1
|
Gu J, Zhu LK, Zhao X, Jiang J, Jiang R. Low testosterone state inhibits erectile function by downregulating the expression of GIT1 in rat penile corpus cavernosum. Sex Med 2023; 11:qfad017. [PMID: 37256221 PMCID: PMC10226814 DOI: 10.1093/sexmed/qfad017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 06/01/2023] Open
Abstract
Background The mechanism of erectile dysfunction (ED) caused by a low androgen level is still not clear. Aim To explore the influence of the low testosterone state on G protein-coupled receptor kinase interactor 1 (GIT1) and its contact to erectile function. Methods Thirty male Sprague-Dawley rats aged 8 weeks were distributed at random into 5 groups: control (sham operated), castration, testosterone supplement after castration, castration + vacant lentiviral transfection, and castration + lentiviral transfection. The testis and epididymis were removed through a scrotal incision to develop castrated rats. Four weeks after castration, a lentivirus carrying the GIT1 gene was injected into the middle of rat penile corpus cavernosum. One week after transfection, maximum intracavernous pressure/mean arterial pressure (ICPmax/MAP), serum testosterone, nitric oxide, GIT1, endothelial nitric oxide synthase (eNOS), phospho-eNOS (p-eNOS), p-eNOS/eNOS, and the interaction between eNOS and GIT1 were assessed in the rats. Outcomes The levels of GIT1 in the penile cavernous tissue of castrated rats are significantly lower than that of controls. Results GIT1 was expressed in the cytoplasm and cell membrane of vascular endothelial cells and smooth muscle cells in rat penile tissue. In comparison with normal rats, the castrated rats showed lower levels of GIT1 expression, GIT1 and eNOS interaction, p-eNOS/eNOS, nitric oxide, and ICPmax/MAP (P < .01). Overexpression of GIT1 can intensively enhance the expression level of GIT1, the interaction between GIT1 and eNOS, p-eNOS/eNOS, nitric oxide, and ICPmax/MAP in rats (P < .01). Clinical Translation Modulating the interaction between eNOS and GIT1 might be a novel method of treating ED caused by a low androgen level. Strengths and Limitations The impact of GIT1 phosphorylation on the activity of eNOS and its possible mechanisms affecting erectile function require further study. Conclusion A low testosterone state inhibits erectile function in rats by reducing the expression of GIT1 and the protein interaction between GIT1 and eNOS.
Collapse
Affiliation(s)
| | | | - Xin Zhao
- Department of Urology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jun Jiang
- Department of Thyroid Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Rui Jiang
- Corresponding author: Department of Urology, Nephropathy Clinical Medical Research Center of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Taiping Road, Luzhou, Sichuan 646000, China. ;
| |
Collapse
|
2
|
Xu T, Gao P, Huang Y, Wu M, Yi J, Zhou Z, Zhao X, Jiang T, Liu H, Qin T, Yang Z, Wang X, Bao T, Chen J, Zhao S, Yin G. Git1-PGK1 interaction achieves self-protection against spinal cord ischemia-reperfusion injury by modulating Keap1/Nrf2 signaling. Redox Biol 2023; 62:102682. [PMID: 36963288 PMCID: PMC10053403 DOI: 10.1016/j.redox.2023.102682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 03/13/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Spinal cord ischemia-reperfusion (IR) injury (SCIRI) is a significant secondary injury that causes damage to spinal cord neurons, leading to the impairment of spinal cord sensory and motor functions. Excessive reactive oxygen species (ROS) production is considered one critical mechanism of neuron damage in SCIRI. Nonetheless, the molecular mechanisms underlying the resistance of neurons to ROS remain elusive. Our study revealed that the deletion of Git1 in mice led to poor recovery of spinal cord motor function after SCIRI. Furthermore, we discovered that Git1 has a beneficial effect on neuron resistance to ROS production. Mechanistically, Git1 interacted with PGK1, regulated PGK1 phosphorylation at S203, and affected the intermediate products of glycolysis in neurons. The influence of Git1 on glycolysis regulates the dimerization of Keap1, which leads to changes in Nrf2 ubiquitination and plays a role in resisting ROS. Collectively, we show that Git1 regulates the Keap1/Nrf2 axis to resist ROS in a PGK1-dependent manner and thus is a potential therapeutic target for SCIRI.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing, 210008, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Peng Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Yifan Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Mengyuan Wu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Jiang Yi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Zheng Zhou
