1
|
Slighoua M, Chebaibi M, Mahdi I, Amrati FEZ, Conte R, Cordero MAW, Alotaibi A, Saghrouchni H, Agour A, Zair T, Bari A, Bousta D. The LC-MS/MS Identification and Analgesic and Wound Healing Activities of Lavandula officinalis Chaix: In Vivo and In Silico Approaches. PLANTS (BASEL, SWITZERLAND) 2022; 11:3222. [PMID: 36501262 PMCID: PMC9738568 DOI: 10.3390/plants11233222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 06/17/2023]
Abstract
We earlier emphasized in vivo the lavender plant's (Lavandula officinalis Chaix.) anti-inflammatory and estrogenic activities and described the chemical compositions of its hydro-ethanolic (HE) extract. We used LC-MS/MS and GC-MS analyses to profile the phytochemical composition of the HE extract and to assess the analgesic and wound-healing effects of both the hydro-ethanolic (HE) and polyphenolic (LOP) extracts in vivo and in silico. The analgesic activity was studied using two methods: acetic acid and formalin injections in mice. The wound-healing activity was carried out over 25 days using a burn model in rats. In the in silico study, the polyphenols identified in the plant were docked in the active sites of three enzymes: casein kinase-1, cyclooxygenase-2, and glycogen synthase kinase-3β. The LC-MS/MS identified some phenolic compounds, mainly apigenin, catechin, and myricetin, and the GC-MS analysis revealed the presence of 19 volatile compounds with triazole, D-glucose, hydroxyphenyl, and D-Ribofuranose as the major compounds. The HE and LOP extracts showed significant decreases in abdominal writhes, and the higher licking time of the paw (57.67%) was observed using the LOP extract at 200 mg/kg. Moreover, both extracts showed high healing percentages, i.e., 99.31 and 92.88%, compared to the control groups, respectively. The molecular docking showed that myricetin, amentoflavone, apigenin, and catechin are the most active molecules against the three enzyme receptors. This study sheds light on the potential of L. officinalis Chaix as a source of natural products for pharmaceutical applications for analgesic purposes as well as their utility in promoting burn-healing activity.
Collapse
Affiliation(s)
- Meryem Slighoua
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Mohamed Chebaibi
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy of the Fez, Sidi Mohamed Ben Abdellah University, B.P. 1893, Km 22, Road of Sidi Harazem, Fez 30000, Morocco
| | - Ismail Mahdi
- AgroBioSciences Research Program, Mohammed VI Polytechnic University, Lot 660-Hay Moulay Rachid, Ben-Guerir 43150, Morocco
| | - Fatima Ez-zahra Amrati
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)—CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Mary Anne W. Cordero
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Amal Alotaibi
- Department of Basic Science, College of Medicine, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Hamza Saghrouchni
- Department of Biotechnology, Institute of Natural and Applied Sciences, Çukurova University, Balcali/Sariçam, Adana 01330, Turkey
| | - Abdelkrim Agour
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health and Quality of Life, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, P.O. Box 1796, Fez 30000, Morocco
| | - Touria Zair
- Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay 19 Ismail University, B.P. 11201, Meknes 50070, Morocco
| | - Amina Bari
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Dalila Bousta
- Laboratory of Biotechnology, Environment, Agro-Food and Health (LBEAS), Faculty of Sciences, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| |
Collapse
|
2
|
Mei S, Li L, Zhou X, Xue C, Livingston MJ, Wei Q, Dai B, Mao Z, Mei C, Dong Z. Susceptibility of renal fibrosis in diabetes: Role of hypoxia inducible factor-1. FASEB J 2022; 36:e22477. [PMID: 35881071 PMCID: PMC9386694 DOI: 10.1096/fj.202200845r] [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: 06/04/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 01/09/2023]
Abstract
Diabetes may prevent kidney repair and sensitize the kidney to fibrosis or scar formation. To test this possibility, we examined renal fibrosis induced by unilateral ureteral obstruction (UUO) in diabetic mouse models. Indeed, UUO induced significantly more renal fibrosis in both Akita and STZ-induced diabetic mice than in nondiabetic mice. The diabetic mice also had more apoptosis and interstitial macrophage infiltration during UUO. In vitro, hypoxia induced higher expression of the fibrosis marker protein fibronectin in high glucose-conditioned renal tubular cells than in normal glucose cells. Mechanistically, hypoxia induced significantly more hypoxia-inducible factor-1 α (HIF-1 α) in high glucose cells than in normal glucose cells. Inhibition of HIF-1 attenuated the expression of fibronectin induced by hypoxia in high-glucose cells. Consistently, UUO induced significantly higher HIF-1α expression along with fibrosis in diabetic mice kidneys than in nondiabetic kidneys. The increased expression of fibrosis induced by UUO in diabetic mice was diminished in proximal tubule-HIF-1α-knockout mice. Together, these results indicate that diabetes sensitizes kidney tissues and cells to fibrogenesis probably by enhancing HIF-1 activation.
