1
|
Tang L, Chen M, Wu M, Liang H, Ge H, Ma Y, Shen Y, Lu S, Shen C, Zhang H, Zhang C, Wang Z. Fgf9 promotes incisor dental epithelial stem cell survival and enamel formation. Stem Cell Res Ther 2024; 15:293. [PMID: 39256850 PMCID: PMC11389439 DOI: 10.1186/s13287-024-03894-y] [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] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/25/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Understanding the role of cytokines in tooth development is critical for advancing dental tissue engineering. Fibroblast growth factor 9 (FGF9) is the only FGF consistently expressed throughout dental epithelial tissue, from the initiation of tooth bud formation to tooth maturation. However, mice lacking Fgf9 (Fgf9-/-) surprisingly show no obvious abnormalities in tooth development, suggesting potential compensation by other FGFs. Here we report findings from an Fgf9S99N mutation mouse model, a loss-of-function mutation with a dominant negative effect. Our study reveals that Fgf9 is crucial for dental epithelial stem cell (DESC) survival and enamel formation. METHODS To dissect the role of Fgf9 in tooth development, we performed the micro-CT, histomorphological analysis and gene expression assay in mice and embryos with S99N mutation. In addition, we assessed the effect of FGF9 on the DESC survival and dental epithelial differentiation by DESC sphere formation assay and tooth explant culture. Cell/tissue culture methods, gene expression analysis, specific inhibitors, and antibody blockage analysis were employed to explore how Fgf9 regulates enamel differentiation and DESC survival through both direct and indirect mechanisms. RESULTS The Fgf9S99N mutation in mice led to reduced ameloblasts, impaired enamel formation, and increased apoptosis in the cervical loop (CL). DESC sphere culture experiments revealed that FGF9 facilitated DESC survival via activating ERK/CREB signaling, without affecting cell proliferation. Furthermore, in vitro tissue culture experiments demonstrated that FGF9 promoted enamel formation in a manner dependent on the presence of mesenchyme. Interestingly, FGF9 stimulation inhibited enamel formation in isolated enamel epithelia and DESC spheres. Further investigation revealed that FGF9 supports DESC survival and promotes amelogenesis by stimulating the secretion of FGF3 and FGF10 in dental mesenchymal cells via the MAPK/ERK signaling pathway. CONCLUSIONS Our study demonstrates that Fgf9 is essential for DESC survival and enamel formation. Fgf9 performs as a dual-directional regulator of the dental enamel epithelium, not only inhibiting DESC differentiation into ameloblasts to preserve the stemness of DESC, but also promoting ameloblast differentiation through epithelial-mesenchymal interactions.
Collapse
Affiliation(s)
- Lingyun Tang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Mingmei Chen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Min Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to SJTUSM, Shanghai, China
| | - Hui Liang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Haoyang Ge
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Yan Ma
- Ruijin Hospital Lu Wan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China
| | - Yan Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Shunyuan Lu
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Chunling Shen
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Hongxin Zhang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China
| | - Chenping Zhang
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, Zhejiang, P.R. China.
| | - Zhugang Wang
- State Key Laboratory of Medical Genomics, Research Center for Experimental Medicine, Rui- Jin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin Road II, Building #17, Shanghai, 200025, P.R. China.
| |
Collapse
|
2
|
Peng X, Zhang X, Sharma G, Dai C. Thymol as a Potential Neuroprotective Agent: Mechanisms, Efficacy, and Future Prospects. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:6803-6814. [PMID: 38507708 DOI: 10.1021/acs.jafc.3c06461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Neurodegenerative diseases pose a growing global health challenge, with limited effective therapeutic options. Mitochondrial dysfunction, oxidative stress, neuroinflammation, apoptosis, and autophagy are common underlying mechanisms in these diseases. Thymol is a phenolic monoterpene compound that has gained attention for its diverse biological properties, including antioxidant, anti-inflammatory, and immunomodulatory activities. Thymol supplementation could provide potential neuroprotection and improve cognitive deficits, depressant-like effects, learning, and memory impairments in rodents. Mechanistic investigations reveal that the neuroprotective effects of thymol involve the improvement of oxidative stress, mitochondrial dysfunction, and inflammatory response. Several signaling pathways, including mitochondrial apoptotic, NF-κB, AKT, Nrf2, and CREB/BDNF pathways are also involved. In this review, the neuroprotective effects of thymol, the potential molecular mechanisms, safety, applications, and current challenges toward development as a neuroprotective agent were summarized and discussed. We hope that this review provides valuable insights for the further development of this promising natural product as a promising neuroprotective agent.