- Department of Emergency Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Xuan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Tao Qin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Zhenqi Yang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Xiaowei Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Tianyi Bao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China; Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
3
|
Wang G, Bai X, Jiang G, Jin S, Wang Q, Wang A, Peng R, Ke A, Bai D. GIT1 overexpression promotes epithelial-mesenchymal transition and predicts poor prognosis in hepatocellular carcinoma. Bioengineered 2021; 12:30-43. [PMID: 33258389 PMCID: PMC8806235 DOI: 10.1080/21655979.2020.1855914] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022] Open
Abstract
Globally, hepatocellular carcinoma (HCC) is one of the most common causes of cancer-associated mortalities. It has a high rate of metastasis and recurrence, which predict a poor prognosis. G-protein-coupled receptor (GPCR)-kinase interacting protein-1 (GIT1) is a multifunctional scaffold protein that mediates the progression of various tumors. Studies have correlated GIT1 with HCC, however, these correlations have not been fully elucidated. Therefore, we aimed at evaluating the expression of GIT1 in HCC tissues and cells, and to investigate its role and potential mechanisms in HCC progression. The expression levels of GIT1 in HCC tissues and other cancers was determined by using the Oncomine and TCGA databases. Functional analysis of GIT1 in HCC was evaluated through in vitro and in vivo experiments, whereby, HCC cells were transfected with synthetically overexpressed and short hairpin RNA (shRNA) lentivirus-mediated plasmids. Kaplan-Meier and Cox regression methods were used to establish the associations between GIT1 and clinical outcomes of 158 HCC patients. GIT1 was found to be elevated in HCC tissues where it promoted the invasion, migration, and proliferation of HCC cells. Moreover, the overexpression of GIT1 prompted epithelial-mesenchymal transition (EMT) by activating extracellular regulated kinase 1/2 (ERK1/2) pathway, which was shown to be reversed by SCH772984, a specific ERK1/2 inhibitor. GIT1 was also found to be associated with malignant features of HCC, leading to a poorer prognosis. In conclusion, GIT1 promotes HCC progression by inducing EMT and may reflect the course of HCC patients.
Collapse
Affiliation(s)
- Guifu Wang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
- The First Clinical Medical College, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Xuesong Bai
- The First Clinical Medical College, Dalian Medical University, Dalian, Liaoning, P.R. China
| | - Guoqing Jiang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Shengjie Jin
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Qian Wang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Aoqing Wang
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Rui Peng
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| | - Aiwu Ke
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Dousheng Bai
- Department of Hepatobiliary Surgery, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu, P.R. China
| |
Collapse
|
4
|
Luo Y, Xu T, Liu W, Rong Y, Wang J, Fan J, Yin G, Cai W. Exosomes derived from GIT1-overexpressing bone marrow mesenchymal stem cells promote traumatic spinal cord injury recovery in a rat model. Int J Neurosci 2021; 131:170-182. [PMID: 32223487 DOI: 10.1080/00207454.2020.1734598] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/27/2019] [Accepted: 01/24/2020] [Indexed: 01/07/2023]
Abstract
OBJECTIVE This study aims to explore the effects of exosomes derived from G protein-coupled receptor kinase 2 interacting protein 1 (GIT1)-overexpressing bone marrow mesenchymal stem cell (GIT1-BMSC-Exos) on the treatment of traumatic spinal cord injury (SCI) in a rat model. METHODS All the rats underwent a T10 laminectomy. A weight-drop impact was performed using a 10-g rod from a height of 12.5 mm except the sham group. Rats with SCI were distributed into three groups randomly and then treated with tail vein injection of GIT1-BMSCs-Exos, BMSCs-Exos and PBS, respectively. The effects of GIT1-Exos on glutamate (GLU)-induced apoptosis in vitro were also evaluated by TUNEL staining. RESULTS The results showed that rats treated with GIT1-BMSCs-Exos had better functional behavioral recovery than those treated with PBS or BMSCs-Exos only. The overexpression of GIT1 in BMSCs-Exos not only restrained glial scar formation and neuroinflammation after SCI, but also attenuated apoptosis and promoted axonal regeneration in the injured lesion area. Neuronal cell death induced by GLU was controlled remarkably in vitro as well. CONCLUSION In conclusion, our study suggested that the application of GIT1-BMSCs-Exos may provide a novel avenue for traumatic SCI treatment.