Collapse
Affiliation(s)
- Shuqin Mei
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Lin Li
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Cheng Xue
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Man J Livingston
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Bing Dai
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zhiguo Mao
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Changlin Mei
- Department of Nephrology, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| |
Collapse
|
3
|
Xiao Y, Zhang Y, Li Y, Peng N, Liu Q, Qiu D, Cho J, Borlongan CV, Yu G. Exosomes Derived From Mesenchymal Stem Cells Pretreated With Ischemic Rat Heart Extracts Promote Angiogenesis via the Delivery of DMBT1. Cell Transplant 2022; 31:9636897221102898. [PMID: 35726847 PMCID: PMC9218457 DOI: 10.1177/09636897221102898] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/26/2022] [Accepted: 05/09/2022] [Indexed: 12/02/2022] Open
Abstract
Mesenchymal stem cell-derived exosomes (MSC-Exos) have been shown to promote angiogenesis. Treating MSCs with ischemic rat brain extracts was sufficient to augment their benefits in stroke. However, no similar analyses of ischemic heart extracts have been performed to date. We aim to determine whether MSC-Exos derived from MSCs pretreated with ischemic rat heart extract were able to promote angiogenesis and to clarify underlying mechanisms. ELISA (enzyme-linked immunosorbent assay) of heart extracts revealed a significant increase of vascular endothelial growth factor (VEGF) at 24 h post-MI (myocardial infarction) modeling, and time-dependent decreases in hypoxia inducible factor-1α (HIF-1α). MTT and wound healing assays revealed human umbilical vein endothelial cells (HUVECs) migration and proliferation increased following MSCE-Exo treatment (exosomes derived from MSC pretreated with ischemic heart extracts of 24 h post-MI) relative to MSCN-Exo treatment (exosomes derived from MSC pretreated with normal heart extracts). Proteomic analyses of MSCE-Exo and MSCN-Exo were conducted to screen for cargo proteins promoting angiogenesis. Result revealed several angiogenesis-related proteins were upregulated in MSCE-Exo, including DMBT1 (deleted in malignant brain tumors 1). When DMBT1 was silenced in MSCs, HUVECs with MSCDMBT1 RNAi-Exo treatment exhibited impaired proliferative and migratory activity and reductions of DMBT1, p-Akt, β-catenin, and VEGF. To explore how ischemic heart extracts took effects, ELISA was conducted showing a significant increase of IL-22 at 24 h post-MI modeling. P-STAT3, IL22RA1, DMBT1, and VEGF proteins were increased in MSCE relative to MSCN, and VEGF and DMBT1 were increased in MSCE-Exos. Together, these suggest that IL-22 upregulation in ischemic heart extracts can increase DMBT1 in MSCs. Exosomes derived from those MSCs deliver DMBT1 to HUVECs, thereby enhancing their migratory and proliferative activity.
Collapse
Affiliation(s)
- Yi Xiao
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Ye Zhang
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Yuzhang Li
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Nanyin Peng
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Qin Liu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Danyang Qiu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| | - Justin Cho
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Cesario V. Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Guolong Yu
- Division of Cardiovascular, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
4
|
Nagavally RR, Sunilkumar S, Akhtar M, Trombetta LD, Ford SM. Chrysin Ameliorates Cyclosporine-A-Induced Renal Fibrosis by Inhibiting TGF-β 1-Induced Epithelial-Mesenchymal Transition. Int J Mol Sci 2021; 22:ijms221910252. [PMID: 34638597 PMCID: PMC8508845 DOI: 10.3390/ijms221910252] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 01/27/2023] Open
Abstract
Cyclosporine A (CsA) is a nephrotoxicant that causes fibrosis via induction of epithelial-mesenchymal transition (EMT). The flavonoid chrysin has been reported to have anti-fibrotic activity and inhibit signaling pathways that are activated during EMT. This study investigated the nephroprotective role of chrysin in the prevention of CsA-induced renal fibrosis and elucidated a mechanism of inhibition against CsA-induced EMT in proximal tubule cells. Treatment with chrysin prevented CsA-induced renal dysfunction in Sprague Dawley rats measured by blood urea nitrogen (BUN), serum creatinine and creatinine clearance. Chrysin inhibited CsA-induced tubulointerstitial fibrosis, characterized by reduced tubular damage and collagen deposition. In vitro, chrysin significantly inhibited EMT in LLC-PK1 cells, evidenced by inhibition of cell migration, decreased collagen expression, reduced presence of mesenchymal markers and elevated epithelial junction proteins. Furthermore, chrysin co-treatment diminished CsA-induced TGF-β1 signaling pathways, decreasing Smad 3 phosphorylation which lead to a subsequent reduction in Snail expression. Chrysin also inhibited activation of the Akt/ GSK-3β pathway. Inhibition of both pathways diminished the cytosolic accumulation of β-catenin, a known trigger for EMT. In conclusion, flavonoids such as chrysin offer protection against CsA-induced renal dysfunction and interstitial fibrosis. Chrysin was shown to inhibit CsA-induced TGF-β1-dependent EMT in proximal tubule cells by modulation of Smad-dependent and independent signaling pathways.
Collapse
Affiliation(s)
- Rohan Reddy Nagavally
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (R.R.N.); (S.S.); (M.A.); (L.D.T.)
- Viatris Inc., 1000 Mylan Blvd, Canonsburg, PA 15317, USA
| | - Siddharth Sunilkumar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (R.R.N.); (S.S.); (M.A.); (L.D.T.)
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Mumtaz Akhtar
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (R.R.N.); (S.S.); (M.A.); (L.D.T.)
| | - Louis D. Trombetta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (R.R.N.); (S.S.); (M.A.); (L.D.T.)
| | - Sue M. Ford
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY 11439, USA; (R.R.N.); (S.S.); (M.A.); (L.D.T.)