Collapse
Affiliation(s)
- Xinyan Peng
- College of Life Sciences, Yantai University, Yantai 264000, P. R. China
| | - Xiaowen Zhang
- College of Life Sciences, Yantai University, Yantai 264000, P. R. China
| | - Gaurav Sharma
- Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Chongshan Dai
- National Key Laboratory of Veterinary Public Health and Safety, College of Veterinary Medicine, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
3
|
Abdel-Fattah MM, Abo-El Fetoh ME, Afify H, Ramadan LAA, Mohamed WR. Probenecid ameliorates testosterone-induced benign prostatic hyperplasia: Implications of PGE-2 on ADAM-17/EGFR/ERK1/2 signaling cascade. J Biochem Mol Toxicol 2023; 37:e23450. [PMID: 37352135 DOI: 10.1002/jbt.23450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/05/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
Benign prostatic hyperplasia (BPH) is one of the most prevalent clinical disorders in the elderly. Probenecid (Prob) is a well-known FDA-approved therapy for gout owing to its uricosuric effect. The present study evaluated the use of Prob for BPH as a COX-2 inhibitor. Prob (100 and 200 mg/kg) was intraperitoneally injected into male Wistar rats daily for 3 weeks. In the second week, testosterone (3 mg/kg) was subcutaneously injected to induce BPH. Compared with BPH-induced rats, Prob treatment reduced prostate weight and index and improved histopathological architecture. The protease activity of ADAM-17/TACE and its ligands (TGF-α and TNF-α) were regulated by prob, which in turn abolished EGFR phosphorylation, and several inflammatory mediators (COX-2, PGE2, NF-κB (p65), and IL-6) were suppressed. By reducing the nuclear import of extracellular regulated kinase protein 1/2 (ERK1/2), Prob helped re-establish the usual equilibrium between antiapoptotic proteins like Bcl-2 and cyclin D1 and proapoptotic proteins like Bax. All of these data point to Prob as a promising treatment for BPH because of its ability to inhibit COX-2-syntheiszed PGE2 and control the ADAM-17/TGF-α-induced EGFR/ERK1/2 signaling cascade. These findings might help to repurpose Prob for the treatment of BPH.
Collapse
Affiliation(s)
- Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mohammed E Abo-El Fetoh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian-Russian University, Cairo, Egypt
| | - Hassan Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian-Russian University, Cairo, Egypt
| | - Laila A A Ramadan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian-Russian University, Cairo, Egypt
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
4
|
Guo T, Wu C, Zhou L, Zhang J, Wang W, Shen Y, Zhang L, Niu M, Zhang X, Yu R, Liu X. Preclinical evaluation of Mito-LND, a targeting mitochondrial metabolism inhibitor, for glioblastoma treatment. J Transl Med 2023; 21:532. [PMID: 37550679 PMCID: PMC10405494 DOI: 10.1186/s12967-023-04332-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/08/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a brain tumor with the highest level of malignancy and the worst prognosis in the central nervous system. Mitochondrial metabolism plays a vital role in the occurrence and development of cancer, which provides critical substances to support tumor anabolism. Mito-LND is a novel small-molecule inhibitor that can selectively inhibit the energy metabolism of tumor cells. However, the therapeutic effect of Mito-LND on GBM remains unclear. METHODS The present study evaluated the inhibitory effect of Mito-LND on the growth of GBM cells and elucidated its potential mechanism. RESULTS The results showed that Mito-LND could inhibit the survival, proliferation and colony formation of GBM cells. Moreover, Mito-LND induced cell cycle arrest and apoptosis. Mechanistically, Mito-LND inhibited the activity of mitochondrial respiratory chain complex I and reduced mitochondrial membrane potential, thus promoting ROS generation. Importantly, Mito-LND could inhibit the malignant proliferation of GBM by blocking the Raf/MEK/ERK signaling pathway. In vivo experiments showed that Mito-LND inhibited the growth of GBM xenografts in mice and significantly prolonged the survival time of tumor-bearing mice. CONCLUSION Taken together, the current findings support that targeting mitochondrial metabolism may be as a potential and promising strategy for GBM therapy, which will lay the theoretical foundation for further clinical trials on Mito-LND in the future.
