and showed that in addition to phospho-Erk1/2, recombinant Pyst2-L binds the phospho-JNK protein. These findings may raise new perspectives regarding the role played by this phosphatase in malignant cells and in activation processes.
Collapse
Affiliation(s)
- Levy-Nissenbaum Orlev
- The Department of Neurobiochemistry, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, 69978 Tel Aviv, Israel
| | | | | | | | | | | |
Collapse
|
17
|
Kelicen P, Tindberg N. Lipopolysaccharide induces CYP2E1 in astrocytes through MAP kinase kinase-3 and C/EBPbeta and -delta. J Biol Chem 2003; 279:15734-42. [PMID: 14670949 DOI: 10.1074/jbc.m311850200] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytochrome P450 2E1 (CYP2E1) is highly inducible in a subset of astrocytes in vivo following ischemic or mechanical injury and in vitro by lipopolysaccharide (LPS) or interleukin-1beta. We have studied the mechanism of induction, and found that transcriptional activation of CYP2E1 occurred within 3 h, and CYP2E1 dependent catalytic activity was induced more than 4-fold within 5 h. The induction was sensitive to several tyrosine kinase inhibitors, and was further modulated by inhibitors of p38 MAP kinase. MAP kinase kinase-3 (MKK3) was phosphorylated in response to LPS, and expression of constitutively active MKK3, but not the MAP kinase kinases MEKK1 or MKK1, activated CYP2E1. Transcriptional activation was mediated through a C/EBPbeta and -delta binding element situated at -486/-474, and appeared to involve activation of prebound factors as well as recruitment of newly synthesized C/EBPbeta and -delta. It is thus suggested that LPS induces MKK3 activation in astrocytes, which in turn stimulates a C/EBPbeta and -delta binding element to mediate transcriptional activation of CYP2E1.
Collapse
Affiliation(s)
- Pelin Kelicen
- Division of Molecular Toxicology, Institute of Environmental Medicine, Karolinska Institute, S-171-77 Stockholm, Sweden
| | | |
Collapse
|
18
|
Kumar A, Knox AJ, Boriek AM. CCAAT/enhancer-binding protein and activator protein-1 transcription factors regulate the expression of interleukin-8 through the mitogen-activated protein kinase pathways in response to mechanical stretch of human airway smooth muscle cells. J Biol Chem 2003; 278:18868-76. [PMID: 12637525 DOI: 10.1074/jbc.m212777200] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Here we investigated the mechanisms by which mechanical stretch regulates the production of IL-8 in primary human airway smooth muscle cells (HASMC). Bronchial HASMC were subjected to cyclic mechanical stretch (12%, 1 Hz) using the computer-controlled Flexcell Strain system. Mechanical stretch increased IL-8 mRNA expression and protein production. Cyclic stretch of HASMC also increased the kinase activities of ERK1/2, JNK1, p38, and the DNA binding activities of AP-1 and C/EBP transcription factors with little effect on NF-kappa B. The inhibition of AP-1 and C/EBP transcriptional activities blocked the production of IL-8 in culture supernatants. Furthermore, the inhibition of ERK1/2 and p38 but not JNK1 caused a significant down-regulation in the expression and production of IL-8 in response to cyclic stretch. Although protein tyrosine kinases were required for the activation of both ERK1/2 and p38 kinase, stretch-activated channels, small GTPase proteins, and extracellular Ca2+ influx were required only for the activation of p38 kinase whereas phosphoinositide 3-kinase was needed for ERK1/2 activation. In addition, the phosphorylation of ERK1/2 was essential for the activation of AP-1 whereas p38 MAP kinase was needed for the activation of C/EBP. Our data demonstrate that the cyclic stretch of HASMC causes the increased production of IL-8 by activating the AP-1 and C/EBP transcription factors through the activation of ERK1/2 and p38 kinase signaling pathways.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Medicine, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | |
Collapse
|
19
|
Li P, Xin F, Fu XB, Yang YH, Guo BC. Effects of EGF on expression of phosphorylated p44/42 MAPK in rat small intestine after ischemia-reperfusion injury. Shijie Huaren Xiaohua Zazhi 2003; 11:578-582. [DOI: 10.11569/wcjd.v11.i5.578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the effects of EGF on the characteristics of phosphrylated p44/42 MAPK expression and its biological significance in EGF-induced gut repair after ischemia-reperfusion (I/R) injury.
