1
|
Ramos C, Gerakopoulos V, Oehler R. Metastasis-associated fibroblasts in peritoneal surface malignancies. Br J Cancer 2024; 131:407-419. [PMID: 38783165 PMCID: PMC11300623 DOI: 10.1038/s41416-024-02717-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/06/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024] Open
Abstract
Over decades, peritoneal surface malignancies (PSMs) have been associated with limited treatment options and poor prognosis. However, advancements in perioperative systemic chemotherapy, cytoreductive surgery (CRS), and hyperthermic intraperitoneal chemotherapy (HIPEC) have significantly improved clinical outcomes. PSMs predominantly result from the spread of intra-abdominal neoplasia, which then form secondary peritoneal metastases. Colorectal, ovarian, and gastric cancers are the most common contributors. Despite diverse primary origins, the uniqueness of the peritoneum microenvironment shapes the common features of PSMs. Peritoneal metastization involves complex interactions between tumour cells and the peritoneal microenvironment. Fibroblasts play a crucial role, contributing to tumour development, progression, and therapy resistance. Peritoneal metastasis-associated fibroblasts (MAFs) in PSMs exhibit high heterogeneity. Single-cell RNA sequencing technology has revealed that immune-regulatory cancer-associated fibroblasts (iCAFs) seem to be the most prevalent subtype in PSMs. In addition, other major subtypes as myofibroblastic CAFs (myCAFs) and matrix CAFs (mCAFs) were frequently observed across PSMs studies. Peritoneal MAFs are suggested to originate from mesothelial cells, submesothelial fibroblasts, pericytes, endothelial cells, and omental-resident cells. This plasticity and heterogeneity of CAFs contribute to the complex microenvironment in PSMs, impacting treatment responses. Understanding these interactions is crucial for developing targeted and local therapies to improve PSMs patient outcomes.
Collapse
Affiliation(s)
- Cristiano Ramos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Vasileios Gerakopoulos
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria
| | - Rudolf Oehler
- Department of General Surgery, Division of Visceral Surgery, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
2
|
Higuchi Y, Ogata T, Nakanishi N, Nishi M, Tsuji Y, Tomita S, Conway SJ, Matoba S. Cavin-2 promotes fibroblast-to-myofibroblast trans-differentiation and aggravates cardiac fibrosis. ESC Heart Fail 2024; 11:167-178. [PMID: 37872863 PMCID: PMC10804157 DOI: 10.1002/ehf2.14571] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 09/22/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023] Open
Abstract
AIMS Transforming growth factor β (TGF-β) signalling is one of the critical pathways in fibroblast activation, and several drugs targeting the TGF-β/Smad signalling pathway in heart failure with cardiac fibrosis are being tested in clinical trials. Some caveolins and cavins, which are components of caveolae on the plasma membrane, are known for their association with the regulation of TGF-β signalling. Cavin-2 is particularly abundant in fibroblasts; however, the detailed association between Cavin-2 and cardiac fibrosis is still unclear. We tried to clarify the involvement and role of Cavin-2 in fibroblasts and cardiac fibrosis. METHODS AND RESULTS To clarify the role of Cavin-2 in cardiac fibrosis, we performed transverse aortic constriction (TAC) operations on four types of mice: wild-type (WT), Cavin-2 null (Cavin-2 KO), Cavin-2flox/flox , and activated fibroblast-specific Cavin-2 conditional knockout (Postn-Cre/Cavin-2flox/flox , Cavin-2 cKO) mice. We collected mouse embryonic fibroblasts (MEFs) from WT and Cavin-2 KO mice and investigated the effect of Cavin-2 in fibroblast trans-differentiation into myofibroblasts and associated TGF-β signalling. Four weeks after TAC, cardiac fibrotic areas in both the Cavin-2 KO and the Cavin-2 cKO mice were significantly decreased compared with each control group (WT 8.04 ± 1.58% vs. Cavin-2 KO 0.40 ± 0.03%, P < 0.01; Cavin-2flox/flox , 7.19 ± 0.50% vs. Cavin-2 cKO 0.88 ± 0.44%, P < 0.01). Fibrosis-associated mRNA expression (Col1a1, Ctgf, and Col3) was significantly attenuated in the Cavin-2 KO mice after TAC. α1 type I collagen deposition and non-vascular αSMA-positive cells (WT 43.5 ± 2.4% vs. Cavin-2 KO 25.4 ± 3.2%, P < 0.01) were reduced in the heart of the Cavin-2 cKO mice after TAC operation. The levels of αSMA protein (0.36-fold, P < 0.05) and fibrosis-associated mRNA expression (Col1a1, 0.69-fold, P < 0.01; Ctgf, 0.27-fold, P < 0.01; Col3, 0.60-fold, P < 0.01) were decreased in the Cavin-2 KO MEFs compared with the WT MEFs. On the other hand, αSMA protein levels were higher in the Cavin-2 overexpressed MEFs compared with the control MEFs (2.40-fold, P < 0.01). TGF-β1-induced Smad2 phosphorylation was attenuated in the Cavin-2 KO MEFs compared with WT MEFs (0.60-fold, P < 0.01). Heat shock protein 90 protein levels were significantly reduced in the Cavin-2 KO MEFs compared with the WT MEFs (0.69-fold, P < 0.01). CONCLUSIONS Cavin-2 loss suppressed fibroblast trans-differentiation into myofibroblasts through the TGF-β/Smad signalling. The loss of Cavin-2 in cardiac fibroblasts suppresses cardiac fibrosis and may maintain cardiac function.
Collapse
Affiliation(s)
- Yusuke Higuchi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Department of Pathology and Cell Regulation, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Naohiko Nakanishi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Masahiro Nishi
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Shinya Tomita
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| | - Simon J. Conway
- Herman B. Wells Center for Pediatric Research, Department of PediatricsIndiana University School of MedicineIndianapolisINUSA
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
| |
Collapse
|
3
|
Zhao T, Ding T, Sun Z, Shao X, Li S, Lu H, Yuan JH, Guo Z. SPHK1/S1P/S1PR pathway promotes the progression of peritoneal fibrosis by mesothelial-mesenchymal transition. FASEB J 2024; 38:e23417. [PMID: 38226856 DOI: 10.1096/fj.202301323r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 11/23/2023] [Accepted: 12/27/2023] [Indexed: 01/17/2024]
Abstract
Long-term exposure to non-physiologically compatible dialysate inevitably leads to peritoneal fibrosis (PF) in patients undergoing peritoneal dialysis (PD), and there is no effective prevention or treatment for PF. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid produced after catalysis by sphingosine kinase (SPHK) 1/2 and activates signals through the S1P receptor (S1PR) via autocrine or paracrine. However, the role of SPHK1/S1P/S1PR signaling has never been elucidated in PF. In our research, we investigated S1P levels in peritoneal effluents and demonstrated the role of SPHK1/S1P/S1PR pathway in peritoneal fibrosis. It was found that S1P levels in peritoneal effluents were positively correlated with D/P Cr (r = 0.724, p < .001) and negatively correlated with 4 h ultrafiltration volume (r = -0.457, p < .001). S1PR1 and S1PR3 on peritoneal cells were increased after high glucose exposure in vivo and in vitro. Fingolimod was applied to suppress S1P/S1PR pathway. Fingolimod restored mouse peritoneal function by reducing interstitial hyperplasia, maintaining ultrafiltration volume, reducing peritoneal transport solute rate, and mitigating the protein expression changes of fibronectin, vimentin, α-SMA, and E-cadherin induced by PD and S1P. Fingolimod preserved the morphology of the human peritoneal mesothelial cells, MeT-5A, and moderated the mesothelial-mesenchymal transition (MMT) process. We further delineated that SPHK1 was elevated in peritoneal cells after high glucose exposure and suppression of SPHK1 in MeT-5A cells reduced S1P release. Overexpression of SPHK1 in MeT-5A cells increased S1P levels in the supernatant and fostered the MMT process. PF-543 treatment, targeting SPHK1, alleviated deterioration of mouse peritoneal function. In conclusion, S1P levels in peritoneal effluent were correlated with the deterioration of peritoneal function. SPHK1/S1P/S1PR pathway played an important role in PF.
Collapse
Affiliation(s)
- Tingting Zhao
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
- Department of Nephrology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tao Ding
- Department of Endocrinology, Xizang Military General Hospital, Lhasa City, China
| | - Zhengyu Sun
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| | - Xin Shao
- Department of Cardiovascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuangxi Li
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| | - Hongtao Lu
- Department of Nutrition, Naval Medical University, Shanghai, China
| | - Ji-Hang Yuan
- Department of Medical Genetics, Naval Medical University, Shanghai, China
| | - Zhiyong Guo
- Department of Nephrology, First Affiliated Hospital of Naval Medical University, Shanghai Changhai Hospital, Shanghai, China
| |
Collapse
|
4
|
Li J, Liu Y, Liu J. A review of research progress on mechanisms of peritoneal fibrosis related to peritoneal dialysis. Front Physiol 2023; 14:1220450. [PMID: 37817984 PMCID: PMC10560738 DOI: 10.3389/fphys.2023.1220450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/13/2023] [Indexed: 10/12/2023] Open
Abstract
Peritoneal dialysis (PD) is an effective alternative treatment for patients with end-stage renal disease (ESRD) and is increasingly being adopted and promoted worldwide. However, as the duration of peritoneal dialysis extends, it can expose problems with dialysis inadequacy and ultrafiltration failure. The exact mechanism and aetiology of ultrafiltration failure have been of great concern, with triggers such as biological incompatibility of peritoneal dialysis solutions, uraemia toxins, and recurrent intraperitoneal inflammation initiating multiple pathways that regulate the release of various cytokines, promote the transcription of fibrosis-related genes, and deposit extracellular matrix. As a result, peritoneal fibrosis occurs. Exploring the pathogenic factors and molecular mechanisms can help us prevent peritoneal fibrosis and prolong the duration of Peritoneal dialysis.