Collapse
Affiliation(s)
- Tongxuan Guo
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Changyong Wu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lingni Zhou
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junhao Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wanzhou Wang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yang Shen
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ludong Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Mingshan Niu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Zhang
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Rutong Yu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xuejiao Liu
- Insititute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Abo-El Fetoh ME, Abdel-Fattah MM, Mohamed WR, Ramadan LAA, Afify H. Cyclooxygenase-2 activates EGFR-ERK1/2 pathway via PGE2-mediated ADAM-17 signaling in testosterone-induced benign prostatic hyperplasia. Inflammopharmacology 2023; 31:499-516. [PMID: 36586043 PMCID: PMC9958186 DOI: 10.1007/s10787-022-01123-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/25/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE AND DESIGN Prostatic inflammation is the driving force in benign prostatic hyperplasia (BPH). This work investigated the potential modulatory effect of COX-2 inhibition on ADAM-17/EGFR/ERK1/2 axis. MATERIALS OR SUBJECTS Adult male Wistar rats were used. TREATMENT Celecoxib (10 and 20 mg/kg; i.p.) was injected i.p. daily for three weeks. Testosterone (TST) (3 mg/kg; s.c.) was used to induce BPH. METHODS Prostatic inflammation and hyperplasia were assessed by organ weight and histopathology. Inflammatory mediators were measured using ELISA technique. Protein analysis was performed using western blotting and immunohistochemistry. Gene expression analysis was performed using qRT-PCR. Statistical analyses included one-way ANOVA and Tukey's multiple comparison test. RESULTS Testosterone-treated rats had a marked increase in COX-2, prostate weight, and index. Moreover, TST-induced COX-2 was inferred from cytoskeletal changes and was attributable to the overexpression of PGE2, NF-κB (p65), and IL-6. COX-2-derived PGE2 increased the activity of ADAM-17, TGF-α, and TNF-α. Consequently, EGFR-ERK1/2 pathway was over-activated, disrupting anti-apoptotic Bcl-2, cyclin D1, and pro-apoptotic Bax. Celecoxib reversed these effects. CONCLUSION COX-2 stimulates the ERK1/2 pathway via PGE2-ADAM-17-catalyzed shedding of TGF-α in testosterone-induced BPH. The results indicate a functional correlation between inflammation and hyperplasia in BPH.
Collapse
Affiliation(s)
- Mohammed E. Abo-El Fetoh
- grid.442695.80000 0004 6073 9704Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Maha M. Abdel-Fattah
- grid.411662.60000 0004 0412 4932Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514 Egypt
| | - Wafaa R. Mohamed
- grid.411662.60000 0004 0412 4932Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514 Egypt
| | - Laila A. A. Ramadan
- grid.442695.80000 0004 6073 9704Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Hassan Afify
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt.