METHODS A total of 80 Wistar rats were randomly divided into four groups, namely EGF treated group (E), normal saline control (R), ischemia group (I) and sham operated control (C). In group E and R, the rats were treated with intravenous EGF 100 μg/kg/rat or normal saline respectively after 45 minutes of superior mesenteric artery occlusion. Blood samples were collected at 2, 6, 12 and 24 hours after reperfusion and plasma D-lactate were determined. Tissue samples from intestine were also taken for histological analysis and immunohistochemical analysis of phospho-p44/42 MAPK.
RESULTS The changes of histological structure and D-lactate indicated that the intestinal barrier was damaged after intestinal I/R injury, while EGF treatment significantly improved the outcome. In group C and I positive signals of phospho-p44/42 MAPK were mainly located in the cytoplasm of the intestinal villi and crypts, while in group I positive cells increased significantly (P<0.05). In group R, positive signals were found in almost all the cells and the percentage of positive nuclei increased with the time of reperfusion, reaching its peak after 12h of reperfusion. In group E, the percentages were higher than those in group R and the peak of nuclear expression was earlier.
CONCLUSION EGF administration improves the outcome of I/R induced intestinal damage. After I/R the expression and nuclear translocation of phspho-p44/42 MAPK increases with the time of reperfusion, suggesting its role in intestinal reconstitution. EGF treatment induces its early expression and translocation into the nucleus, suggesting the significance of p44/42 MAPK signaling pathway in EGF-induced gut repair.
Collapse
Affiliation(s)
- Ping Li
- Department of Anesthesiology, 304 Hospital of PLA, Bei jing 100037, China
| | - Feng Xin
- Department of Anesthesiology, the Second Artillery General Hospital of PLA, Beijing 100088, China
| | - Xiao-Bing Fu
- Trauma Research Institute, 304 Hospital of PLA, Beijing, 100037, China
| | - Yin-Hui Yang
- Trauma Research Institute, 304 Hospital of PLA, Beijing, 100037, China
| | - Bao-Chen Guo
- Department of Anesthesiology, 304 Hospital of PLA, Bei jing 100037, China
| |
Collapse
|
20
|
Aigner A, Renneberg H, Bojunga J, Apel J, Nelson PS, Czubayko F. Ribozyme-targeting of a secreted FGF-binding protein (FGF-BP) inhibits proliferation of prostate cancer cells in vitro and in vivo. Oncogene 2002; 21:5733-42. [PMID: 12173043 DOI: 10.1038/sj.onc.1205560] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2001] [Revised: 03/28/2002] [Accepted: 04/04/2002] [Indexed: 12/19/2022]
Abstract
Prostate cancer is one of the most common malignant tumors with increasing incidence rates in the aging male. Since locally advanced or metastatic prostate tumors are essentially incurable, identification of new target molecules and treatment strategies is of critical importance. Fibroblast growth factor-2 (FGF-2) acts as potent mitogen which is upregulated in prostate cancers modulating cancer cell proliferation and development of an invasive phenotype. Normally it is tightly bound to the extracellular matrix that quenches its biological activity. The FGF-binding proteins (FGF-BP, HBp17) is a secreted protein which is able to mobilize and activate FGF-2 from the extracellular matrix. Here we show that FGF-BP is highly expressed in prostate tumor cells. To study the functional role of FGF-BP, we use a ribozyme-targeting approach to selectively deplete FGF-BP in prostate cancer cells achieving a more than 50% reduction of FGF-BP mRNA and protein levels in two mass-transfected cell lines. FGF-BP depletion reduces proliferation of the cells in vitro without changes in cell cycle distribution or apoptosis. Using cDNA microarrays, Northern blotting and RT-PCR, we show a complex pattern of changes in the gene expression profiles upon FGF-BP depletion. Most strikingly, ribozyme-mediated reduction of FGF-BP levels completely abolishes the ability of the highly metastatic PC-3 prostate carcinoma cells to grow tumors in an athymic nude mouse in vivo model which is far beyond the effects of FGF-BP ribozyme targeting observed previously in cells from other tumors in the same model. Taken together, our study identifies FGF-BP as a potential rate-limiting factor for prostate cancer growth and, due to its restricted expression pattern in adults, a potentially attractive target for prostate cancer therapy.