Collapse
Affiliation(s)
- Jin’e Li
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yinghong Liu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Sotodosos-Alonso L, Pulgarín-Alfaro M, Del Pozo MA. Caveolae Mechanotransduction at the Interface between Cytoskeleton and Extracellular Matrix. Cells 2023; 12:cells12060942. [PMID: 36980283 PMCID: PMC10047380 DOI: 10.3390/cells12060942] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
The plasma membrane (PM) is subjected to multiple mechanical forces, and it must adapt and respond to them. PM invaginations named caveolae, with a specific protein and lipid composition, play a crucial role in this mechanosensing and mechanotransduction process. They respond to PM tension changes by flattening, contributing to the buffering of high-range increases in mechanical tension, while novel structures termed dolines, sharing Caveolin1 as the main component, gradually respond to low and medium forces. Caveolae are associated with different types of cytoskeletal filaments, which regulate membrane tension and also initiate multiple mechanotransduction pathways. Caveolar components sense the mechanical properties of the substrate and orchestrate responses that modify the extracellular matrix (ECM) according to these stimuli. They perform this function through both physical remodeling of ECM, where the actin cytoskeleton is a central player, and via the chemical alteration of the ECM composition by exosome deposition. Here, we review mechanotransduction regulation mediated by caveolae and caveolar components, focusing on how mechanical cues are transmitted through the cellular cytoskeleton and how caveolae respond and remodel the ECM.
Collapse
Affiliation(s)
- Laura Sotodosos-Alonso
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Marta Pulgarín-Alfaro
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
| |
Collapse
|
6
|
Anerillas C, Altés G, Gorospe M. MAPKs in the early steps of senescence implemEMTation. Front Cell Dev Biol 2023; 11:1083401. [PMID: 37009481 PMCID: PMC10060890 DOI: 10.3389/fcell.2023.1083401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
Evidence is accumulating that the earliest stages of the DNA damage response can direct cells toward senescence instead of other cell fates. In particular, tightly regulated signaling through Mitogen-Activated Protein Kinases (MAPKs) in early senescence can lead to a sustained pro-survival program and suppress a pro-apoptotic program. Importantly, an epithelial-to-mesenchymal Transition (EMT)-like program appears essential for preventing apoptosis and favoring senescence following DNA damage. In this review, we discuss how MAPKs might influence EMT features to promote a senescent phenotype that increases cell survival at the detriment of tissue function.
Collapse
|
7
|
Role of Ferroptosis in Regulating the Epithelial-Mesenchymal Transition in Pulmonary Fibrosis. Biomedicines 2023; 11:biomedicines11010163. [PMID: 36672671 PMCID: PMC9856078 DOI: 10.3390/biomedicines11010163] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Idiopathic pulmonary fibrosis is a chronic interstitial lung disease whose pathogenesis involves a complex interaction of cell types and signaling pathways. Lung epithelial cells responding to repeated injury experience persistent inflammation and sustained epithelial-mesenchymal transition (EMT). The persistence of EMT-induced signals generates extracellular matrix accumulation, thereby causing fibrosis. Ferroptosis is a newly characterized iron-dependent non-apoptotic regulated cell death. Increased iron accumulation can increase iron-induced oxidant damage in alveolar epithelial cells. Studies have demonstrated that iron steady states and oxidation steady states play an important role in the iron death regulation of EMT. This review summarizes the role of ferroptosis in regulating EMT in pulmonary fibrosis, aiming to provide a new idea for the prevention and treatment of this disease.
Collapse
|
8
|
De Belly H, Paluch EK, Chalut KJ. Interplay between mechanics and signalling in regulating cell fate. Nat Rev Mol Cell Biol 2022; 23:465-480. [PMID: 35365816 DOI: 10.1038/s41580-022-00472-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2022] [Indexed: 12/11/2022]
Abstract
Mechanical signalling affects multiple biological processes during development and in adult organisms, including cell fate transitions, cell migration, morphogenesis and immune responses. Here, we review recent insights into the mechanisms and functions of two main routes of mechanical signalling: outside-in mechanical signalling, such as mechanosensing of substrate properties or shear stresses; and mechanical signalling regulated by the physical properties of the cell surface itself. We discuss examples of how these two classes of mechanical signalling regulate stem cell function, as well as developmental processes in vivo. We also discuss how cell surface mechanics affects intracellular signalling and, in turn, how intracellular signalling controls cell surface mechanics, generating feedback into the regulation of mechanosensing. The cooperation between mechanosensing, intracellular signalling and cell surface mechanics has a profound impact on biological processes. We discuss here our understanding of how these three elements interact to regulate stem cell fate and development.
Collapse
Affiliation(s)
- Henry De Belly
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ewa K Paluch
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| | - Kevin J Chalut
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
- Wellcome/MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Wu L, Wang Z, He X, Jiang Y, Pan R, Chen S, Chen Y, Han Y, Yu H, Zhang T. GJA1 reverses arsenic-induced EMT via modulating MAPK/ERK signaling pathway. Toxicol Appl Pharmacol 2022; 450:116138. [PMID: 35750204 DOI: 10.1016/j.taap.2022.116138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 10/18/2022]
Abstract
Arsenic is known as a well-established human carcinogen. Gap Junction Protein Alpha 1 (GJA1) is a multifunction protein that forms gap junction channels and is important for intercellular communication. Recently, its aberrant expression has been shown to associate with cancer recurrence and metastatic spread. However, whether GJA1 plays a role in arsenic carcinogenesis remains unknown. Here, we demonstrated that chronic exposure of human bronchial epithelial BEAS-2B cells to sodium arsenite promoted epithelial-mesenchymal transition (EMT) via increasing the expression of EMT inducer S100A4 and activation of MAPK/ERK signaling. In vitro and in vivo experiments showed that chronic exposure to sodium arsenite reduced GJA1 expression. Forced expression of GJA1 inhibited sodium arsenite-induced EMT via suppressing MAPK/ERK signaling whereas GJA1 knockdown produced an opposite effect. Intriguingly, chronic exposure to sodium arsenite increased autophagy flux. Inhibition of autophagy by pharmacological intervention or genetic deletion of autophagy core gene Beclin-1 upregulated GJA1 expression. These results suggested that GJA1 restrained the carcinogenic effect of sodium arsenite by limiting MAPK/ERK signaling, and GJA1 expression was decreased by arsenic-activated autophagy. In addition, interventions directed at enhancing the level or functional activity of GJA1 could be of preventive and therapeutic interest.
Collapse
Affiliation(s)
- Linqing Wu
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Zengbin Wang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiaoli He
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | | | | | - Shi Chen
- Kindstar Global Medical Special Inspection Group, Wuhan, China
| | - Yizhong Chen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yanfei Han
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huahui Yu
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Tao Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
10
|
Yu M, Zhu W, Wang J, Chen X, He X, Lin B, Cen L, Zhou T, Lu C, Yu C, Sun J. Caveolin-1 Alleviates Crohn's Disease-induced Intestinal Fibrosis by Inhibiting Fibroblasts Autophagy Through Modulating Sequestosome 1. Inflamm Bowel Dis 2022; 28:923-935. [PMID: 35020883 DOI: 10.1093/ibd/izab342] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Intestinal fibrosis is a common complication of Crohn's disease (CD) and is characterized by the excessive accumulation of extracellular matrix produced by activated myofibroblasts. Caveolin-1 (CAV1) inhibits fibrosis. However, limited data show that CAV1 affects intestinal fibrosis. METHODS Human CD tissue samples were gained from patients with CD who underwent surgical resection of the intestine and were defined as stenotic or nonstenotic areas. A dextran sodium sulfate-induced mouse model of intestinal fibrosis was established. For in vitro experiments, we purchased CCD-18Co intestinal fibrosis cells and isolated and cultured human primary colonic fibroblasts. These fibroblasts were activated by transforming growth factor β administration for 48 hours. In the functional experiments, a specific small interfering RNA or overexpression plasmid was transfected into fibroblasts. The messenger RNA levels of fibrosis markers, such as α-smooth muscle actin, fibronectin, connective tissue growth factor, and collagen I1α, were determined using quantitative polymerase chain reaction. Western blot analysis was applied to detect the expression of CAV1, SQSTM1/p62 (sequestosome 1), and other fibrosis markers. RESULTS In human CD samples and the dextran sodium sulfate-induced mouse model of intestinal fibrosis, we observed a downregulation of CAV1 in fibrosis-activated areas. Mechanistically, CAV1 knockdown in both human primary colonic fibroblasts and CCD-18Co cells promoted fibroblast activation, while CAV1 overexpression inhibited fibroblast activation in vitro. We found that SQSTM1/p62 positively correlated with CAV1 expression levels in patients with CD and that it was indirectly modulated by CAV1 expression. Rescue experiments showed that CAV1 decreased primary human intestinal fibroblast activation by inhibiting fibroblast autophagy through the modulation of SQSTM1/p62. CONCLUSIONS Our data demonstrate that CAV1 deficiency induces fibroblast activation by indirectly regulating SQSTM1/p62 to promote fibroblast autophagy. CAV1 or SQSTM1/p62 may be potential therapeutic targets for intestinal fibrosis.
Collapse
Affiliation(s)
- Mengli Yu
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Zhu
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jinhai Wang
- Department of Colorectal Surgery, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xueyang Chen
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xinjue He
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Bingru Lin
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Li Cen
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Tianyu Zhou
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chao Lu
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Chaohui Yu
- Department of Gastroenterology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Sun
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Fibrosis of Peritoneal Membrane as Target of New Therapies in Peritoneal Dialysis. Int J Mol Sci 2022; 23:ijms23094831. [PMID: 35563220 PMCID: PMC9102299 DOI: 10.3390/ijms23094831] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.