| |
Collapse
|
6
|
Reich N, Hölscher C. The neuroprotective effects of glucagon-like peptide 1 in Alzheimer's and Parkinson's disease: An in-depth review. Front Neurosci 2022; 16:970925. [PMID: 36117625 PMCID: PMC9475012 DOI: 10.3389/fnins.2022.970925] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 12/16/2022] Open
Abstract
Currently, there is no disease-modifying treatment available for Alzheimer's and Parkinson's disease (AD and PD) and that includes the highly controversial approval of the Aβ-targeting antibody aducanumab for the treatment of AD. Hence, there is still an unmet need for a neuroprotective drug treatment in both AD and PD. Type 2 diabetes is a risk factor for both AD and PD. Glucagon-like peptide 1 (GLP-1) is a peptide hormone and growth factor that has shown neuroprotective effects in preclinical studies, and the success of GLP-1 mimetics in phase II clinical trials in AD and PD has raised new hope. GLP-1 mimetics are currently on the market as treatments for type 2 diabetes. GLP-1 analogs are safe, well tolerated, resistant to desensitization and well characterized in the clinic. Herein, we review the existing evidence and illustrate the neuroprotective pathways that are induced following GLP-1R activation in neurons, microglia and astrocytes. The latter include synaptic protection, improvements in cognition, learning and motor function, amyloid pathology-ameliorating properties (Aβ, Tau, and α-synuclein), the suppression of Ca2+ deregulation and ER stress, potent anti-inflammatory effects, the blockage of oxidative stress, mitochondrial dysfunction and apoptosis pathways, enhancements in the neuronal insulin sensitivity and energy metabolism, functional improvements in autophagy and mitophagy, elevated BDNF and glial cell line-derived neurotrophic factor (GDNF) synthesis as well as neurogenesis. The many beneficial features of GLP-1R and GLP-1/GIPR dual agonists encourage the development of novel drug treatments for AD and PD.
Collapse
Affiliation(s)
- Niklas Reich
- Biomedical and Life Sciences Division, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Christian Hölscher
- Neurology Department, Second Associated Hospital, Shanxi Medical University, Taiyuan, China
- Henan University of Chinese Medicine, Academy of Chinese Medical Science, Zhengzhou, China
| |
Collapse
|
7
|
Ogaly HA, Abdel-Rahman RF, Mohamed MAE, O A AF, Khattab MS, Abd-Elsalam RM. Thymol ameliorated neurotoxicity and cognitive deterioration in a thioacetamide-induced hepatic encephalopathy rat model; involvement of the BDNF/CREB signaling pathway. Food Funct 2022; 13:6180-6194. [PMID: 35583008 DOI: 10.1039/d1fo04292k] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the present study, we aimed to delineate the neuroprotective potential of thymol (THY) against neurotoxicity and cognitive deterioration induced by thioacetamide (TAA) in an experimental model of hepatic encephalopathy (HE). Rats received TAA (100 mg kg-1, intraperitoneally injected, three times per week) for two weeks. THY (30 and 60 mg kg-1), and Vit E (100 mg k-1) were administered daily by oral gavage for 30 days after HE induction. Supplementation with THY significantly improved liver function, reduced serum ammonia level, and ameliorated the locomotor and cognitive deficits. THY effectively modulated the alteration in oxidative stress markers, neurotransmitters, and brain ATP content. Histopathology of liver and brain tissues showed that THY had ameliorated TAA-induced damage, astrocyte swelling and brain edema. Furthermore, THY downregulated NF-kB and upregulated GFAP protein expression. In addition, THY significantly promoted CREB and BDNF expression at both mRNA and protein levels, together with enhancing brain cAMP level. In conclusion, THY exerted hepato- and neuroprotective effects against HE by mitigating hepatotoxicity, hyperammonemia and brain ATP depletion via its antioxidant, anti-inflammatory effects in addition to activation of the CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Hanan A Ogaly
- Department of Chemistry, College of Science, King Khalid University, Abha, Saudi Arabia. .,Department of Biochemistry, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Rehab F Abdel-Rahman
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Dokki, Giza, Egypt
| | - Marawan Abd Elbaset Mohamed
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Dokki, Giza, Egypt
| | - Ahmed-Farid O A
- Department of Physiology, National Organization for Drug Control and Research, Giza, Egypt
| | - Marwa S Khattab
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Reham M Abd-Elsalam