Collapse
Affiliation(s)
- Yongjun Luo
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Xu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Liu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yuluo Rong
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiaxing Wang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin Fan
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Weihua Cai
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Hendrickx JO, van Gastel J, Leysen H, Santos-Otte P, Premont RT, Martin B, Maudsley S. GRK5 - A Functional Bridge Between Cardiovascular and Neurodegenerative Disorders. Front Pharmacol 2018; 9:1484. [PMID: 30618771 PMCID: PMC6304357 DOI: 10.3389/fphar.2018.01484] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022] Open
Abstract
Complex aging-triggered disorders are multifactorial programs that comprise a myriad of alterations in interconnected protein networks over a broad range of tissues. It is evident that rather than being randomly organized events, pathophysiologies that possess a strong aging component such as cardiovascular diseases (hypertensions, atherosclerosis, and vascular stiffening) and neurodegenerative conditions (dementia, Alzheimer's disease, mild cognitive impairment, Parkinson's disease), in essence represent a subtly modified version of the intricate molecular programs already in place for normal aging. To control such multidimensional activities there are layers of trophic protein control across these networks mediated by so-called "keystone" proteins. We propose that these "keystones" coordinate and interconnect multiple signaling pathways to control whole somatic activities such as aging-related disease etiology. Given its ability to control multiple receptor sensitivities and its broad protein-protein interactomic nature, we propose that G protein coupled receptor kinase 5 (GRK5) represents one of these key network controllers. Considerable data has emerged, suggesting that GRK5 acts as a bridging factor, allowing signaling regulation in pathophysiological settings to control the connectivity between both the cardiovascular and neurophysiological complications of aging.
Collapse
Affiliation(s)
- Jhana O. Hendrickx
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Jaana van Gastel
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Hanne Leysen
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt-Universitat zu Berlin, Berlin, Germany
| | - Richard T. Premont
- Harrington Discovery Institute, Case Western Reserve University, Cleveland, GA, United States
| | - Bronwen Martin
- Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Department of Biomedical Science, University of Antwerp, Antwerp, Belgium
- Center for Molecular Neurology, University of Antwerp – Flanders Institute for Biotechnology (VIB), Antwerp, Belgium
| |
Collapse
|
6
|
Qian B, Zhao L, Wang X, Xu J, Teng F, Gao L, Shen R. RETRACTED: miR-149 regulates the proliferation and apoptosis of cervical cancer cells by targeting GIT1. Biomed Pharmacother 2018; 105:1106-1116. [DOI: 10.1016/j.biopha.2018.06.075] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 06/07/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
|
7
|
Chen J, Wang Q, Zhou W, Zhou Z, Tang PY, Xu T, Liu W, Li LW, Cheng L, Zhou ZM, Fan J, Yin GY. GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling. FASEB J 2018; 32:fj201800548. [PMID: 29912587 DOI: 10.1096/fj.201800548] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