Collapse
Affiliation(s)
- Achim Aigner
- Department of Pharmacology and Toxicology, Philipps-University School of Medicine, Marburg, Germany.
| | | | | | | | | | | |
Collapse
|
21
|
Stöppler H, Malerczyk C, Block K, Aigner A, Czubayko F. The human papillomavirus (HPV) 16 E6 oncoprotein leads to an increase in gene expression of the angiogenic switch molecule FGF-BP in non-immortalized human keratinocytes. Oncogene 2001; 20:7430-6. [PMID: 11704874 DOI: 10.1038/sj.onc.1204957] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2001] [Revised: 08/27/2001] [Accepted: 09/04/2001] [Indexed: 11/09/2022]
Abstract
Fibroblast growth factor binding protein (FGF-BP) is a secreted protein that binds FGF-1 and FGF-2 and is involved in mobilization and activation of FGFs from the extracellular matrix. FGF-BP overexpression as well as ribozyme-mediated reduction of endogenous FGF-BP revealed that FGF-BP can be rate-limiting for tumor growth and angiogenesis. Recent studies showed that FGF-BP expression is up-regulated during early phases of tumorigenesis, indicating that the role of FGF-BP in angiogenesis is a critical early step in the development and progression of tumors. Human papillomavirus type 16 (HPV 16) is highly associated with the development of anogenital cancers. Here we demonstrate that the stable expression of the E6 oncogene of HPV 16 leads to an activation of the FGF-BP promoter in primary human foreskin keratinocytes (one of the natural host cells of these viruses). This is associated with an increase in the steady state levels of FGF-BP mRNA and FGF-BP protein in cells stably expressing E6. Transient E6 expression revealed that the observed activation of the FGF-BP promoter by the viral oncogene is an early process which is independent from immortalization/transformation events in the cells.
Collapse
Affiliation(s)
- H Stöppler
- Department of Pharmacology, Philipps University, Marburg, Germany
| | | | | | | | | |
Collapse
|
22
|
Cattaruzza M, Eberhardt I, Hecker M. Mechanosensitive transcription factors involved in endothelin B receptor expression. J Biol Chem 2001; 276:36999-7003. [PMID: 11457849 DOI: 10.1074/jbc.m105158200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Growing evidence suggests an involvement of the endothelin B receptor (ET(B)-R) in blood pressure-dependent arterial remodeling. To study the molecular mechanisms leading to deformation-induced ET(B)-R expression, we have cultured rat aortic smooth muscle cells on flexible elastomers and, when grown to 70% confluence, periodically stretched them for 6 h (15% elongation, 0.5 Hz). The cells responded with an increase both in ET(B)-R mRNA (12-fold compared with control) and protein (4-fold). According to nuclear run-on analysis this increase in ET(B)-R expression occurred at the level of transcription. Among various kinase pathways, Rho kinase (ROCK) and p38 mitogen-activated protein kinase (p38 MAPK) mediated part of the deformation-induced increase in ET(B)-R expression, as judged by the inhibitory effect of Y27632 (1 microm, 38% inhibition) and SB202190 (10 microm, 44% inhibition), respectively. Gel shift assaying of the three transcription factors principally activated by these kinases revealed a transient deformation-induced activation of activator protein-1 (AP-1) and CCAAT/enhancer-binding protein (C/EBP), but not activating transcription factor, that was sensitive to both Y27632 and SB212190. The potential role of AP-1 and C/EBP in deformation-induced ET(B)-R expression was verified both by using decoy oligodeoxynucleotides mimicking the DNA-binding sites of these transcription factors and a nuclear run-on-based assay employing specific antibodies directed against AP-1 and C/EBP. Both techniques unequivocally demonstrated that activation of these transcription factors, namely that of C/EBP beta, contribute to the increase of ET(B)-R gene expression in response to cyclic stretch.
Collapse
Affiliation(s)
- M Cattaruzza
- Department of Cardiovascular Physiology, University of Goettingen, Humboldtallee 23, 37073 Goettingen, Germany
| | | | | |
Collapse
|