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
8
|
The Prostate-Associated Gene 4 (PAGE4) Could Play a Role in the Development of Benign Prostatic Hyperplasia under Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7041739. [PMID: 35633887 PMCID: PMC9135540 DOI: 10.1155/2022/7041739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/19/2022] [Accepted: 05/05/2022] [Indexed: 11/18/2022]
Abstract
Benign prostatic hyperplasia (BPH) is a common disease in elderly men with uncertain molecular mechanism, and oxidative stress (OS) has also been found associated with BPH development. Recently, we found that prostate-associated gene 4 (PAGE4) was one of the most significantly changed differentially expressed genes (DEGs) in BPH, which can protect cells against stress stimulation. However, the exact role of PAGE4 in BPH remains unclear. This study is aimed at exploring the effect of PAGE4 in BPH under OS. Human prostate tissues and cultured WPMY-1 and PrPF cells were utilized. The expression and localization of PAGE4 were determined with qRT-PCR, Western blotting, and immunofluorescence staining. OS cell models induced with H2O2 were treated with PAGE4 silencing or PAGE4 overexpression or inhibitor (N-acetyl-L-cysteine (NAC)) of OS. The proliferation activity, apoptosis, OS markers, and MAPK signaling pathways were detected by CCK-8 assay, flow cytometry analysis, and Western blotting. PAGE4 was shown to be upregulated in human hyperplastic prostate and mainly located in the stroma. Acute OS induced with H2O2 increased PAGE4 expression (which was prevented by OS inhibitor), apoptosis, cell cycle arrest, and reactive oxygen species (ROS) accumulation in WPMY-1 and PrPF cells. siPAGE4 plus H2O2 potentiated H2O2 effect via reducing the p-ERK1/2 level and increasing p-JNK1/2 level. Consistently, overexpression of PAGE4 offset the effect of H2O2 and partially reversed the PAGE4 silencing effect. However, knocking down and overexpression of PAGE4 alone determined no significant effects. Our novel data demonstrated that augmented PAGE4 promotes cell survival by activating p-ERK1/2 and decreases cell apoptosis by inhibiting p-JNK1/2 under the OS, which could contribute to the development of BPH.
Collapse
|
9
|
Sharma VK, Yang X, Kim SK, Mafi A, Saiz-Sanchez D, Villanueva-Anguita P, Xiao L, Inoue A, Goddard WA, Loh YP. Novel interaction between neurotrophic factor-α1/carboxypeptidase E and serotonin receptor, 5-HTR1E, protects human neurons against oxidative/neuroexcitotoxic stress via β-arrestin/ERK signaling. Cell Mol Life Sci 2021; 79:24. [PMID: 34966948 PMCID: PMC8732845 DOI: 10.1007/s00018-021-04021-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/14/2021] [Accepted: 11/01/2021] [Indexed: 12/18/2022]
Abstract
Protecting neurons from death during oxidative and neuroexcitotoxic stress is key for preventing cognitive dysfunction. We uncovered a novel neuroprotective mechanism involving interaction between neurotrophic factor-α1 (NF-α1/carboxypeptidase E, CPE) and human 5-HTR1E, a G protein-coupled serotonin receptor with no previously known neurological function. Co-immunoprecipitation and pull-down assays confirmed interaction between NFα1/CPE and 5-HTR1E and 125I NF-α1/CPE-binding studies demonstrated saturable, high-affinity binding to 5-HTR1E in stably transfected HEK293 cells (Kd = 13.82 nM). Treatment of 5-HTR1E stable cells with NF-α1/CPE increased pERK 1/2 and pCREB levels which prevented a decrease in pro-survival protein, BCL2, during H2O2-induced oxidative stress. Cell survival assay in β-arrestin Knockout HEK293 cells showed that the NF-α1/CPE-5-HTR1E-mediated protection against oxidative stress was β-arrestin-dependent. Molecular dynamics studies revealed that NF-α1/CPE interacts with 5-HTR1E via 3 salt bridges, stabilized by several hydrogen bonds, independent of the serotonin pocket. Furthermore, after phosphorylating the C-terminal tail and intracellular loop 3 (ICL3) of NF-α1/CPE-5-HTR1E, it recruited β-arrestin1 by forming numerous salt bridges and hydrogen bonds to ICL2 and ICL3, leading to activation of β-arrestin1. Immunofluorescence studies showed 5-HTR1E and NF-α1/CPE are highly expressed and co-localized on cell surface of human hippocampal neurons. Importantly, knock-down of 5-HTR1E in human primary neurons diminished the NF-α1/CPE-mediated protection of these neurons against oxidative stress and glutamate neurotoxicity-induced cell death. Thus, NF-α1/CPE uniquely interacts with serotonin receptor 5-HTR1E to activate the β-arrestin/ERK/CREB/BCL2 pathway to mediate stress-induced neuroprotection.