GPCR kinase 2-interacting protein-1 (GIT1) is a scaffold protein that plays an important role in cell adaptation, proliferation, migration, and differentiation; however, the role of GIT1 in the regulation of neuronal death after spinal cord injury remains obscure. Here, we demonstrate that GIT1 deficiency remarkably increased neuronal apoptosis and enhanced JNK/p38 signaling, which resulted in stronger motor deficits by ischemia-reperfusion in vivo, consistent with the finding of oxygen-glucose deprivation/reoxygenation-induced neuronal injury in vitro. After treatment with JNK and p38 inhibitors, abnormally necroptotic cell death caused by GIT1 knockdown could be partially rescued, with the recovery of neuronal viability, which was still poorer than that in control neurons. Meanwhile, overactivation of JNK/p38 after GIT1 depletion was concomitant with excessive activity of apoptosis signal-regulating kinase-1 (ASK1) that could be abolished by ASK1 silencing in HEK293T cells. Finally, GIT1 could disrupt the oligomerization of ASK1 via interaction between the synaptic localization domain that contains the coiled-coil (CC)-2 domain of GIT1 and the C-terminal CC domain of ASK1. It suppressed the autophosphorylation of ASK1 and led to decreasing activity of the ASK1/JNK/p38 pathway. These data reveal a protective role for GIT1 in neuronal damage by modulating ASK1/JNK/p38 signaling.-Chen, J., Wang, Q., Zhou, W., Zhou, Z., Tang, P.-Y., Xu, T., Liu, W., Li, L.-W., Cheng, L., Zhou, Z.-M., Fan, J., Yin, G.-Y. GPCR kinase 2-interacting protein-1 protects against ischemia-reperfusion injury of the spinal cord by modulating ASK1/JNK/p38 signaling.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zheng Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Peng-Yu Tang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin-Wei Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lin Cheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Min Zhou
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guo-Yong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Edogawa S, Peters SA, Jenkins GD, Gurunathan SV, Sundt WJ, Johnson S, Lennon RJ, Dyer RB, Camilleri M, Kashyap PC, Farrugia G, Chen J, Singh RJ, Grover M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota. FASEB J 2018; 32:fj201800560R. [PMID: 29897814 PMCID: PMC6219825 DOI: 10.1096/fj.201800560r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022]
Abstract
Intestinal barrier function and microbiota are integrally related and play critical roles in maintenance of host physiology. Sex is a key biologic variable for several disorders. Our aim was to determine sex-based differences in response to perturbation and subsequent recovery of intestinal barrier function and microbiota in healthy humans. Twenty-three volunteers underwent duodenal biopsies, mucosal impedance, and in vivo permeability measurement. Permeability testing was repeated after administration of indomethacin, then 4 to 6 wk after its discontinuation. Duodenal and fecal microbiota composition was determined using 16S rRNA amplicon sequencing. Healthy women had lower intestinal permeability and higher duodenal and fecal microbial diversity than healthy men. Intestinal permeability increases after indomethacin administration in both sexes. However, only women demonstrated decreased fecal microbial diversity, including an increase in Prevotella abundance, after indomethacin administration. Duodenal microbiota composition did not show sex-specific changes. The increase in permeability and microbiota changes normalized after discontinuation of indomethacin. In summary, women have lower intestinal permeability and higher microbial diversity. Intestinal permeability is sensitive to perturbation but recovers to baseline. Gut microbiota in women is sensitive to perturbation but appears to be more stable in men. Sex-based differences in intestinal barrier function and microbiome should be considered in future studies.-Edogawa, S., Peters, S. A., Jenkins, G. D., Gurunathan, S. V., Sundt, W. J., Johnson, S., Lennon, R. J., Dyer, R. B., Camilleri, M., Kashyap, P. C., Farrugia, G., Chen, J., Singh, R. J., Grover, M. Sex differences in NSAID-induced perturbation of human intestinal barrier function and microbiota.