Collapse
|
12
|
Yu R, Tian M, He P, Chen J, Zhao Z, Zhang Y, Zhang B. Suppression of LMCD1 ameliorates renal fibrosis by blocking the activation of ERK pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119200. [PMID: 34968577 DOI: 10.1016/j.bbamcr.2021.119200] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/10/2021] [Accepted: 12/18/2021] [Indexed: 12/29/2022]
Abstract
Tubulointerstitial fibrosis is a common pathway of chronic kidney disease (CKD) and is closely related to the progression of CKD. LMCD1, acting as an intermediary, has been reported to play a role in cardiac fibrosis. However, its role in renal fibrosis is yet to be deciphered. Based on the GEO database, we found the expression of LMCD1 is increased in kidney tissues of CKD patients and in human proximal tubular epithelial (HK-2) cells treated with transforming growth factor-β1 (TGF-β1), suggesting that LMCD1 may be involved in tubulointerstitial fibrosis. Herein, we investigated the role of LMCD1 in mice with unilateral ureteral obstruction (UUO) and in TGF-β1-stimulated HK-2 cells. In the UUO model, the expression of LMCD1 was upregulated. UUO-induced renal histopathological changes were mitigated by knockdown of LMCD1. LMCD1 silence alleviated renal interstitial fibrosis in UUO mice by decreasing the expression of TGF-β1, fibronectin, collagen I, and collagen III. LMCD1 deficiency suppressed cell apoptosis in kidney to prevent UUO-triggered renal injury. Furthermore, LMCD1 deficiency blocked the activation of ERK signaling in UUO mice. In vitro, LMCD1 was upregulated in HK-2 cells after TGF-β1 stimulation. LMCD1 silence abrogated TGF-β1-mediated upregulation of fibrotic genes. Treatment of HK-2 cells with ERK-specific inhibitor SCH772984 and agonist TPA validated LMCD1 exerted its function via activating ERK signaling. Together, our findings suggest that inhibition of LMCD1 protects against renal interstitial fibrosis by impeding ERK activation.
Collapse
Affiliation(s)
- Rui Yu
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Mi Tian
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Ping He
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Jie Chen
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Zixia Zhao
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Yongzhe Zhang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China
| | - Beiru Zhang
- Department of Nephrology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China.
| |
Collapse
|
13
|
Erichsen L, Thimm C, Bohndorf M, Rahman MS, Wruck W, Adjaye J. Activation of the Renin–Angiotensin System Disrupts the Cytoskeletal Architecture of Human Urine-Derived Podocytes. Cells 2022; 11:cells11071095. [PMID: 35406662 PMCID: PMC8997628 DOI: 10.3390/cells11071095] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/21/2022] [Indexed: 01/27/2023] Open
Abstract
High blood pressure is one of the major public health problems that causes severe disorders in several tissues including the human kidney. One of the most important signaling pathways associated with the regulation of blood pressure is the renin–angiotensin system (RAS), with its main mediator angiotensin II (ANGII). Elevated levels of circulating and intracellular ANGII and aldosterone lead to pro-fibrotic, -inflammatory, and -hypertrophic milieu that causes remodeling and dysfunction in cardiovascular and renal tissues. Furthermore, ANGII has been recognized as a major risk factor for the induction of apoptosis in podocytes, ultimately leading to chronic kidney disease (CKD). In the past, disease modeling of kidney-associated diseases was extremely difficult, as the derivation of kidney originated cells is very challenging. Here we describe a differentiation protocol for reproducible differentiation of sine oculis homeobox homolog 2 (SIX2)-positive urine-derived renal progenitor cells (UdRPCs) into podocytes bearing typical cellular processes. The UdRPCs-derived podocytes show the activation of the renin–angiotensin system by being responsive to ANGII stimulation. Our data reveal the ANGII-dependent downregulation of nephrin (NPHS1) and synaptopodin (SYNPO), resulting in the disruption of the podocyte cytoskeletal architecture, as shown by immunofluorescence-based detection of α-Actinin. Furthermore, we show that the cytoskeletal disruption is mainly mediated through angiotensin II receptor type 1 (AGTR1) signaling and can be rescued by AGTR1 inhibition with the selective, competitive angiotensin II receptor type 1 antagonist, losartan. In the present manuscript we confirm and propose UdRPCs differentiated to podocytes as a unique cell type useful for studying nephrogenesis and associated diseases. Furthermore, the responsiveness of UdRPCs-derived podocytes to ANGII implies potential applications in nephrotoxicity studies and drug screening.
Collapse
|
14
|
Zhou J, Wang G, Han R, Wang R, Kong Y, Zhang R, Hou L, Meng M. Glycopeptides from Paecilomyces sinensis ameliorate ethanol-induced gastric ulcers via anti-inflammation and the miR-9-5p-MEK/ERK signaling pathway. Food Funct 2021; 12:7664-7675. [PMID: 34236362 DOI: 10.1039/d1fo00911g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of this study was to investigate the immunomodulatory effect and mechanism of the glycopeptides from Paecilomyces sinensis (CPS-II) on ethanol induced ulcers in mice. In this study, histopathological evaluation (H&E staining) and the gastric ulcer score, ulcer index, total acid secretion and gastric pH value were used to determine the anti-ulcer activity. The expression levels of interleukin (IL)-6, interleukin (IL)-10 and tumor necrosis factor-α (TNF-α) were detected by ELISA. The contents of superoxide dismutase (SOD), malondialdehyde (MDA) and epidermal growth factor (PEG2) in serum were measured according to the instructions for the reagents. Western blotting was used to detect the effect of CPS-II on the MEK/ERK pathway. The results showed that CPS-II could inhibit the ulcer score and ulcer index compared with the disease control group. CPS-II could significantly increase gastric pH and decrease gastric acid secretion in mice. The ELISA analysis showed that the expression levels of IL-6 and TNF-α in the CPS-II treatment group were significantly decreased, while the expression levels of IL-10 were significantly increased in the CPS-II treatment group. In the resveratrol treatment group, the content of MDA in serum was decreased, and the level of PEG2 and the activity of SOD in serum were significantly increased, which indicated that CPS-II has immunoregulation and anti-ulcer properties. The CPS-II treatment group could reduce the expression level of miR-9-5p in gastric tissue. pEGFR had been identified as a potential target of miR-9-5p. Western blot analysis showed that CPS-II could up-regulate the relative protein expression of pEGFR/EGFR, pRaf/Raf, pMEK/MEK, pERK/ERK, and ZO-1. The results showed that CPS-II could reduce oxidative stress and inflammatory response by regulating the miR-9-5p-MEK/ERK signaling pathway, thus protecting the gastric mucosa and improving stress gastric ulcers.
Collapse
Affiliation(s)
- Jiaping Zhou
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, College of Food Engineering and Biotechnology, Tianjin University of Science and Technology, No. 29, 13th Avenue, Tianjin Economy Technological Development Area, Tianjin 300457, People's Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Terri M, Trionfetti F, Montaldo C, Cordani M, Tripodi M, Lopez-Cabrera M, Strippoli R. Mechanisms of Peritoneal Fibrosis: Focus on Immune Cells-Peritoneal Stroma Interactions. Front Immunol 2021; 12:607204. [PMID: 33854496 PMCID: PMC8039516 DOI: 10.3389/fimmu.2021.607204] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
Peritoneal fibrosis is characterized by abnormal production of extracellular matrix proteins leading to progressive thickening of the submesothelial compact zone of the peritoneal membrane. This process may be caused by a number of insults including pathological conditions linked to clinical practice, such as peritoneal dialysis, abdominal surgery, hemoperitoneum, and infectious peritonitis. All these events may cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy. Among the cellular processes implicated in these peritoneal alterations is the generation of myofibroblasts from mesothelial cells and other cellular sources that are central in the induction of fibrosis and in the subsequent functional deterioration of the peritoneal membrane. Myofibroblast generation and activity is actually integrated in a complex network of extracellular signals generated by the various cellular types, including leukocytes, stably residing or recirculating along the peritoneal membrane. Here, the main extracellular factors and the cellular players are described with emphasis on the cross-talk between immune system and cells of the peritoneal stroma. The understanding of cellular and molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.
Collapse
Affiliation(s)
- Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Flavia Trionfetti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Claudia Montaldo
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Marco Cordani
- instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA) Nanociencia, Madrid, Spain
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
- Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Manuel Lopez-Cabrera
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular “Severo Ochoa”-Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
16
|
Chen H, Qian Z, Zhang S, Tang J, Fang L, Jiang F, Ge D, Chang J, Cao J, Yang L, Cao X. Silencing COX-2 blocks PDK1/TRAF4-induced AKT activation to inhibit fibrogenesis during skeletal muscle atrophy. Redox Biol 2021; 38:101774. [PMID: 33152664 PMCID: PMC7645269 DOI: 10.1016/j.redox.2020.101774] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 02/08/2023] Open
Abstract
Skeletal muscle atrophy with high prevalence can induce weakness and fatigability and place huge burden on both health and quality of life. During skeletal muscle degeneration, excessive fibroblasts and extracellular matrix (ECM) accumulated to replace and impair the resident muscle fiber and led to loss of muscle mass. Cyclooxygenase-2 (COX-2), the rate-limiting enzyme in synthesis of prostaglandin, has been identified as a positive regulator in pathophysiological process like inflammation and oxidative stress. In our study, we found injured muscles of human subjects and mouse model overexpressed COX-2 compared to the non-damaged region and COX-2 was also upregulated in fibroblasts following TGF-β stimulation. Then we detected the effect of selective COX-2 inhibitor celecoxib on fibrogenesis. Celecoxib mediated anti-fibrotic effect by inhibiting fibroblast differentiation, proliferation and migration as well as inactivating TGF-β-dependent signaling pathway, non-canonical TGF-β pathways and suppressing generation of reactive oxygen species (ROS) and oxidative stress. In vivo pharmacological inhibition of COX-2 by celecoxib decreased tissue fibrosis and increased skeletal muscle fiber preservation reflected by less ECM formation and myofibroblast accumulation with decreased p-ERK1/2, p-Smad2/3, TGF-βR1, VEGF, NOX2 and NOX4 expression. Expression profiling further found that celecoxib could suppress PDK1 expression. The interaction between COX-2 and PDK1/AKT signaling remained unclear, here we found that COX-2 could bind to PDK1/AKT to form compound. Knockdown of COX-2 in fibroblasts by pharmacological inactivation or by siRNA restrained PDK1 expression and AKT phosphorylation induced by TGF-β treatment. Besides, si-COX-2 prevented TGF-β-induced K63-ubiquitination of AKT by blocking the interaction between AKT and E3 ubiquitin ligase TRAF4. In summary, we found blocking COX-2 inhibited fibrogenesis after muscle atrophy induced by injury and suppressed AKT signaling pathway by inhibiting upstream PDK1 expression and preventing the recruitment of TRAF4 to AKT, indicating that COX-2/PDK1/AKT signaling pathway promised to be target for treating muscle atrophy in the future.