Collapse
Affiliation(s)
- Vinay Kumar Sharma
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49, Convent Drive, Bldg 49, Rm 6A-10, Bethesda, MD, 20892, USA
| | - Xuyu Yang
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49, Convent Drive, Bldg 49, Rm 6A-10, Bethesda, MD, 20892, USA
| | - Soo-Kyung Kim
- Materials and Process Simulation Center, California Institute of Technology, Pasedena, CA, 91125, USA
| | - Amirhossein Mafi
- Materials and Process Simulation Center, California Institute of Technology, Pasedena, CA, 91125, USA
| | - Daniel Saiz-Sanchez
- Neuroplasticity and Neurodegeneration Laboratory, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Patricia Villanueva-Anguita
- Neuroplasticity and Neurodegeneration Laboratory, Medical School, Regional Center for Biomedical Research, University of Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Lan Xiao
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49, Convent Drive, Bldg 49, Rm 6A-10, Bethesda, MD, 20892, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - William A Goddard
- Materials and Process Simulation Center, California Institute of Technology, Pasedena, CA, 91125, USA.
| | - Y Peng Loh
- Section on Cellular Neurobiology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 49, Convent Drive, Bldg 49, Rm 6A-10, Bethesda, MD, 20892, USA.
| |
Collapse
|
10
|
Madecassic acid protects human periodontal ligament fibroblasts against hydrogen peroxide-induced cell damage by maintaining mitochondrial membrane potential. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00174-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
11
|
Farhat D, Ghayad SE, Icard P, Le Romancer M, Hussein N, Lincet H. Lipoic acid-induced oxidative stress abrogates IGF-1R maturation by inhibiting the CREB/furin axis in breast cancer cell lines. Oncogene 2020; 39:3604-3610. [PMID: 32060422 DOI: 10.1038/s41388-020-1211-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 12/21/2022]
Abstract
The beneficial effects of lipoic acid (LA) in cancer treatment have been well documented in the last decade. Indeed, LA exerts crucial antiproliferative effects by reducing breast cancer cell viability, cell cycle progression and the epithelial-to-mesenchymal transition (EMT). However, the mechanisms of action (MOA) underlying these antiproliferative effects remain to be elucidated. Recently, we demonstrated that LA decreases breast cancer cell proliferation by inhibiting IGF-1R maturation via the downregulation of the proprotein convertase furin. The aim of the present study was to investigate the MOA by which LA inhibits furin expression in estrogen receptor α (ERα) (+) and (-) breast cancer cell lines. We unveil that LA exerts a pro-oxidant effect on these cell lines, the resulting reactive oxygen species (ROS) generated being responsible for the reduction in the expression of the major (CREB) protein. This transcription factor is overexpressed in many types of cancers and regulates the expression of furin in breast cancer cells independently of ERα, as evidenced herein by the inhibition of furin expression following CREB silencing. Consequently, our findings expose for the first time the complete MOA of LA via the CREB/furin axis leading to inhibition of breast cancer cell proliferation.
Collapse
Affiliation(s)
- Diana Farhat
- Université Lyon 1, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France.,Lebanese University, Faculty of Sciences, Cancer biology Stem Cells and Molecular Immunology, Hadath-Beirut, Lebanon
| | - Sandra E Ghayad
- Department of Biology, Faculty of Science II, Lebanese University, Fanar, Lebanon
| | - Philippe Icard
- Normandie Univ, UNICAEN, CHU de Caen Normandie, Unité de recherche BioTICLA INSERM U 119, 14000, Caen, France.,Service de chirurgie thoracique, Hôpital Cochin, Hôpitaux Universitaires Paris Centre, Paris, France
| | - Muriel Le Romancer
- Université Lyon 1, Lyon, France.,Inserm U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France.,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France
| | - Nader Hussein
- Lebanese University, Faculty of Sciences, Cancer biology Stem Cells and Molecular Immunology, Hadath-Beirut, Lebanon
| | - Hubert Lincet
- Université Lyon 1, Lyon, France. .,Inserm U1052, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France. .,CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon (CRCL), Lyon, France. .,ISPB, Faculté de Pharmacie, Lyon, France.
| |
Collapse
|