Collapse
Affiliation(s)
- Shoko Edogawa
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephanie A. Peters
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gregory D. Jenkins
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Wendy J. Sundt
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Stephen Johnson
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan J. Lennon
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Roy B. Dyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael Camilleri
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Purna C. Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jun Chen
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ravinder J. Singh
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Madhusudan Grover
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
9
|
Majumder S, Zhu G, Xu X, Senchanthisai S, Jiang D, Liu H, Xue C, Wang X, Coia H, Cui Z, Smolock EM, Libby RT, Berk BC, Pang J. G-Protein-Coupled Receptor-2-Interacting Protein-1 Controls Stalk Cell Fate by Inhibiting Delta-like 4-Notch1 Signaling. Cell Rep 2017; 17:2532-2541. [PMID: 27926858 DOI: 10.1016/j.celrep.2016.11.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 04/12/2016] [Accepted: 11/01/2016] [Indexed: 10/20/2022] Open
Abstract
The spatiotemporal localization and expression of Dll4 are critical for sprouting angiogenesis. However, the related mechanisms are poorly understood. Here, we show that G-protein-coupled receptor-kinase interacting protein-1 (GIT1) is a robust endogenous inhibitor of Dll4-Notch1 signaling that specifically controls stalk cell fate. GIT1 is highly expressed in stalk cells but not in tip cells. GIT1 deficiency remarkably enhances Dll4 expression and Notch1 signaling, resulting in impaired retinal sprouting angiogenesis, which can be rescued by treatment with the Notch inhibitor or Dll4 neutralizing antibody. Notch1 regulates Dll4 expression by binding to recombining binding protein suppressor of hairless (RBP-J, a transcriptional regulator of Notch) via a highly conserved ankyrin (ANK) repeat domain. We show that GIT1, which also contains an ANK domain, inhibits the Notch1-Dll4 signaling pathway by competing with Notch1 ANK domain for binding to RBP-J in stalk cells.
Collapse
Affiliation(s)
- Syamantak Majumder
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - GuoFu Zhu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiangbin Xu
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Sharon Senchanthisai
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Dongyang Jiang
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Hao Liu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chao Xue
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Xiaoqun Wang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Heidi Coia
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Zhaoqiang Cui
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Elaine M Smolock
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Richard T Libby
- Flaum Eye Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Jinjiang Pang
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA; Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China.
| |
Collapse
|
10
|
Jiang D, Zhuang J, Peng W, Lu Y, Liu H, Zhao Q, Chi C, Li X, Zhu G, Xu X, Yan C, Xu Y, Ge J, Pang J. Phospholipase Cγ1 Mediates Intima Formation Through Akt-Notch1 Signaling Independent of the Phospholipase Activity. J Am Heart Assoc 2017; 6:JAHA.117.005537. [PMID: 28698260 PMCID: PMC5586285 DOI: 10.1161/jaha.117.005537] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background Vascular smooth muscle cell proliferation, migration, and dedifferentiation are critical for vascular diseases. Recently, it was demonstrated that Notch receptors have opposing effects on intima formation after vessel injury. Therefore, it is important to investigate the specific regulatory pathways that activate the different Notch receptors. Methods and Results There was a time‐ and dose‐dependent activation of Notch1 by angiotensin II and platelet‐derived growth factor in vascular smooth muscle cells. When phospholipase Cγ1 (PLCγ1) expression was reduced by small interfering RNA, Notch1 activation and Hey2 expression (Notch target gene) induced by angiotensin II or platelet‐derived growth factor were remarkably inhibited, while Notch2 degradation was not affected. Mechanistically, we observed an association of PLCγ1 and Akt, which increased after angiotensin II or platelet‐derived growth factor stimulation. PLCγ1 knockdown significantly inhibited Akt activation. Importantly, PLCγ1 phospholipase site mutation (no phospholipase activity) did not affect Akt activation. Furthermore, PLCγ1 depletion inhibited platelet‐derived growth factor–induced vascular smooth muscle cell proliferation, migration, and dedifferentiation, while it increased apoptosis. In vivo, PLCγ1 and control small interfering RNA were delivered periadventitially in pluronic gel and complete carotid artery ligation was performed. Morphometric analysis 21 days after ligation demonstrated that PLCγ1 small interfering RNA robustly attenuated intima area and intima/media ratio compared with the control group. Conclusions PLCγ1‐Akt–mediated Notch1 signaling is crucial for intima formation. This effect is attributable to PLCγ1‐Akt interaction but not PLCγ1 phospholipase activity. Specific inhibition of the PLCγ1 and Akt interaction will be a promising therapeutic strategy for preventing vascular remodeling.