- Correspondence: ; Tel.: +1-71-8990-6220
| |
Collapse
|
5
|
Rao P, Qiao X, Hua W, Hu M, Tahan M, Chen T, Yu H, Ren X, Cao Q, Wang Y, Yang Y, Wang YM, Lee VW, Alexander SI, Harris DC, Zheng G. Promotion of β-Catenin/Forkhead Box Protein O Signaling Mediates Epithelial Repair in Kidney Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:993-1009. [PMID: 33753026 PMCID: PMC8351131 DOI: 10.1016/j.ajpath.2021.03.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 03/01/2021] [Accepted: 03/05/2021] [Indexed: 01/24/2023]
Abstract
Fibrosis is characterized by progressively excessive deposition of matrix components and may lead to organ failure. Transforming growth factor-β (TGF-β) is a key cytokine involved in tissue repair and fibrosis. TGF-β's profibrotic signaling pathways converge at activation of β-catenin. β-Catenin is an important transcription cofactor whose function depends on its binding partner. Promoting β-catenin binding to forkhead box protein O (Foxo) via inhibition of its binding to T-cell factor (TCF) reduces kidney fibrosis in experimental murine models. Herein, we investigated whether β-catenin/Foxo diverts TGF-β signaling from profibrotic to physiological epithelial healing. In an in vitro model of wound healing (scratch assay), and in an in vivo model of kidney injury, unilateral renal ischemia reperfusion, TGF-β treatment in combination with either ICG-001 or iCRT3 (β-catenin/TCF inhibitors) increased β-catenin/Foxo interaction, increased scratch closure by increased cell proliferation and migration, reduced the TGF-β-induced mesenchymal differentiation, and healed the ischemia reperfusion injury with less fibrosis. In addition, administration of ICG-001 or iCRT3 reduced the contractile activity induced by TGF-β in C1.1 cells. Together, our results indicate that redirection of β-catenin binding from TCF to Foxo promotes β-catenin/Foxo-mediated epithelial repair. Targeting β-catenin/Foxo may rebuild normal structure of injured kidney.
Collapse
Affiliation(s)
- Padmashree Rao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Xi Qiao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Nephrology, Shanxi Medical University, Taiyuan, China
| | - Winston Hua
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Min Hu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Mariah Tahan
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Titi Chen
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Hong Yu
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Xiaojun Ren
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Nephrology, Shanxi Medical University, Taiyuan, China
| | - Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia
| | - Ying Yang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Nephrology, Shanxi Medical University, Taiyuan, China
| | - Yuan M Wang
- Centre for Kidney Research Children's Hospital at Westmead, Australia
| | - Vincent W Lee
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Nephrology, Westmead Hospital, Australia
| | | | - David C Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia; Department of Nephrology, Westmead Hospital, Australia
| | - Guoping Zheng
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
6
|
Ardalan M, Hejazian SM, Sharabiyani HF, Farnood F, Ghafari Aghdam A, Bastami M, Ahmadian E, Zununi Vahed S, Cucchiarini M. Dysregulated levels of glycogen synthase kinase-3β (GSK-3β) and miR-135 in peripheral blood samples of cases with nephrotic syndrome. PeerJ 2020; 8:e10377. [PMID: 33362958 PMCID: PMC7749650 DOI: 10.7717/peerj.10377] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/26/2020] [Indexed: 12/14/2022] Open
Abstract
Background Glycogen synthase kinase-3 (GSK-3β) is a serine/threonine kinase with multifunctions in various physiological procedures. Aberrant level of GSK-3β in kidney cells has a harmful role in podocyte injury. Methods In this article, the expression levels of GSK-3β and one of its upstream regulators, miR-135a-5p, were measured in peripheral blood mononuclear cells (PBMCs) of cases with the most common types of nephrotic syndrome (NS); focal segmental glomerulosclerosis (FSGS) and membranous glomerulonephritis (MGN). In so doing, fifty-two cases along with twenty-four healthy controls were included based on the strict criteria. Results Levels of GSK-3β mRNA and miR-135 were measured with quantitative real-time PCR. There were statistically significant increases in GSK-3β expression level in NS (P = 0.001), MGN (P = 0.002), and FSGS (P = 0.015) groups compared to the control group. Dysregulated levels of miR-135a-5p in PBMCs was not significant between the studied groups. Moreover, a significant decrease was observed in the expression level of miR-135a-5p in the plasma of patients with NS (P = 0.020), MGN (P = 0.040), and FSGS (P = 0.046) compared to the control group. ROC curve analysis approved a diagnostic power of GSK-3β in discriminating patients from healthy controls (AUC: 0.72, P = 0.002) with high sensitivity and specificity. Conclusions Dysregulated levels of GSK-3β and its regulator miR-135a may participate in the pathogenesis of NS with different etiology. Therefore, more research is needed for understanding the relationship between them.