Collapse
Affiliation(s)
- Hongtao Chen
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhanyang Qian
- Department of Orthopedics, Zhongda Hospital of Southeast University, Nanjing, Jiangsu, China
| | - Sheng Zhang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Tang
- Department of Plastic and Burn Surgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Le Fang
- Department of Critical Care Medicine, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Jiang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dawei Ge
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jie Chang
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lei Yang
- Department of Orthopedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiaojian Cao
- Department of Orthopedics, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
17
|
IL-17A as a Potential Therapeutic Target for Patients on Peritoneal Dialysis. Biomolecules 2020; 10:biom10101361. [PMID: 32987705 PMCID: PMC7598617 DOI: 10.3390/biom10101361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) is a health problem reaching epidemic proportions. There is no cure for CKD, and patients may progress to end-stage renal disease (ESRD). Peritoneal dialysis (PD) is a current replacement therapy option for ESRD patients until renal transplantation can be achieved. One important problem in long-term PD patients is peritoneal membrane failure. The mechanisms involved in peritoneal damage include activation of the inflammatory and immune responses, associated with submesothelial immune infiltrates, angiogenesis, loss of the mesothelial layer due to cell death and mesothelial to mesenchymal transition, and collagen accumulation in the submesothelial compact zone. These processes lead to fibrosis and loss of peritoneal membrane function. Peritoneal inflammation and membrane failure are strongly associated with additional problems in PD patients, mainly with a very high risk of cardiovascular disease. Among the inflammatory mediators involved in peritoneal damage, cytokine IL-17A has recently been proposed as a potential therapeutic target for chronic inflammatory diseases, including CKD. Although IL-17A is the hallmark cytokine of Th17 immune cells, many other cells can also produce or secrete IL-17A. In the peritoneum of PD patients, IL-17A-secreting cells comprise Th17 cells, γδ T cells, mast cells, and neutrophils. Experimental studies demonstrated that IL-17A blockade ameliorated peritoneal damage caused by exposure to PD fluids. This article provides a comprehensive review of recent advances on the role of IL-17A in peritoneal membrane injury during PD and other PD-associated complications.
Collapse
|
18
|
Cellular and molecular events of inflammation induced transdifferentiation (EMT) and regeneration (MET) in mesenteric mesothelial cells. Inflamm Res 2020; 69:1173-1179. [PMID: 32920669 PMCID: PMC7486969 DOI: 10.1007/s00011-020-01400-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
In this review we summarize the cellular and molecular events of inflammation induced epithelial-to-mesenchymal (EMT) and mesothelial-to-macrophage transition (MET) during regeneration. Since the receptor transmits the environmental stimulus, downregulating or upregulating the process on an epigenetic level, the intracellular localization of receptors (signaling organelles: early endosomes or lysosomal degradation: late endosomes) plays a crucial role in the signaling events regulating inflammation and regeneration. Therefore, we focused on the internalization of the receptors as well as the intracellular compartmentalization of signaling molecules during EMT and MET. The review draws the reader's attention to the plasticity of mesothelial cells and supports the idea that during inflammation an ambient macrophage population might derive from mesothelial cells.
Collapse
|
19
|
Zhou J, Liu S, Guo L, Wang R, Chen J, Shen J. NMDA receptor-mediated CaMKII/ERK activation contributes to renal fibrosis. BMC Nephrol 2020; 21:392. [PMID: 32907546 PMCID: PMC7488001 DOI: 10.1186/s12882-020-02050-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 08/30/2020] [Indexed: 02/08/2023] Open
Abstract
Background This study aimed to understand the mechanistic role of N-methyl-D-aspartate receptor (NMDAR) in acute fibrogenesis using models of in vivo ureter obstruction and in vitro TGF-β administration. Methods Acute renal fibrosis (RF) was induced in mice by unilateral ureteral obstruction (UUO). Histological changes were observed using Masson’s trichrome staining. The expression levels of NR1, which is the functional subunit of NMDAR, and fibrotic and epithelial-to-mesenchymal transition markers were measured by immunohistochemical and Western blot analysis. HK-2 cells were incubated with TGF-β, and NMDAR antagonist MK-801 and Ca2+/calmodulin-dependent protein kinase II (CaMKII) antagonist KN-93 were administered for pathway determination. Chronic RF was introduced by sublethal ischemia–reperfusion injury in mice, and NMDAR inhibitor dextromethorphan hydrobromide (DXM) was administered orally. Results The expression of NR1 was upregulated in obstructed kidneys, while NR1 knockdown significantly reduced both interstitial volume expansion and the changes in the expression of α-smooth muscle actin, S100A4, fibronectin, COL1A1, Snail, and E-cadherin in acute RF. TGF-β1 treatment increased the elongation phenotype of HK-2 cells and the expression of membrane-located NR1 and phosphorylated CaMKII and extracellular signal–regulated kinase (ERK). MK801 and KN93 reduced CaMKII and ERK phosphorylation levels, while MK801, but not KN93, reduced the membrane NR1 signal. The levels of phosphorylated CaMKII and ERK also increased in kidneys with obstruction but were decreased by NR1 knockdown. The 4-week administration of DXM preserved renal cortex volume in kidneys with moderate ischemic–reperfusion injury. Conclusions NMDAR participates in both acute and chronic renal fibrogenesis potentially via CaMKII-induced ERK activation.
Collapse
Affiliation(s)
- Jingyi Zhou
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Shuaihui Liu
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Luying Guo
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Rending Wang
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China.,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China.,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China.,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China
| | - Jianghua Chen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China. .,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China. .,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China. .,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China.
| | - Jia Shen
- Kidney Disease Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China. .,Key Laboratory of Nephropathy, Hangzhou, Zhejiang Province, China. .,Kidney Disease Immunology Laboratory, the Third-Grade Laboratory, State Administration of Traditional Chinese Medicine of China, Hangzhou, China. .,Key Laboratory of Multiple Organ Transplantation, Ministry of Health of China, Hangzhou, China.
| |
Collapse
|
20
|
Strippoli R, Sandoval P, Moreno-Vicente R, Rossi L, Battistelli C, Terri M, Pascual-Antón L, Loureiro M, Matteini F, Calvo E, Jiménez-Heffernan JA, Gómez MJ, Jiménez-Jiménez V, Sánchez-Cabo F, Vázquez J, Tripodi M, López-Cabrera M, Del Pozo MÁ. Caveolin1 and YAP drive mechanically induced mesothelial to mesenchymal transition and fibrosis. Cell Death Dis 2020; 11:647. [PMID: 32811813 PMCID: PMC7435273 DOI: 10.1038/s41419-020-02822-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/27/2022]
Abstract
Despite their emerging relevance to fully understand disease pathogenesis, we have as yet a poor understanding as to how biomechanical signals are integrated with specific biochemical pathways to determine cell behaviour. Mesothelial-to-mesenchymal transition (MMT) markers colocalized with TGF-β1-dependent signaling and yes-associated protein (YAP) activation across biopsies from different pathologies exhibiting peritoneal fibrosis, supporting mechanotransduction as a central driving component of these class of fibrotic lesions and its crosstalk with specific signaling pathways. Transcriptome and proteome profiling of the response of mesothelial cells (MCs) to linear cyclic stretch revealed molecular changes compatible with bona fide MMT, which (i) overlapped with established YAP target gene subsets, and were largely dependent on endogenous TGF-β1 signaling. Importantly, TGF-β1 blockade blunts the transcriptional upregulation of these gene signatures, but not the mechanical activation and nuclear translocation of YAP per se. We studied the role therein of caveolin-1 (CAV1), a plasma membrane mechanotransducer. Exposure of CAV1-deficient MCs to cyclic stretch led to a robust upregulation of MMT-related gene programs, which was blunted upon TGF-β1 inhibition. Conversely, CAV1 depletion enhanced both TGF-β1 and TGFBRI expression, whereas its re-expression blunted mechanical stretching-induced MMT. CAV1 genetic deficiency exacerbated MMT and adhesion formation in an experimental murine model of peritoneal ischaemic buttons. Taken together, these results support that CAV1-YAP/TAZ fine-tune the fibrotic response through the modulation of MMT, onto which TGF-β1-dependent signaling coordinately converges. Our findings reveal a cooperation between biomechanical and biochemical signals in the triggering of MMT, representing a novel potential opportunity to intervene mechanically induced disorders coursing with peritoneal fibrosis, such as post-surgical adhesions.