Collapse
Affiliation(s)
- Dongyang Jiang
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianhui Zhuang
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenhui Peng
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuyan Lu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Liu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Zhao
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Chi
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiankai Li
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guofu Zhu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiangbin Xu
- Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Chen Yan
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Yawei Xu
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Junbo Ge
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinjiang Pang
- Department of Cardiology, Pan-Vascular Research Institute, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China .,Aab Cardiovascular Research Institute and Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
11
|
Tao WY, Wang CY, Sun YH, Su YH, Pang D, Zhang GQ. MicroRNA-34c Suppresses Breast Cancer Migration and Invasion by Targeting GIT1. J Cancer 2016; 7:1653-1662. [PMID: 27698902 PMCID: PMC5039386 DOI: 10.7150/jca.14762] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 06/29/2016] [Indexed: 01/03/2023] Open
Abstract
Abnormal expression of microRNAs plays important role in tumor metastasis. Migration and invasion of cancer cells accord for the metastasis and deterioration of breast cancer. However, the regulatory role of microRNAs in the invasion and migration of breast cancer cells has not completely understood yet. Here we found that microRNA-34c (miR-34c) was significantly downregulated in metastatic tissue of breast cancer. In vitro study showed that miR-34c negatively regulated GIT1 protein expression by binding to the 3'UTR of GIT1 mRNA. Consistently, GIT1 protein expression was found upregulated significantly in metastatic breast cancer. Moreover, miR-34c overexpression suppressed the expression of GIT1 protein, and this effect was restored by AMO-miR-34c in breast cancer cells. Overexpression of miR-34c suppressed cell migration and invasion in both MCF-7 and MDA-MD-231 breast cancer cells. Furthermore, knockdown of endogenous GIT1 expression reduced the migration and invasion of both two breast cancer cells. Collectively, miR-34c downregulation in breast cancer cells resulted in the upregulation of GIT1, which in turn enhanced the migration and invasion of breast cancer. This study highlights molecular mechanism of migration and invasion of breast cancer cells.
Collapse
Affiliation(s)
- Wei-Yang Tao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China;; Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China
| | - Chun-Yang Wang
- Key Laboratory of Cardiovascular Medicine Research (Harbin Medical University), Ministry of Education, Harbin, China;; Department of Urology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong-Hui Sun
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yong-Hui Su
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Da Pang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guo-Qiang Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
12
|
Specific dephosphorylation at tyr-554 of git1 by ptprz promotes its association with paxillin and hic-5. PLoS One 2015; 10:e0119361. [PMID: 25742295 PMCID: PMC4351203 DOI: 10.1371/journal.pone.0119361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/12/2015] [Indexed: 11/29/2022] Open
Abstract
G protein-coupled receptor kinase-interactor 1 (Git1) is involved in cell motility control by serving as an adaptor that links signaling proteins such as Pix and PAK to focal adhesion proteins. We previously demonstrated that Git1 was a multiply tyrosine-phosphorylated protein, its primary phosphorylation site was Tyr-554 in the vicinity of the focal adhesion targeting-homology (FAH) domain, and this site was selectively dephosphorylated by protein tyrosine phosphatase receptor type Z (Ptprz). In the present study, we showed that Tyr-554 phosphorylation reduced the association of Git1 with the FAH-domain-binding proteins, paxillin and Hic-5, based on immunoprecipitation experiments using the Tyr-554 mutants of Git1. The Tyr-554 phosphorylation of Git1 was higher, and its binding to paxillin was consistently lower in the brains of Ptprz-deficient mice than in those of wild-type mice. We then investigated the role of Tyr-554 phosphorylation in cell motility control using three different methods: random cell motility, wound healing, and Boyden chamber assays. The shRNA-mediated knockdown of endogenous Git1 impaired cell motility in A7r5 smooth muscle cells. The motility defect was rescued by the exogenous expression of wild-type Git1 and a Git1 mutant, which only retained Tyr-554 among the multiple potential tyrosine phosphorylation sites, but not by the Tyr-554 phosphorylation-defective or phosphorylation-state mimic Git1 mutant. Our results suggested that cyclic phosphorylation-dephosphorylation at Tyr-554 of Git1 was crucial for dynamic interactions between Git1 and paxillin/Hic-5 in order to ensure coordinated cell motility.