Collapse
Affiliation(s)
| | - Seyyedeh Mina Hejazian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farahnoosh Farnood
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Ghafari Aghdam
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Bastami
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Universität des Saarlandes, Homburg/Saar, Germany
| |
Collapse
|
7
|
Huang X, Liang P, Jiang B, Zhang P, Yu W, Duan M, Guo L, Cui X, Huang M, Huang X. Hyperbaric oxygen potentiates diabetic wound healing by promoting fibroblast cell proliferation and endothelial cell angiogenesis. Life Sci 2020; 259:118246. [DOI: 10.1016/j.lfs.2020.118246] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/31/2022]
|
8
|
Liu L, Zhang P, Bai M, He L, Zhang L, Liu T, Yang Z, Duan M, Liu M, Liu B, Du R, Qian Q, Sun S. p53 upregulated by HIF-1α promotes hypoxia-induced G2/M arrest and renal fibrosis in vitro and in vivo. J Mol Cell Biol 2020; 11:371-382. [PMID: 30032308 PMCID: PMC7727266 DOI: 10.1093/jmcb/mjy042] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 04/21/2018] [Accepted: 07/17/2018] [Indexed: 12/21/2022] Open
Abstract
Hypoxia plays an important role in the genesis and progression of renal fibrosis. The underlying mechanisms, however, have not been sufficiently elucidated. We examined the role of p53 in hypoxia-induced renal fibrosis in cell culture (human and rat renal tubular epithelial cells) and a mouse unilateral ureteral obstruction (UUO) model. Cell cycle of tubular cells was determined by flow cytometry, and the expression of profibrogenic factors was determined by RT-PCR, immunohistochemistry, and western blotting. Chromatin immunoprecipitation and luciferase reporter experiments were performed to explore the effect of HIF-1α on p53 expression. We showed that, in hypoxic tubular cells, p53 upregulation suppressed the expression of CDK1 and cyclins B1 and D1, leading to cell cycle (G2/M) arrest (or delay) and higher expression of TGF-β, CTGF, collagens, and fibronectin. p53 suppression by siRNA or by a specific p53 inhibitor (PIF-α) triggered opposite effects preventing the G2/M arrest and profibrotic changes. In vivo experiments in the UUO model revealed similar antifibrotic results following intraperitoneal administration of PIF-α (2.2 mg/kg). Using gain-of-function, loss-of-function, and luciferase assays, we further identified an HRE3 region on the p53 promoter as the HIF-1α-binding site. The HIF-1α–HRE3 binding resulted in a sharp transcriptional activation of p53. Collectively, we show the presence of a hypoxia-activated, p53-responsive profibrogenic pathway in the kidney. During hypoxia, p53 upregulation induced by HIF-1α suppresses cell cycle progression, leading to the accumulation of G2/M cells, and activates profibrotic TGF-β and CTGF-mediated signaling pathways, causing extracellular matrix production and renal fibrosis.
Collapse
Affiliation(s)
- Limin Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ming Bai
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lijie He
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lei Zhang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Ting Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Zhen Yang
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Menglu Duan
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Minna Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Baojian Liu
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Rui Du
- Department of Radiation Oncology, Navy General Hospital, Beijing, China
| | - Qi Qian
- Department of Medicine, Mayo Clinic College of Medicine and Mayo Graduate School, Rochester, MN, USA
| | - Shiren Sun
- Department of Nephrology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.,State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
9
|
Kang J, Wang J, Tian J, Shi R, Jia H, Wang Y. The emerging role of EGFL6 in angiogenesis and tumor progression. Int J Med Sci 2020; 17:1320-1326. [PMID: 32624687 PMCID: PMC7330666 DOI: 10.7150/ijms.45129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/29/2020] [Indexed: 01/05/2023] Open
Abstract
Epidermal growth factor-like domain-containing protein 6 (EGFL6) belongs to the epidermal growth factor (EGF) superfamily. EGFL6 is expressed at higher levels in embryos and various malignant tumors than in normal tissues. Recent studies suggest that EGFL6 participates in the development of a variety of tumors. In this review, we summarize findings that support the role for EGFL6 in tumor proliferation, invasion and migration. Furthermore, our review results indicate the mechanism of EGFL6 activity angiogenesis. We also describe work toward the preparation of monoclonal antibodies against EGFL6. Altogether, the work of this review promotes understanding of the role of EGFL6 in tumor development, the mechanism of that action, and the potential of EGFL6 as a therapeutic target for tumor prevention and treatment.
Collapse
Affiliation(s)
- Jing Kang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juanjuan Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jihua Tian
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruyi Shi
- Department of Cell biology and Genetics, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hongyan Jia
- Department of General Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
10
|
Role of the PI3K/AKT (mTOR and GSK3β) signalling pathway and photobiomodulation in diabetic wound healing. Cytokine Growth Factor Rev 2019; 50:52-59. [DOI: 10.1016/j.cytogfr.2019.03.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 03/11/2019] [Indexed: 12/14/2022]
|
11
|
HIF stabilization inhibits renal epithelial cell migration and is associated with cytoskeletal alterations. Sci Rep 2018; 8:9497. [PMID: 29934555 PMCID: PMC6015081 DOI: 10.1038/s41598-018-27918-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/13/2018] [Indexed: 11/23/2022] Open
Abstract
Acute kidney injury (AKI) is a common and potentially lethal complication in the hospitalized patients, with hypoxic injury being as a major cause. The loss of renal tubular epithelial cells (TEC), one of the AKI hallmarks, is potentially followed by tubular regeneration process orchestrated by the remaining uninjured TECs that undergo proliferation and migration. In this study, we used human primary TEC to investigate the initiation of tubular cell migration and associated cytoskeletal alterations in response to pharmacological HIF stabilization which resembles the pathophysiology of hypoxia. Tubular cells have been shown to migrate as cohorts in a wound healing assay. Importantly, cells of distal tubular origin moved faster than those of proximal origin. HIF stabilization impaired TEC migration, which was confirmed by live single cell tracking. HIF stabilization significantly reduced tubular cell migration velocity and promoted cell spreading. In contrast to the control conditions, HIF stabilization induced actin filaments rearrangement and cell adhesion molecules including paxillin and focal adhesion kinase. Condensed bundling of keratin fibers was also observed, while the expression of different types of keratins, phosphorylation of keratin 18, and the microtubule structure were not altered. In summary, HIF stabilization reduced the ability of renal tubular cells to migrate and led to cytoskeleton reorganization. Our data suggested an important involvement of HIF stabilization during the epithelial migration underlying the mechanism of renal regeneration in response to AKI.