Collapse
Affiliation(s)
- Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy. .,National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy. .,Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain.
| | - Pilar Sandoval
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular "Severo Ochoa"-CSIC, 28049, Madrid, Spain
| | - Roberto Moreno-Vicente
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Lucia Rossi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Cecilia Battistelli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Michela Terri
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.,National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy
| | - Lucía Pascual-Antón
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular "Severo Ochoa"-CSIC, 28049, Madrid, Spain
| | - Marta Loureiro
- Cardiovascular Proteomics laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) and CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Francesca Matteini
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy
| | - Enrique Calvo
- Cardiovascular Proteomics laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) and CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - José Antonio Jiménez-Heffernan
- Departamento de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), 28006, Madrid, Spain
| | - Manuel José Gómez
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029, Madrid, Spain
| | - Victor Jiménez-Jiménez
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029, Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) and CIBER Cardiovascular Diseases (CIBERCV), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Marco Tripodi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161, Rome, Italy.,National Institute for Infectious Diseases L. Spallanzani, IRCCS, Via Portuense, 292, 00149, Rome, Italy
| | - Manuel López-Cabrera
- Programa de Homeostasis de Tejidos y Organos, Centro de Biología Molecular "Severo Ochoa"-CSIC, 28049, Madrid, Spain.
| | - Miguel Ángel Del Pozo
- Mechanoadaptation & Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029, Madrid, Spain.
| |
Collapse
|
21
|
Cell polarity and oncogenesis: common mutations contribute to altered cellular polarity and promote malignancy. THE NUCLEUS 2020. [DOI: 10.1007/s13237-020-00313-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
22
|
Membrane dynamics in cell migration. Essays Biochem 2020; 63:469-482. [PMID: 31350382 DOI: 10.1042/ebc20190014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 06/27/2019] [Accepted: 07/11/2019] [Indexed: 12/20/2022]
Abstract
Migration of cells is required in multiple tissue-level processes, such as in inflammation or cancer metastasis. Endocytosis is an extremely regulated cellular process by which cells uptake extracellular molecules or internalise cell surface receptors. While the role of endocytosis of focal adhesions (FA) and plasma membrane (PM) turnover at the leading edge of migratory cells is wide known, the contribution of endocytic proteins per se in migration has been frequently disregarded. In this review, we describe the novel functions of the most well-known endocytic proteins in cancer cell migration, focusing on clathrin, caveolin, flotillins and GRAF1. In addition, we highlight the relevance of the macropinocytic pathway in amoeboid-like cell migration.
Collapse
|
23
|
Wei X, Zhu X, Jiang L, Huang X, Zhang Y, Zhao D, Du Y. Recent advances in understanding the role of hypoxia-inducible factor 1α in renal fibrosis. Int Urol Nephrol 2020; 52:1287-1295. [PMID: 32378138 DOI: 10.1007/s11255-020-02474-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 04/15/2020] [Indexed: 12/11/2022]
Abstract
Renal fibrosis is the most common pathological manifestation of chronic kidney disease (CKD), and with numerous influencing factors, its pathogenesis is complex. Epithelial-mesenchymal transition (EMT) is known to promote the progression of renal fibrosis via alterations in the secreted proteome. Moreover, blocking or even reversing EMT can effectively reduce the degree of fibrosis. As such, targeting the key molecules responsible for promoting EMT may be an effective strategy for inhibiting renal fibrosis. Research in recent years has demonstrated that hypoxia-inducible factor 1α (HIF-1α) acts to promote renal fibrosis through regulation of EMT. However, the relationship between HIF-1α and EMT remains incompletely understood. In the present review, the underlying mechanism of the interaction between HIF-1α and EMT is explored to provide novel insight into the pathogenesis of renal fibrosis and new ideas for early targeted intervention.
Collapse
Affiliation(s)
- Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Lili Jiang
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Xiu Huang
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Yangyang Zhang
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Dan Zhao
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, 71 XinMin Street, Changchun, Jilin, China.
| |
Collapse
|
24
|
Su W, Wang H, Feng Z, Sun J. Nitro-oleic acid inhibits the high glucose-induced epithelial-mesenchymal transition in peritoneal mesothelial cells and attenuates peritoneal fibrosis. Am J Physiol Renal Physiol 2019; 318:F457-F467. [PMID: 31760768 DOI: 10.1152/ajprenal.00425.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As an electrophilic nitroalkene fatty acid, nitro-oleic acid (OA-NO2) exerts multiple biological effects that contribute to anti-inflammation, anti-oxidative stress, and antiapoptosis. However, little is known about the role of OA-NO2 in peritoneal fibrosis. Thus, in the present study, we examined the effects of OA-NO2 on the high glucose (HG)-induced epithelial-mesenchymal transition (EMT) in human peritoneal mesothelial cells (HPMCs) and evaluated the morphological and immunohistochemical changes in a rat model of peritoneal dialysis-related peritoneal fibrosis. In in vitro experiments, we found that HG reduced the expression level of E-cadherin and increased Snail, N-cadherin, and α-smooth muscle actin expression levels in HPMCs. The above-mentioned changes were attenuated by pretreatment with OA-NO2. Additionally, OA-NO2 also inhibited HG-induced activation of the transforming growth factor-β1/Smad signaling pathway and NF-κB signaling pathway. Meanwhile, OA-NO2 inhibited HG-induced phosphorylation of Erk and JNK. The results from the in vivo experiments showed that OA-NO2 notably relieved peritoneal fibrosis by decreasing the thickness of the peritoneum; it also inhibited expression of transforming growth factor-β1, α-smooth muscle actin, N-cadherin, and vimentin and enhanced expression of E-cadherin in the peritoneum. Collectively, these results suggest that OA-NO2 inhibits the HG-induced epithelial-mesenchymal transition in HPMCs and attenuates peritoneal dialysis-related peritoneal fibrosis.
Collapse
Affiliation(s)
- Wenyan Su
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| | - Haiping Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| | - ZiYan Feng
- Department of Dialysis, JuanCheng People's Hospital, Heze, Shangdong, China
| | - Jing Sun
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shangdong, China
| |
Collapse
|
25
|
Miyagawa K, Shi M, Chen PI, Hennigs JK, Zhao Z, Wang M, Li CG, Saito T, Taylor S, Sa S, Cao A, Wang L, Snyder MP, Rabinovitch M. Smooth Muscle Contact Drives Endothelial Regeneration by BMPR2-Notch1-Mediated Metabolic and Epigenetic Changes. Circ Res 2019; 124:211-224. [PMID: 30582451 DOI: 10.1161/circresaha.118.313374] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
RATIONALE Maintaining endothelial cells (EC) as a monolayer in the vessel wall depends on their metabolic state and gene expression profile, features influenced by contact with neighboring cells such as pericytes and smooth muscle cells (SMC). Failure to regenerate a normal EC monolayer in response to injury can result in occlusive neointima formation in diseases such as atherosclerosis and pulmonary arterial hypertension. OBJECTIVE We investigated the nature and functional importance of contact-dependent communication between SMC and EC to maintain EC integrity. METHODS AND RESULTS We found that in SMC and EC contact cocultures, BMPR2 (bone morphogenetic protein receptor 2) is required by both cell types to produce collagen IV to activate ILK (integrin-linked kinase). This enzyme directs p-JNK (phospho-c-Jun N-terminal kinase) to the EC membrane, where it stabilizes presenilin1 and releases N1ICD (Notch1 intracellular domain) to promote EC proliferation. This response is necessary for EC regeneration after carotid artery injury. It is deficient in EC-SMC Bmpr2 double heterozygous mice in association with reduced collagen IV production, decreased N1ICD, and attenuated EC proliferation, but can be rescued by targeting N1ICD to EC. Deletion of EC- Notch1 in transgenic mice worsens hypoxia-induced pulmonary hypertension, in association with impaired EC regenerative function associated with loss of precapillary arteries. We further determined that N1ICD maintains EC proliferative capacity by increasing mitochondrial mass and by inducing the phosphofructokinase PFKFB3 (fructose-2,6-bisphosphatase 3). Chromatin immunoprecipitation sequencing analyses showed that PFKFB3 is required for citrate-dependent H3K27 acetylation at enhancer sites of genes regulated by the acetyl transferase p300 and by N1ICD or the N1ICD target MYC and necessary for EC proliferation and homeostasis. CONCLUSIONS Thus, SMC-EC contact is required for activation of Notch1 by BMPR2, to coordinate metabolism with chromatin remodeling of genes that enable EC regeneration, and to maintain monolayer integrity and vascular homeostasis in response to injury.