Collapse
|
13
|
Zhang LQ, Zhao GZ, Xu XY, Fang J, Chen JM, Li JW, Gao XJ, Hao LJ, Chen YZ. Integrin-β1 regulates chondrocyte proliferation and apoptosis through the upregulation of GIT1 expression. Int J Mol Med 2015; 35:1074-80. [PMID: 25715677 DOI: 10.3892/ijmm.2015.2114] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/05/2015] [Indexed: 01/25/2023] Open
Abstract
Chondrocytes play a critical role in the repair process of osteoarthritis, which is also known as degenerative arthritis. Integrins, as the key family of cell surface receptors, are responsible for the regulation of chondrocyte proliferation, differentiation, survival and apoptosis through the recruitment and activation of downstream adaptor proteins. Moreover, G-protein-coupled receptor kinase interacting protein-1 (GIT1) exerts its effects on cell proliferation and migration through interaction with various cytokines. It has been previously suggested that GIT1 acts as a vital protein downstream of the integrin-mediated pathway. In the present study, we investigated the effects of integrin-β1 on cell proliferation and apoptosis, as well as the underlying mechanisms in chondrocytes in vitro. Following transfection with a vector expressing integrin-β1, our results revealed that the overexpression of integrin-β1 enhanced GIT1 expression, whereas the knockdown of integrin-β1 by siRNA suppressed GIT1 expression. However, no significant effect was observed on integrin-β1 expression following the enforced overexpression of GIT1, which suggests that GIT1 is localized downstream of integrin-β1. In other words, integrin-β1 regulates the expression of GIT1. Furthermore, this study demonstrated that integrin-β1 and GIT1 increased the expression levels of aggrecan and type II collagen, thus promoting chondrocyte proliferation; however, they inhibited chondrocyte apoptosis. Taken together, our data demonstrate that integrin-β1 plays a vital role in chondrocyte proliferation, differentiation and apoptosis. GIT1 exerts effects similar to those of integrin-β1 and is a downstream target of integrin-β1.
Collapse
Affiliation(s)
- Long-Qiang Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Guang-Zong Zhao
- Department of Orthopedics, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Xiao-Yan Xu
- Department of Oncology, Qingzhou Hospital of Traditional Chinese Medicine, Qingzhou, Shandong 262500, P.R. China
| | - Jun Fang
- Department of Orthopedics, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Jing-Ming Chen
- Department of Orthopedics, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Ji-Wen Li
- Department of Orthopedics, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Xue-Jian Gao
- Department of Orthopedics, The 89th Hospital of PLA, Weifang, Shandong 261021, P.R. China
| | - Li-Juan Hao
- Department of Urologic Surgery, Yidu Central Hospital of Weifang, Weifang, Shandong 262500, P.R. China
| | - Yun-Zhen Chen
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
14
|
Maegdefessel L, Rayner KJ, Leeper NJ. MicroRNA Regulation of Vascular Smooth Muscle Function and Phenotype. Arterioscler Thromb Vasc Biol 2015; 35:2-6. [DOI: 10.1161/atvbaha.114.304877] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Lars Maegdefessel
- From the Department of Medicine, Center for Molecular Medicine (L8:03), Karolinska Institute, 17176 Stockholm, Sweden (L.M.); Cardiometabolic microRNA Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada (K.J.R.); and Division of Vascular Surgery, Stanford University, CA (N.J.L.)
| | - Katey J. Rayner
- From the Department of Medicine, Center for Molecular Medicine (L8:03), Karolinska Institute, 17176 Stockholm, Sweden (L.M.); Cardiometabolic microRNA Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada (K.J.R.); and Division of Vascular Surgery, Stanford University, CA (N.J.L.)
| | - Nicholas J. Leeper
- From the Department of Medicine, Center for Molecular Medicine (L8:03), Karolinska Institute, 17176 Stockholm, Sweden (L.M.); Cardiometabolic microRNA Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada (K.J.R.); and Division of Vascular Surgery, Stanford University, CA (N.J.L.)