Collapse
|
12
|
Renal Tubule Repair: Is Wnt/β-Catenin a Friend or Foe? Genes (Basel) 2018; 9:genes9020058. [PMID: 29364168 PMCID: PMC5852554 DOI: 10.3390/genes9020058] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/14/2018] [Accepted: 01/16/2018] [Indexed: 12/15/2022] Open
Abstract
Wnt/β-catenin signaling is extremely important for proper kidney development. This pathway is also upregulated in injured renal tubular epithelia, both in acute kidney injury and chronic kidney disease. The renal tubular epithelium is an important target of kidney injury, and its response (repair versus persistent injury) is critical for determining whether tubulointerstitial fibrosis, the hallmark of chronic kidney disease, develops. This review discusses how Wnt/β-catenin signaling in the injured tubular epithelia promotes either repair or fibrosis after kidney injury. There is data suggesting that epithelial Wnt/β-catenin signaling is beneficial in acute kidney injury and important in tubular progenitors responsible for epithelial repair. The role of Wnt/β-catenin signaling in chronically injured epithelia is less clear. There is convincing data that Wnt/β-catenin signaling in interstitial fibroblasts and pericytes contributes to the extracellular matrix accumulation that defines fibrosis. However, some recent studies question whether Wnt/β-catenin signaling in chronically injured epithelia actually promotes fibrosis or repair.
Collapse
|
13
|
Noh K, Mangala LS, Han HD, Zhang N, Pradeep S, Wu SY, Ma S, Mora E, Rupaimoole R, Jiang D, Wen Y, Shahzad MMK, Lyons Y, Cho M, Hu W, Nagaraja AS, Haemmerle M, Mak CSL, Chen X, Gharpure KM, Deng H, Xiong W, Kingsley CV, Liu J, Jennings N, Birrer MJ, Bouchard RR, Lopez-Berestein G, Coleman RL, An Z, Sood AK. Differential Effects of EGFL6 on Tumor versus Wound Angiogenesis. Cell Rep 2017; 21:2785-2795. [PMID: 29212026 PMCID: PMC5749980 DOI: 10.1016/j.celrep.2017.11.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 09/18/2017] [Accepted: 11/02/2017] [Indexed: 11/25/2022] Open
Abstract
Angiogenesis inhibitors are important for cancer therapy, but clinically approved anti-angiogenic agents have shown only modest efficacy and can compromise wound healing. This necessitates the development of novel anti-angiogenesis therapies. Here, we show significantly increased EGFL6 expression in tumor versus wound or normal endothelial cells. Using a series of in vitro and in vivo studies with orthotopic and genetically engineered mouse models, we demonstrate the mechanisms by which EGFL6 stimulates tumor angiogenesis. In contrast to its antagonistic effects on tumor angiogenesis, EGFL6 blockage did not affect normal wound healing. These findings have significant implications for development of anti-angiogenesis therapies.
Collapse
Affiliation(s)
- Kyunghee Noh
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Gene Therapy Research Unit, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hee-Dong Han
- Department of Immunology, School of Medicine, Konkuk University, Chungju 380-701, South Korea
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sherry Y Wu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shaolin Ma
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Edna Mora
- Department of Surgery, University of Puerto Rico, San Juan 00936, Puerto Rico; University of Puerto Rico Comprehensive Cancer Center, San Juan 00936, Puerto Rico; Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77584, USA
| | - Rajesha Rupaimoole
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dahai Jiang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mian M K Shahzad
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yasmin Lyons
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - MinSoon Cho
- Department of Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Archana S Nagaraja
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Monika Haemmerle
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Celia S L Mak
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiuhui Chen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kshipra M Gharpure
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Charles V Kingsley
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael J Birrer
- University of Alabama Comprehensive Cancer Center, Birmingham, AL 35294, USA
| | - Richard R Bouchard
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gabriel Lopez-Berestein
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
14
|
He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA, Dong Z. AKI on CKD: heightened injury, suppressed repair, and the underlying mechanisms. Kidney Int 2017; 92:1071-1083. [PMID: 28890325 DOI: 10.1016/j.kint.2017.06.030] [Citation(s) in RCA: 277] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected. Although AKI-to-CKD transition has been intensively studied, the information of AKI on CKD is very limited. Nonetheless, AKI, when occurring in patients with CKD, is known to be more severe and difficult to recover. CKD is associated with significant changes in cell signaling in kidney tissues, including the activation of transforming growth factor-β, p53, hypoxia-inducible factor, and major developmental pathways. At the cellular level, CKD is characterized by mitochondrial dysfunction, oxidative stress, and aberrant autophagy. At the tissue level, CKD is characterized by chronic inflammation and vascular dysfunction. These pathologic changes may contribute to the heightened sensitivity of, and nonrecovery from, AKI in patients with CKD.