Collapse
Affiliation(s)
- Kazuya Miyagawa
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Minyi Shi
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Pin-I Chen
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Jan K Hennigs
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Zhixin Zhao
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Mouer Wang
- Department of Medicine (M.W.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Caiyun G Li
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Toshie Saito
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Shalina Taylor
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Silin Sa
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Aiqin Cao
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Lingli Wang
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| | - Michael P Snyder
- Department of Genetics (M.S., Z.Z., M.P.S.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA
| | - Marlene Rabinovitch
- From the Department of Pediatrics (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA.,Cardiovascular Institute (K.M., M.S., P.-I.C., J.K.H., Z.Z., M.W., C.G.L., T.S., S.T., S.S., A.C., L.W., M.P.S., M.R.), Stanford University School of Medicine, CA.,Vera Moulton Wall Center for Pulmonary Vascular Disease (K.M., P.-I.C., J.K.H., C.G.L., T.S., S.T., S.S., A.C., L.W., M.R.), Stanford University School of Medicine, CA
| |
Collapse
|
26
|
Wu J, Huang Q, Li P, Wang Y, Zheng C, Lei X, Li S, Gong W, Yin B, Luo C, Xiao J, Zhou W, Xu Z, Chen Y, Peng F, Long H. MicroRNA-145 promotes the epithelial-mesenchymal transition in peritoneal dialysis-associated fibrosis by suppressing fibroblast growth factor 10. J Biol Chem 2019; 294:15052-15067. [PMID: 31431501 PMCID: PMC6791318 DOI: 10.1074/jbc.ra119.007404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 08/14/2019] [Indexed: 12/11/2022] Open
Abstract
Peritoneal fibrosis is a common complication of long-term peritoneal dialysis (PD) and the principal cause of ultrafiltration failure during PD. The initial and reversible step in PD-associated peritoneal fibrosis is the epithelial-mesenchymal transition (EMT). Although the mechanisms in the EMT have been the focus of many studies, only limited information is currently available concerning microRNA (miRNA) regulation in peritoneal fibrosis. In this study, we aimed to characterize the roles of microRNA-145 (miR-145) and fibroblast growth factor 10 (FGF10) in peritoneal fibrosis. After inducing EMT with transforming growth factor-β1 (TGF-β1) in vitro, we found that miR-145 is significantly up-regulated, whereas FGF10 is markedly down-regulated, suggesting a close link between miR-145 and FGF10 in peritoneal fibrosis, further confirmed in luciferase reporter experiments. Furthermore, in human peritoneal mesothelial cells (i.e. HMrSV5 cells), miR-145 mimics induced EMT, whereas miR-145 inhibition suppressed EMT, and we also observed that miR-145 suppressed FGF10 expression. In vivo, we found that the exogenous delivery of an miR-145 expression plasmid both blocked FGF10 and intensified the EMT, whereas miR-145 inhibition promoted the expression of FGF10 and reversed the EMT. In conclusion, miR-145 promotes the EMT during the development of peritoneal fibrosis by suppressing FGF10 activity, suggesting that miR-145 represents a potential therapeutic target for managing peritoneal fibrosis.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qianyin Huang
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Peilin Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxian Wang
- Department of Gerontology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chenghao Zheng
- Second Clinical Medical School, Southern Medical University, Guangzhou 510280, China
| | - Xianghong Lei
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuting Li
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Wangqiu Gong
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Bohui Yin
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Congwei Luo
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jing Xiao
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weidong Zhou
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhaozhong Xu
- Department of Emergency, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yihua Chen
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Fenfen Peng
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Haibo Long
- Department of Nephrology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| |
Collapse
|
27
|
Sobierajska K, Wawro ME, Ciszewski WM, Niewiarowska J. Transforming Growth Factor-β Receptor Internalization via Caveolae Is Regulated by Tubulin-β2 and Tubulin-β3 during Endothelial-Mesenchymal Transition. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2531-2546. [PMID: 31539520 DOI: 10.1016/j.ajpath.2019.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/28/2019] [Accepted: 08/26/2019] [Indexed: 02/02/2023]
Abstract
Fibrotic disorders, which are caused by long-term inflammation, are observed in numerous organs. These disorders are regulated mainly through transforming growth factor (TGF)-β family proteins by a fundamental cellular mechanism, known as the endothelial-mesenchymal transition. Therefore, there is a pressing need to identify the mechanisms and potential therapeutic targets that enable the inhibition of endothelial transdifferentiation. This study is the first to demonstrate that glycosylation of tubulin-β2 and tubulin-β3 in microtubules enhances sensitivity to TGF-β1 stimulation in human microvascular endothelial cells. We observed that the microtubules enriched in glycosylated tubulin-β2 and tubulin-β3 were necessary for caveolae-dependent TGF-β receptor internalization. Post-translational modulation is critical for the generation of myofibroblasts through endothelial-mesenchymal transition during fibrosis development. We suggest that microtubule glycosylation may become the target of new effective therapies for patients with recognized fibrotic diseases.
Collapse
Affiliation(s)
| | - Marta E Wawro
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland
| | - Wojciech M Ciszewski
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland
| | - Jolanta Niewiarowska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
28
|
Nagaraja MR, Tiwari N, Shetty SK, Marudamuthu AS, Fan L, Ostrom RS, Fu J, Gopu V, Radhakrishnan V, Idell S, Shetty S. p53 Expression in Lung Fibroblasts Is Linked to Mitigation of Fibrotic Lung Remodeling. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 188:2207-2222. [PMID: 30253845 DOI: 10.1016/j.ajpath.2018.07.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 06/11/2018] [Accepted: 07/02/2018] [Indexed: 02/06/2023]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a debilitating, incurable, and life-threatening disease. A cardinal feature of the pathogenesis of IPF is excessive extracellular matrix deposition attributable to proliferation of activated fibrotic lung fibroblasts (fLfs). To assess the underlying mechanism, we analyzed the status of the tumor suppressor protein p53 in fLfs from the lungs of IPF patients or mice with bleomycin-induced established PF. We report that basal expression of p53 is markedly reduced in fLfs. Forced expression of caveolin-1 in fLfs increased basal p53 and reduced profibrogenic proteins, including collagen-1. Transduction of fLfs with adenovirus expressing p53 reduced expression of these proteins. Conversely, inhibition of baseline p53 in control lung fibroblasts from lung tissues increased profibrogenic protein expression. Lung transduction of adenovirus expressing p53 reduced bleomycin-induced PF in wild-type or caveolin-1-deficient mice. Furthermore, treatment of fLfs or fibrotic lung tissues with caveolin-1 scaffolding domain peptide (CSP) or its fragment, CSP7, restored p53 and reduced profibrogenic proteins. Treatment of wild-type mice with i.p. CSP or CSP7 resolved bleomycin-induced PF. These peptides failed to resolve PF in inducible conditional knockout mice lacking p53 in fLfs, indicating the induction of baseline fLf p53 as the basis of the antifibrotic effects.
Collapse
Affiliation(s)
- M R Nagaraja
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Nivedita Tiwari
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Shwetha K Shetty
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Amarnath S Marudamuthu
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Liang Fan
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Rennolds S Ostrom
- Department of Pharmacology, Chapman University School of Pharmacy, Irvine, California
| | - Jian Fu
- Center for Research on Environmental Disease and Toxicology, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Venkadesaperumal Gopu
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Vijay Radhakrishnan
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Steven Idell
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas
| | - Sreerama Shetty
- Texas Lung Injury Institute, Department of Medicine, University of Texas Health Science Center at Tyler, Tyler, Texas.
| |
Collapse
|
29
|
Yu H, Sun J, Zhao C, Wang H, Liu Y, Xiong J, Chang J, Wang M, Wang W, Ye D, Zhou H, Yu T. SET domain containing protein 5 (SETD5) enhances tumor cell invasion and is associated with a poor prognosis in non-small cell lung cancer patients. BMC Cancer 2019; 19:736. [PMID: 31345185 PMCID: PMC6659235 DOI: 10.1186/s12885-019-5944-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 07/16/2019] [Indexed: 12/25/2022] Open
Abstract
Background SET domain containing 5 (SETD5) is related to the aggressiveness of prostate and mammary cancers, but its association with non-small cell lung cancer (NSCLC) is unknown. Therefore, the purpose of this research was to determine the expression pattern and function of SETD5 in NSCLC. Methods SETD5 was detected by immunohistochemical analysis in 147 patients with non-small cell lung cancer. SETD5 was overexpressed in A549 cells or suppressed with siRNA in H1299 cells. Wound healing and transwell assays were performed. The expression levels of SETD5, p-AKT/AKT, Snail, p-JNK/JNK, Slug, E-cadherin, Zo-1, p-P38/P38, occludin, α-catenin, p-ERK/ERK, and p-P90RSK/ P90RSK were assessed by western blot. Results Online analysis of overall survival in 1928 patients with NSCLC showed that the SETD5 gene was related to worse overall survival (OS)(P < 0.001). The positive expression rate of SETD5 in noncancerous tissues was lower than that in cancerous tissues (16.7% vs. 44.2%, P < 0.001). SETD5 was significantly correlated with advanced TNM stage (P < 0.001), lymph node metastasis (P < 0.001) and overall survival rate (P < 0.001). Overexpression of SETD5 in A549 cells increased migration and invasion, while deletion of SETD5 in H1299 cells decreased migration and invasion. After overexpression of SETD5, the expression of ZO-1 was downregulated, and that of Snail was upregulated. After overexpression of SETD5, the levels of p-ERK and its downstream factor p-p90rsk increased. Conclusion These results suggest that SETD5 could regulate p-P90RSK and facilitate the migration and invasion of NSCLC and may be related to the poor prognosis of patients with NSCLC.
Collapse
Affiliation(s)
- Hairu Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Jiayi Sun
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Congxuan Zhao
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Haotian Wang
- The First Clinical College, Dalian Medical University, No. 9 West Section of Lushun South Road, Dalian City, Liaoning Province, China
| | - Yeqiu Liu
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Jiajia Xiong
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Jing Chang
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Mixue Wang
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Wenhui Wang
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Dongman Ye
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Hongyan Zhou
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China. .,Department of Medical Imaging, Liaoning Cancer Hospital and Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning Province, China.
| |
Collapse
|
30
|
Olea-Flores M, Zuñiga-Eulogio MD, Mendoza-Catalán MA, Rodríguez-Ruiz HA, Castañeda-Saucedo E, Ortuño-Pineda C, Padilla-Benavides T, Navarro-Tito N. Extracellular-Signal Regulated Kinase: A Central Molecule Driving Epithelial-Mesenchymal Transition in Cancer. Int J Mol Sci 2019; 20:E2885. [PMID: 31200510 PMCID: PMC6627365 DOI: 10.3390/ijms20122885] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/09/2019] [Accepted: 06/11/2019] [Indexed: 12/18/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a reversible cellular process, characterized by changes in gene expression and activation of proteins, favoring the trans-differentiation of the epithelial phenotype to a mesenchymal phenotype. This process increases cell migration and invasion of tumor cells, progression of the cell cycle, and resistance to apoptosis and chemotherapy, all of which support tumor progression. One of the signaling pathways involved in tumor progression is the MAPK pathway. Within this family, the ERK subfamily of proteins is known for its contributions to EMT. The ERK subfamily is divided into typical (ERK 1/2/5), and atypical (ERK 3/4/7/8) members. These kinases are overexpressed and hyperactive in various types of cancer. They regulate diverse cellular processes such as proliferation, migration, metastasis, resistance to chemotherapy, and EMT. In this context, in vitro and in vivo assays, as well as studies in human patients, have shown that ERK favors the expression, function, and subcellular relocalization of various proteins that regulate EMT, thus promoting tumor progression. In this review, we discuss the mechanistic roles of the ERK subfamily members in EMT and tumor progression in diverse biological systems.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Miriam Daniela Zuñiga-Eulogio
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Miguel Angel Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Hugo Alberto Rodríguez-Ruiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Carlos Ortuño-Pineda
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n Chilpancingo, Gro. 39090, Mexico.