| |
Collapse
|
15
|
Xiao J, Chen X, Xu L, Zhang Y, Yin Q, Wang F. PDGF regulates chondrocyte proliferation through activation of the GIT1- and PLCγ1-mediated ERK1/2 signaling pathway. Mol Med Rep 2014; 10:2409-14. [PMID: 25175053 DOI: 10.3892/mmr.2014.2506] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 03/17/2014] [Indexed: 11/06/2022] Open
Abstract
Studies investigating the effects of cytokines on chondrocytes have significant application potential, since the culture of cartilage cells in vitro is a vital step for cartilage tissue engineering. Platelet-derived growth factor (PDGF), one of the growth factors occurring at the early stage of the healing process of damaged tissue, is critical in bone healing. The present study investigated the effects of the activation of PDGF on cell proliferation, apoptosis and the underlying mechanisms of chondrocytes in vitro. The results indicated that the stimulation of PDGF led to overexpression of the G-protein-coupled receptor kinase interacting protein-1 (GIT1) and promotion of the phosphorylation of phospholipase Cγ1 (PLCγ1). Furthermore, PDGF induced chondrocyte proliferation and inhibited apoptosis via activation of the extracellular signal-regulated kinase (ERK) 1/2 pathway. Following knocking down GIT1 expression by small interfering RNA, phosphorylation of PLCγ1 and activation of the ERK1/2 pathway was no longer promoted by PDGF. In addition, the effects of PDGF on proliferation and apoptosis were suppressed. The expression levels of GIT1 were not affected; however, the phosphorylation of ERK1/2 was suppressed through inhibition of the phosphorylation of PLCγ1 by U73122. The results demonstrated that GIT1 is upstream of PLCγ1. Although the ability of PDGF to induce cell proliferation was inhibited by the inhibition of the ERK1/2 pathway by PD98059, apoptosis was not suppressed. In conclusion, the present study demonstrated that PDGF was able to activate the GIT1‑PLCγ1‑mediated ERK1/2 pathway to control chondrocyte proliferation.
Collapse
Affiliation(s)
- Jin Xiao
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Xuqiong Chen
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Lipeng Xu
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Ying Zhang
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Qingshui Yin
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| | - Fei Wang
- Department of Orthopedics, Liuhuaqiao Hospital, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
16
|
Wu Y, Zhang Y, Yin Q, Xia H, Wang J. Platelet‑derived growth factor promotes osteoblast proliferation by activating G‑protein‑coupled receptor kinase interactor‑1. Mol Med Rep 2014; 10:1349-54. [PMID: 25017023 DOI: 10.3892/mmr.2014.2374] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 03/19/2014] [Indexed: 11/05/2022] Open
Abstract
Platelet‑derived growth factor (PDGF) has been reported to stimulate bone fracture‑healing. Multiple studies have demonstrated that PDGF has a critical role in osteoblast or osteoprogenitor cell activation, although the underlying mechanism remains unclear. Studies have found that G‑protein‑coupled receptor kinase interactor‑1 (GIT1) is activated by PDGF and described as an important factor in bone metabolism. In the present study, the effects of PDGF and GIT1 on the proliferation and apoptosis of osteoblasts were investigated in cultured osteoblasts isolated from rat calvaria with PDGF stimulation and GIT1 small interfering RNA transfection. The results demonstrated that PDGF rapidly stimulated GTI1 expression in osteoblasts, increased osteoblast proliferation and inhibited cell apoptosis. Furthermore, cyclin D1 expression was significantly upregulated, the number of cells in the G0/G1 phase was decreased, while the number in the S phase was increased. In cells with knockdown of GIT1, the change stimulated by PDGF was not evident. The results indicate that, PDGF stimulated GIT1 activation of cyclin D1 expression, thereby promoting osteoblasts to enter the S phase from the stationary G0/G1 phase, leading to the proliferation of osteoblasts.
Collapse
Affiliation(s)
- You Wu
- Department of Traumatic Orthopedics, General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Ying Zhang
- Department of Traumatic Orthopedics, General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Qingshui Yin
- Department of Traumatic Orthopedics, General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Hong Xia
- Department of Traumatic Orthopedics, General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong 510010, P.R. China
| | - Jiajia Wang
- Department of Traumatic Orthopedics, General Hospital of Guangzhou Military Area Command of Chinese PLA, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|