Collapse
Affiliation(s)
- Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Jing Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Mixuan Yi
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manjeri A Venkatachalam
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA.
| |
Collapse
|
15
|
Zhang W, Zhou X, Yao Q, Liu Y, Zhang H, Dong Z. HIF-1-mediated production of exosomes during hypoxia is protective in renal tubular cells. Am J Physiol Renal Physiol 2017; 313:F906-F913. [PMID: 28679592 DOI: 10.1152/ajprenal.00178.2017] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/21/2017] [Accepted: 06/26/2017] [Indexed: 11/22/2022] Open
Abstract
Exosomes are nano-sized vesicles produced and secreted by cells to mediate intercellular communication. The production and function of exosomes in kidney tissues and cells remain largely unclear. Hypoxia is a common pathophysiological condition in kidneys. This study was designed to characterize exosome production during hypoxia of rat renal proximal tubular cells (RPTCs), investigate the regulation by hypoxia-inducible factor-1 (HIF-1), and determine the effect of the exosomes on ATP-depletion-induced tubular cell injury. Hypoxia did not change the average sizes of exosomes secreted by RPTCs, but it significantly increased exosome production in a time-dependent manner. HIF-1 induction with dimethyloxalylglycine also promoted exosome secretion, whereas pharmacological and genetic suppression of HIF-1 abrogated the increase of exosome secretion under hypoxia. The exosomes from hypoxic RPTCs had inhibitory effects on apoptosis of RPTCs following ATP depletion. The protective effects were lost in the exosomes from HIF-1α knockdown cells. It is concluded that hypoxia stimulates exosome production and secretion in renal tubular cells. The exosomes from hypoxic cells are protective against renal tubular cell injury. HIF-1 mediates exosome production during hypoxia and contributes to the cytoprotective effect of the exosomes.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| | - Xiangjun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| | - Qisheng Yao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yutao Liu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia; and
| | - Hao Zhang
- Department of Nephrology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, Georgia; and.,Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
16
|
Peng J, Zheng H, Wang X, Cheng Z. Upregulation of TLR4 via PKC activation contributes to impaired wound healing in high-glucose-treated kidney proximal tubular cells. PLoS One 2017; 12:e0178147. [PMID: 28542370 PMCID: PMC5443579 DOI: 10.1371/journal.pone.0178147] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 05/09/2017] [Indexed: 12/15/2022] Open
Abstract
Acute kidney injury (AKI) leads to a worse prognosis in diabetic patients compared with prognoses in non-diabetic patients, but whether and how diabetes affects kidney repair after AKI remains unknown. Here, we used scratch-wound healing and transwell migration models to examine whether and how wound healing is affected by high glucose levels in cultured kidney proximal tubular cells (RPTC). The results show that scratch-wound healing and transwell migration were significantly slower in high-glucose-treated kidney tubular cells (30 mM glucose) than in low-glucose-treated cells (5.5 mM). Toll-like receptor 4 (TLR4), MyD88, phospho-protein kinase C (PKC), phospho-p38 MAPK and monocyte chemoattractant protein-1 (MCP-1) mRNA levels were upregulated after high glucose treatments. Staurosporine, a selective PKC inhibitor, inhibited TLR4, MyD88 and p-p38 upregulation in the high-glucose-treated cells, indicating the involvement of PKC in high-glucose-induced TLR4 upregulation. The pharmacological inhibition of TLR4 or shRNA-mediated TLR4 knockdown improved wound healing and transwell migration in high-glucose-treated RPTC. In contrast, the overexpression of TLR4 in low-glucose-treated RPTC suppressed wound healing, mimicking the effects of high glucose levels. These results suggest that the upregulation of TLR4 expression via PKC activation contributes to defective wound healing in high-glucose-treated kidney tubular cells.
Collapse
Affiliation(s)
- Jianping Peng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Hang Zheng
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
- * E-mail:
| | - Xia Wang
- Department of Internal Medicine, Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zhixiang Cheng
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Flampouri E, Mavrikou S, Mouzaki-Paxinou AC, Kintzios S. Alterations of cellular redox homeostasis in cultured fibroblast-like renal cells upon exposure to low doses of cytochrome bc1 complex inhibitor kresoxim-methyl. Biochem Pharmacol 2016; 113:97-109. [DOI: 10.1016/j.bcp.2016.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/01/2016] [Indexed: 12/18/2022]
|
18
|
Xu W, Ge Y, Liu Z, Gong R. Glycogen synthase kinase 3β dictates podocyte motility and focal adhesion turnover by modulating paxillin activity: implications for the protective effect of low-dose lithium in podocytopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 184:2742-56. [PMID: 25239564 DOI: 10.1016/j.ajpath.2014.06.027] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 05/19/2014] [Accepted: 06/10/2014] [Indexed: 12/27/2022]
Abstract
Aberrant focal adhesion turnover is centrally involved in podocyte actin cytoskeleton disorganization and foot process effacement. The structural and dynamic integrity of focal adhesions is orchestrated by multiple cell signaling molecules, including glycogen synthase kinase 3β (GSK3β), a multitasking kinase lately identified as a mediator of kidney injury. However, the role of GSK3β in podocytopathy remains obscure. In doxorubicin (Adriamycin)-injured podocytes, lithium, a GSK3β inhibitor and neuroprotective mood stabilizer, obliterated the accelerated focal adhesion turnover, rectified podocyte hypermotility, and restored actin cytoskeleton integrity. Mechanistically, lithium counteracted the doxorubicin-elicited GSK3β overactivity and the hyperphosphorylation and overactivation of paxillin, a focal adhesion-associated adaptor protein. Moreover, forced expression of a dominant negative kinase dead mutant of GSK3β highly mimicked, whereas ectopic expression of a constitutively active GSK3β mutant abolished, the effect of lithium in doxorubicin-injured podocytes, suggesting that the effect of lithium is mediated, at least in part, through inhibition of GSK3β. Furthermore, paxillin interacted with GSK3β and served as its substrate. In mice with doxorubicin nephropathy, a single low dose of lithium ameliorated proteinuria and glomerulosclerosis. Consistently, lithium therapy abrogated GSK3β overactivity, blunted paxillin hyperphosphorylation, and reinstated actin cytoskeleton integrity in glomeruli associated with an early attenuation of podocyte foot process effacement. Thus, GSK3β-modulated focal adhesion dynamics might serve as a novel therapeutic target for podocytopathy.