| |
Collapse
|
31
|
Phosphorylation Regulates CAP1 (Cyclase-Associated Protein 1) Functions in the Motility and Invasion of Pancreatic Cancer Cells. Sci Rep 2019; 9:4925. [PMID: 30894654 PMCID: PMC6426867 DOI: 10.1038/s41598-019-41346-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 03/05/2019] [Indexed: 12/28/2022] Open
Abstract
Pancreatic cancer has the worst prognosis among major malignancies, largely due to its highly invasive property and difficulty in early detection. Mechanistic insights into cancerous transformation and especially metastatic progression are imperative for developing novel treatment strategies. The actin-regulating protein CAP1 is implicated in human cancers, while the role still remains elusive. In this study, we investigated roles for CAP1 and its phosphor-regulation in pancreatic cancer cells. No evidence supports remarkable up-regulation of CAP1 in the panel of cancer cell lines examined. However, knockdown of CAP1 in cancer cells led to enhanced stress fibers, reduced cell motility and invasion into Matrigel. Phosphorylation of CAP1 at the S308/S310 tandem regulatory site was elevated in cancer cells, consistent with hyper-activated GSK3 reported in pancreatic cancer. Inhibition of GSK3, a kinase for S310, reduced cell motility and invasion. Moreover, phosphor mutants had defects in alleviating actin stress fibers and rescuing the reduced invasiveness in the CAP1-knockdown PANC-1 cells. These results suggest a required role for transient phosphorylation for CAP1 function in controlling cancer cell invasiveness. Depletion of CAP1 also reduced FAK activity and cell adhesion, but did not cause significant alterations in ERK or cell proliferation. CAP1 likely regulates cancer cell invasiveness through effects on both actin filament turnover and cell adhesion. Finally, the growth factor PDGF induced CAP1 dephosphorylation, suggesting CAP1 may mediate extracellular signals to control cancer cell invasiveness. These findings may ultimately help develop strategies targeting CAP1 or its regulatory signals for controlling the invasive cycle of the disease.
Collapse
|
32
|
Role of the Endocytosis of Caveolae in Intracellular Signaling and Metabolism. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 57:203-234. [PMID: 30097777 DOI: 10.1007/978-3-319-96704-2_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Caveolae are 60-80 nm invaginated plasma membrane (PM) nanodomains, with a specific lipid and protein composition, which assist and regulate multiple processes in the plasma membrane-ranging from the organization of signalling complexes to the mechanical adaptation to changes in PM tension. However, since their initial descriptions, these structures have additionally been found tightly linked to internalization processes, mechanoadaptation, to the regulation of signalling events and of endosomal trafficking. Here, we review caveolae biology from this perspective, and its implications for cell physiology and disease.
Collapse
|
33
|
Li X, Liu H, Sun L, Zhou X, Yuan X, Chen Y, Liu F, Liu Y, Xiao L. MicroRNA-302c modulates peritoneal dialysis-associated fibrosis by targeting connective tissue growth factor. J Cell Mol Med 2019; 23:2372-2383. [PMID: 30693641 PMCID: PMC6433681 DOI: 10.1111/jcmm.14029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
Long‐term peritoneal dialysis (PD) can lead to the induction of mesothelial/epithelial‐mesenchymal transition (MMT/EMT) and fibrosis; these effects eventually result in ultrafiltration failure and the discontinuation of PD. MicroRNA‐302c (miR‐302c) is believed to be involved in regulating tumour cell growth and metastasis by suppressing MMT, but the effect of miR‐302c on MMT in the context of PD is unknown. MiR‐302c levels were measured in mesothelial cells isolated from the PD effluents of continuous ambulatory peritoneal dialysis patients. After miR‐302c overexpression using lentivirus, human peritoneal mesothelial cell line (HMrSV5) and PD mouse peritoneum were treated with TGF‐β1 or high glucose peritoneal dialysate respectively. MiR‐302c expression level and MMT‐related factors alteration were observed. In addition, fibrosis of PD mouse peritoneum was alleviated by miR‐302c overexpression. Furthermore, the expression of connective tissue growth factor (CTGF) was negatively related by miR‐302c, and LV‐miR‐302c reversed the up‐regulation of CTGF induced by TGF‐β1. These data suggest that there is a novel TGF‐β1/miR‐302c/CTGF pathway that plays a significant role in the process of MMT and fibrosis during PD. MiR‐302c might be a potential biomarker for peritoneal fibrosis and a novel therapeutic target for protection against peritoneal fibrosis in PD patients.
Collapse
Affiliation(s)
- Xiejia Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xun Zhou
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xinke Yuan
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yusa Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li Xiao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
34
|
Ji L, Zhao G, Zhang P, Huo W, Dong P, Watari H, Jia L, Pfeffer LM, Yue J, Zheng J. Knockout of MTF1 Inhibits the Epithelial to Mesenchymal Transition in Ovarian Cancer Cells. J Cancer 2018; 9:4578-4585. [PMID: 30588241 PMCID: PMC6299381 DOI: 10.7150/jca.28040] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022] Open
Abstract
Due to peritoneal metastasis and frequent recurrence, ovarian cancer has the highest mortality among gynecological cancers. Epithelial to mesenchymal transition (EMT) contributes to ovarian tumor metastasis. In this study, we report for the first time that metal regulatory transcription factor 1 (MTF1) was upregulated in ovarian cancer, and its high expression was associated with poor patient survival and disease relapse. Knockout of MTF1 using lentiviral CRISPR/Cas9 nickase vector-mediated gene editing inhibited EMT by upregulating epithelial cell markers E-cadherin and cytokeratin 7, and downregulating mesenchymal markers Snai2 and β-catenin in ovarian cancer SKOV3 and OVCAR3 cells. Loss of MTF1 reduced cell proliferation, migration, and invasion in both SKOV3 and OVCAR3 cells. Knockout of MTF1 upregulated the expression of the KLF4 transcription factor, and attenuated two cellular survival pathways, ERK1/2 and AKT. Our studies demonstrated that MTF1 plays an oncogenic role and contributes to ovarian tumor metastasis by promoting EMT. MTF1 may be a novel biomarker for early diagnosis as well as a drug target for clinical therapy.
Collapse
Affiliation(s)
- Liang Ji
- Department of Anatomy, College of Basic Medical Science, Harbin Medical University, Harbin, China
| | - Guannan Zhao
- Department of Pathology and Laboratory Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Peng Zhang
- Department of Pathology and Laboratory Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Wenying Huo
- Department of Pathology and Laboratory Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Peixin Dong
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Obstetrics and Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Limin Jia
- Department of Anatomy, College of Basic Medical Science, Harbin Medical University, Harbin, China
| | - Lawrence M Pfeffer
- Department of Pathology and Laboratory Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, the University of Tennessee Health Science Center, Memphis, TN, 38163, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Jinhua Zheng
- Department of Anatomy, College of Basic Medical Science, Harbin Medical University, Harbin, China
| |
Collapse
|
35
|
Olea-Flores M, Juárez-Cruz JC, Mendoza-Catalán MA, Padilla-Benavides T, Navarro-Tito N. Signaling Pathways Induced by Leptin during Epithelial⁻Mesenchymal Transition in Breast Cancer. Int J Mol Sci 2018; 19:E3493. [PMID: 30404206 PMCID: PMC6275018 DOI: 10.3390/ijms19113493] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 10/27/2018] [Accepted: 11/01/2018] [Indexed: 12/12/2022] Open
Abstract
Leptin is an adipokine that is overexpressed in obese and overweight people. Interestingly, women with breast cancer present high levels of leptin and of its receptor ObR. Leptin plays an important role in breast cancer progression due to the biological processes it participates in, such as epithelial⁻mesenchymal transition (EMT). EMT consists of a series of orchestrated events in which cell⁻cell and cell⁻extracellular matrix interactions are altered and lead to the release of epithelial cells from the surrounding tissue. The cytoskeleton is also re-arranged, allowing the three-dimensional movement of epithelial cells into the extracellular matrix. This transition provides cells with the ability to migrate and invade adjacent or distal tissues, which is a classic feature of invasive or metastatic carcinoma cells. In recent years, the number of cases of breast cancer has increased, making this disease a public health problem worldwide and the leading cause of death due to cancer in women. In this review, we focus on recent advances that establish: (1) leptin as a risk factor for the development of breast cancer, and (2) leptin as an inducer of EMT, an event that promotes tumor progression.
Collapse
Affiliation(s)
- Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, México.
| | - Juan Carlos Juárez-Cruz
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, México.
| | - Miguel A Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, México.
| | - Teresita Padilla-Benavides
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, 364 Plantation Street, Worcester, MA 01605, USA.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, México.
| |
Collapse
|
36
|
Zhang X, Shang J, Wang X, Cheng G, Jiang Y, Liu D, Xiao J, Zhao Z. Microarray analysis reveals long non‑coding RNA SOX2OT as a novel candidate regulator in diabetic nephropathy. Mol Med Rep 2018; 18:5058-5068. [PMID: 30320339 PMCID: PMC6236268 DOI: 10.3892/mmr.2018.9534] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/14/2018] [Indexed: 01/24/2023] Open
Abstract
Diabetic nephropathy (DN) is a highly complex syndrome involving multiple dysregulated biological processes. Long non-coding RNAs (lncRNAs) are now believed to have an important function in various diseases. However, their roles in DN remain largely unknown. Therefore, the present study was performed in order to investigate the lncRNAs that have a crucial role in DN. db/db mice were used as a DN model while db/m mice served as a control to search for lncRNAs which may have important roles in DN. Microarray and bioinformatics analysis gave an overview of the features of differentially expressed genes. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analysis demonstrated the typical biological alterations in DN. A co-expression network of lncRNAs and mRNAs revealed the complex interaction pattern in DN conditions. Further data investigation indicated that SOX2-overlapping transcript (SOX2OT), which was significantly downregulated in DN mice, may be the potentially functional lncRNA contributing to the onset of DN. The UCSC database demonstrated that SOX2OT was highly conserved in mice and humans. Additionally further study using cultured human podocytes and mesangial cells confirmed the downregulation of SOX2OT using reverse transcription-quantitative polymerase chain reaction and fluorescence in situ hybridization. However, the cellular location of SOX2OT depended on certain cell types. Taken together, the results of the present study indicated that SOX2OT may act as an important regulator in the pathogenesis of DN by interacting with various mRNAs with critical roles in DN.