Collapse
Affiliation(s)
- Weiwei Xu
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China; Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island
| | - Yan Ge
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island
| | - Zhihong Liu
- National Clinical Research Center of Kidney Disease, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Rujun Gong
- Division of Kidney Disease and Hypertension, Department of Medicine, Rhode Island Hospital, Brown University School of Medicine, Providence, Rhode Island.
| |
Collapse
|
19
|
Chaudhary A, Bag S, Barui A, Banerjee P, Chatterjee J. Honey dilution impact on in vitro wound healing: Normoxic and hypoxic condition. Wound Repair Regen 2015; 23:412-22. [PMID: 25845442 DOI: 10.1111/wrr.12297] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 03/26/2015] [Indexed: 12/15/2022]
Affiliation(s)
- Amrita Chaudhary
- School of Medical Science and Technology, Indian Institute of Technology- Kharagpur; Kharagpur West Bengal India
| | - Swarnendu Bag
- School of Medical Science and Technology, Indian Institute of Technology- Kharagpur; Kharagpur West Bengal India
| | - Ananya Barui
- Centre for Healthcare Science and Technology; Indian Institute of Engineering Science and Technology-Shibpur; Howrah West Bengal India
| | - Provas Banerjee
- Banerjees’ Biomedical Research Foundation-Sainthia; Birbhum West Bengal India
| | - Jyotirmoy Chatterjee
- School of Medical Science and Technology, Indian Institute of Technology- Kharagpur; Kharagpur West Bengal India
| |
Collapse
|
20
|
Litherland GJ, Hui W, Elias MS, Wilkinson DJ, Watson S, Huesa C, Young DA, Rowan AD. Glycogen synthase kinase 3 inhibition stimulates human cartilage destruction and exacerbates murine osteoarthritis. Arthritis Rheumatol 2014; 66:2175-87. [PMID: 24757033 DOI: 10.1002/art.38681] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 04/17/2014] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To assess the role of glycogen synthase kinase 3 (GSK-3) as a regulator of cartilage destruction in human tissue and a murine model of osteoarthritis (OA). METHODS Surgical destabilization of the medial meniscus (DMM) was performed to induce experimental murine OA, and joint damage was assessed histologically. Bovine nasal and human OA cartilage samples were incubated with interleukin-1 (IL-1) plus oncostatin M (OSM) and GSK-3 inhibitor. Collagen and proteoglycan release was assessed by hydroxyproline measurement and dye binding assay, collagenase activity was assessed by bioassay, and gene expression was analyzed by real-time polymerase chain reaction. Human articular chondrocytes were isolated by enzymatic digestion and cultured prior to gene silencing and immunoblotting of cell lysates and nuclear fractions. RESULTS Mice treated with GSK-3 inhibitor exhibited significantly greater cartilage damage compared with sham-operated control mice. GSK-3 inhibition in bovine cartilage dramatically accelerated IL-1 plus OSM-stimulated degradation, concomitant with a profound increase in collagenase activity. GSK-3 inhibitor induced collagen release from human OA cartilage in the presence of IL-1 plus OSM and increased proteoglycan loss. Gene expression profiling of resorbing OA cartilage revealed a marked procatabolic switch in gene expression upon GSK-3 inhibition. This was mirrored in human articular chondrocytes following GSK3 silencing, particularly with the GSK-3β isoform. GSK-3 inhibition or silencing led to enhanced IL-1 plus OSM-stimulated abundance and activity of Jun, and silencing of c-jun ameliorated GSK-3 inhibitor-mediated procatabolic gene expression. CONCLUSION GSK-3 is an important regulator of matrix metalloproteinase (MMP)-mediated joint destruction, the inhibition of which by proinflammatory stimuli de-represses catabolic gene expression. Therapeutic strategies that maintain cartilage GSK-3 activity may therefore help curtail aberrant MMP activity during pathologic joint destruction.
Collapse
|
21
|
Mennerich D, Dimova EY, Kietzmann T. Direct phosphorylation events involved in HIF-α regulation: the role of GSK-3β. HYPOXIA 2014; 2:35-45. [PMID: 27774465 PMCID: PMC5045055 DOI: 10.2147/hp.s60703] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hypoxia-inducible factors (HIFs), consisting of α- and β-subunits, are critical regulators of the transcriptional response to hypoxia under both physiological and pathological conditions. To a large extent, the protein stability and the recruitment of coactivators to the C-terminal transactivation domain of the HIF α-subunits determine overall HIF activity. The regulation of HIF α-subunit protein stability and coactivator recruitment is mainly achieved by oxygen-dependent posttranslational hydroxylation of conserved proline and asparagine residues, respectively. Under hypoxia, the hydroxylation events are inhibited and HIF α-subunits stabilize, translocate to the nucleus, dimerize with the β-subunits, and trigger a transcriptional response. However, under normal oxygen conditions, HIF α-subunits can be activated by various growth and coagulation factors, hormones, cytokines, or stress factors implicating the involvement of different kinase pathways in their regulation, thereby making HIF-α-regulating kinases attractive therapeutic targets. From the kinases known to regulate HIF α-subunits, only a few phosphorylate HIF-α directly. Here, we review the direct phosphorylation of HIF-α with an emphasis on the role of glycogen synthase kinase-3β and the consequences for HIF-1α function.
Collapse
Affiliation(s)
- Daniela Mennerich
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Elitsa Y Dimova
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
22
|
Bao H, Ge Y, Wang Z, Zhuang S, Dworkin L, Peng A, Gong R. Delayed administration of a single dose of lithium promotes recovery from AKI. J Am Soc Nephrol 2014; 25:488-500. [PMID: 24408869 DOI: 10.1681/asn.2013040350] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Evidence suggests that glycogen synthase kinase 3β (GSK3β) contributes to AKI; however, its role in post-AKI kidney repair remains uncertain. Here, delayed treatment with a single dose of lithium, a selective inhibitor of GSK3β and a US Food and Drug Administration-approved mood stabilizer, accelerated recovery of renal function, promoted repopulation of renal tubular epithelia, and improved kidney repair in murine models of cisplatin- and ischemia/reperfusion-induced AKI. These effects associated with reduced GSK3β activity and elevated expression of proproliferative molecules, including cyclin D1, c-Myc, and hypoxia-inducible factor 1α (HIF-1α), in renal tubular epithelia. In cultured renal tubular cells, cisplatin exposure led to transient repression of GSK3β activity followed by a prolonged upregulation of activity. Rescue treatment with lithium inhibited GSK3β activity, enhanced nuclear expression of cyclin D1, c-Myc, and HIF-1α, and boosted cellular proliferation. Similarly, ectopic expression of a kinase-dead mutant of GSK3β enhanced the expression of cyclin D1, c-Myc, and HIF-1α and amplified cellular proliferation after cisplatin injury, whereas forced expression of a constitutively active mutant of GSK3β abrogated the effects of lithium. Mechanistically, GSK3β colocalized and physically interacted with cyclin D1, c-Myc, and HIF-1α in tubular cells. In silico analysis revealed that cyclin D1, c-Myc, and HIF-1α harbor putative GSK3β consensus phosphorylation motifs, implying GSK3β-directed phosphorylation and subsequent degradation of these molecules. Notably, cotreatment with lithium enhanced the proapoptotic effects of cisplatin in cultured colon cancer cells. Collectively, our findings suggest that pharmacologic targeting of GSK3β by lithium may be a novel therapeutic strategy to improve renal salvage after AKI.
Collapse
Affiliation(s)
- Hui Bao
- Department of Nephrology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; and
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Using conditional knockout models, Zhou et al. firmly establish a renoprotective role of β-catenin in acute kidney injury. Although β-catenin is protective at the injury phase, whether it helps kidney repair remains in question. In a renal cell scratch model, β-catenin suppresses wound healing. Moreover, continuous activation of β-catenin may lead to renal fibrosis. Further investigation should elucidate the distinct roles played by β-catenin and related signaling in kidney injury, repair, and fibrosis.
Collapse
|
24
|
Borges FT, Melo SA, Özdemir BC, Kato N, Revuelta I, Miller CA, Gattone VH, LeBleu VS, Kalluri R. TGF-β1-containing exosomes from injured epithelial cells activate fibroblasts to initiate tissue regenerative responses and fibrosis. J Am Soc Nephrol 2012; 24:385-92. [PMID: 23274427 DOI: 10.1681/asn.2012101031] [Citation(s) in RCA: 333] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hypoxia is associated with tissue injury and fibrosis but its functional role in fibroblast activation and tissue repair/regeneration is unknown. Using kidney injury as a model system, we demonstrate that injured epithelial cells produce an increased number of exosomes with defined genetic information to activate fibroblasts. Exosomes released by injured epithelial cells promote proliferation, α-smooth muscle actin expression, F-actin expression, and type I collagen production in fibroblasts. Fibroblast activation is dependent on exosomes delivering TGF-β1 mRNA among other yet to be identified moieties. This study suggests that TGF-β1 mRNA transported by exosomes constitutes a rapid response to initiate tissue repair/regenerative responses and activation of fibroblasts when resident parenchyma is injured. The results also inform potential utility of exosome-targeted therapies to control tissue fibrosis.
Collapse
Affiliation(s)
- Fernanda T Borges
- Division of Matrix Biology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Howard C, Tao S, Yang HC, Fogo AB, Woodgett JR, Harris RC, Rao R. Specific deletion of glycogen synthase kinase-3β in the renal proximal tubule protects against acute nephrotoxic injury in mice. Kidney Int 2012; 82:1000-9. [PMID: 22785175 PMCID: PMC3472082 DOI: 10.1038/ki.2012.239] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Renal proximal tubular damage and repair are hallmarks of acute kidney injury. Because glycogen synthase kinase-3β (GSK-3β) is an important cellular regulator of survival and proliferation, we determined its role during injury and recovery of proximal tubules in a mercuric chloride-induced nephrotoxic model of acute kidney injury. Renal proximal tubule-specific GSK-3β knockout mice exposed to mercuric chloride had improved survival and renal function compared to wild type mice. Apoptosis, measured by TUNEL staining, Bax activation, and caspase 3 cleavage were all reduced in the knockout mice. The restoration of renal structure, function, and cell proliferation was also accelerated in the GSK-3β knockout mice. This enhanced repair, evidenced by increased Ki-67 and BrdU staining, along with increased cyclin D1 and c-myc levels, was recapitulated by treatment of wild type mice with the small-molecule GSK-3 inhibitor TDZD-8 following injury. This confirmed that hastened repair in the knockout mice was not merely due to lower initial injury levels. Thus, inhibition of GSK-3β prior to nephrotoxic insult protects from renal injury. Such treatment after acute kidney injury may accelerate repair and regeneration.
Collapse
Affiliation(s)
- Christiana Howard
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | | | | | | | | | | |
Collapse
|