Collapse
Affiliation(s)
- Xiaoxue Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaoyang Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Genyang Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yumin Jiang
- Department of Emergency, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Dong Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jing Xiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhanzheng Zhao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
37
|
Tanaka S, Fujio Y, Nakayama H. Caveolae-Specific CaMKII Signaling in the Regulation of Voltage-Dependent Calcium Channel and Cardiac Hypertrophy. Front Physiol 2018; 9:1081. [PMID: 30131723 PMCID: PMC6090180 DOI: 10.3389/fphys.2018.01081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/19/2018] [Indexed: 02/04/2023] Open
Abstract
Cardiac hypertrophy is a major risk for the progression of heart failure; however, the underlying molecular mechanisms contributing to this process remain elusive. The caveolae microdomain plays pivotal roles in various cellular processes such as lipid homeostasis, signal transduction, and endocytosis, and also serves as a signaling platform. Although the caveolae microdomain has been postulated to have a major contribution to the development of cardiac pathologies, including cardiac hypertrophy, recent evidence has placed this role into question. Lack of direct evidence and appropriate methods for determining activation of caveolae-specific signaling has thus far limited the ability to obtain a definite answer to the question. In this review, we focus on the potential physiological and pathological roles of the multifunctional kinase Ca2+/calmodulin-dependent kinase II and voltage-dependent L-type calcium channel in the caveolae, toward gaining a better understanding of the contribution of caveolae-based signaling in cardiac hypertrophy.
Collapse
Affiliation(s)
- Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Hiroyuki Nakayama
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| |
Collapse
|
38
|
Rossi L, Battistelli C, de Turris V, Noce V, Zwergel C, Valente S, Moioli A, Manzione A, Palladino M, Bordoni V, Domenici A, Menè P, Mai A, Tripodi M, Strippoli R. HDAC1 inhibition by MS-275 in mesothelial cells limits cellular invasion and promotes MMT reversal. Sci Rep 2018; 8:8492. [PMID: 29855565 PMCID: PMC5981641 DOI: 10.1038/s41598-018-26319-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 05/10/2018] [Indexed: 12/20/2022] Open
Abstract
Peritoneal fibrosis is a pathological alteration of the peritoneal membrane occurring in a variety of conditions including peritoneal dialysis (PD), post-surgery adhesions and peritoneal metastases. The acquisition of invasive and pro-fibrotic abilities by mesothelial cells (MCs) through induction of MMT, a cell-specific form of EMT, plays a main role in this process. Aim of this study was to evaluate possible effects of histone deacetylase (HDAC) inhibitors, key components of the epigenetic machinery, in counteracting MMT observed in MCs isolated from effluent of PD patients. HDAC inhibitors with different class/isoform selectivity have been used for pharmacological inhibition. While the effect of other inhibitors was limited to a partial E-cadherin re-expression, MS-275, a HDAC1-3 inhibitor, promoted: (i) downregulation of mesenchymal markers (MMP2, Col1A1, PAI-1, TGFβ1, TGFβRI) (ii) upregulation of epithelial markers (E-cadherin, Occludin), (iii) reacquisition of an epithelial-like morphology and (iv) marked reduction of cellular invasiveness. Results were confirmed by HDAC1 genetic silencing. Mechanistically, MS-275 causes: (i) increase of nuclear histone H3 acetylation (ii) rescue of the acetylation profile on E-cadherin promoter, (iii) Snail functional impairment. Overall, our study, pinpointing a role for HDAC1, revealed a new player in the regulation of peritoneal fibrosis, providing the rationale for future therapeutic opportunities.
Collapse
Affiliation(s)
- Lucia Rossi
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Valeria de Turris
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Valeria Noce
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy
| | - Clemens Zwergel
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Sergio Valente
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Alessandra Moioli
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Andrea Manzione
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Marco Palladino
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Veronica Bordoni
- Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy
| | - Alessandro Domenici
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Paolo Menè
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Nephrology Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Antonello Mai
- Department of Chemistry and Technologies of Drugs, Sapienza University of Rome, Rome, Italy
| | - Marco Tripodi
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy. .,Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy.
| | - Raffaele Strippoli
- Department of Cellular Biotechnologies and Hematology, Section of Molecular Genetics, Sapienza University of Rome, Rome, Italy. .,Gene Expression Laboratory, National Institute for Infectious Diseases "Lazzaro Spallanzani" I.R.C.C.S., Rome, Italy.
| |
Collapse
|
39
|
Hosseinzadeh A, Javad-Moosavi SA, Reiter RJ, Hemati K, Ghaznavi H, Mehrzadi S. Idiopathic pulmonary fibrosis (IPF) signaling pathways and protective roles of melatonin. Life Sci 2018; 201:17-29. [PMID: 29567077 DOI: 10.1016/j.lfs.2018.03.032] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by the progressive loss of lung function due to tissue scarring. A variety of pro-inflammatory and pro-fibrogenic factors including interleukin‑17A, transforming growth factor β, Wnt/β‑catenin, vascular endothelial growth factor, platelet-derived growth factor, fibroblast growth factors, endotelin‑1, renin angiotensin system and impaired caveolin‑1 function are involved in the IPF pathogenesis. Current therapies for IPF have some limitations and this highlights the need for effective therapeutic agents to treat this fatal disease. Melatonin and its metabolites are broad-spectrum antioxidants that not only remove reactive oxygen and nitrogen species by radical scavenging but also up-regulate the expression and activity of endogenous antioxidants. Via these actions, melatonin and its metabolites modulate a variety of molecular pathways in different pathophysiological conditions. Herein, we review the signaling pathways involved in the pathophysiology of IPF and the potentially protective effects of melatonin on these pathways.
Collapse
Affiliation(s)
- Azam Hosseinzadeh
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Russel J Reiter
- Department of Cellular and Structural Biology, UT Health, San Antonio, TX, USA
| | - Karim Hemati
- Department of Anesthesiology, Iran University of Medical Sciences, Tehran, Iran; Department of Anesthesiology, Ilam University of Medical Sciences, Ilam, Iran
| | - Habib Ghaznavi
- Department of Pharmacology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saeed Mehrzadi
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Cav-1 deficiency promotes liver fibrosis in carbon tetrachloride (CCl 4)-induced mice by regulation of oxidative stress and inflammation responses. Biomed Pharmacother 2018; 102:26-33. [PMID: 29549726 DOI: 10.1016/j.biopha.2018.03.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/28/2018] [Accepted: 03/05/2018] [Indexed: 12/30/2022] Open
Abstract
Caveolin-1 (Cav-1), as a membrane protein involved in the formation of caveolae, binds steroid receptors and endothelial nitric oxide synthase, limiting its translocation and activation. In the present study, we investigated the role of Cav-1 in the progression of hepatic fibrosis induced by carbon tetrachloride (CCl4) in murine animals. Therefore, the wild type (WT) and Cav-1-knockout (Cav-1-/-) mice were used in our study and subjected to CCl4. The results indicated that CCl4 induced the decrease of Cav-1 expression in liver tissue samples. And Cav-1-/- intensified CCl4-triggered hepatic injury, evidenced by the stronger hepatic histological alterations, serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels and liver terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells. CCl4 led to oxidative stress, supported by the reduced superoxide dismutase (SOD) activity and glutathione (GSH) levels, as well as enhanced malondialdehyde (MDA) and O2- levels in liver samples. And the process was intensified by Cav-1-/-. Additionally, CCl4-caused hepatic inflammation was aggregated by Cav-1-/- via further increasing the secretion of pro-inflammatory cytokines. Moreover, CCl4-caused fibrosis was strengthened by Cav-1-/-, which was evidenced by the up-regulation of α-smooth muscle actin (α-SMA), collagen alpha 1 type 1 (Col1A1), lysyl oxidase (Lox) and transforming growth factor-β1 (TGF-β1) in liver tissues. Similar results were observed in TGF-β1-stimulated hepatic stellate cells (HSCs) and LX-2 cells without Cav-1 expressions that in vitro, suppressing Cav-1 further accelerated TGF-β1-induced oxidative stress, inflammation and fibrosis development. In conclusion, our results indicated that Cav-1 played an important role in CCl4-induced hepatic injury, which may be used as potential therapeutic target for hepatic fibrosis treatment.
Collapse
|
41
|
Magistri P, Battistelli C, Strippoli R, Petrucciani N, Pellinen T, Rossi L, Mangogna L, Aurello P, D'Angelo F, Tripodi M, Ramacciato G, Nigri G. SMO Inhibition Modulates Cellular Plasticity and Invasiveness in Colorectal Cancer. Front Pharmacol 2018; 8:956. [PMID: 29456503 PMCID: PMC5801594 DOI: 10.3389/fphar.2017.00956] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/15/2017] [Indexed: 12/17/2022] Open
Abstract
HIGHLIGHTS Preliminary results of this work were presented at the 2016 Academic Surgical Congress, Jacksonville (FL), February 2-4 2016 (Original title: Selective Smo-Inhibition Interferes With Cellular Energetic Metabolism In Colorectal Cancer)This study was funded by "Sapienza-University of Rome" (Funds for young researchers) and "AIRC" (Italian Association for Cancer Research)Hedgehog inhibitor was kindly provided by Genentech, Inc.®. Colon Cancer (CC) is the fourth most frequently diagnosed tumor and the second leading cause of death in the USA. Abnormalities of Hedgehog pathway have been demonstrated in several types of human cancers, however the role of Hedgehog (Hh) in CC remain controversial. In this study, we analyzed the association between increased mRNA expression of GLI1 and GLI2, two Hh target genes, and CC survival and recurrence by gene expression microarray from a cohort of 382 CC patients. We found that patients with increased expression of GLI1 showed a statistically significant reduction in survival. In order to demonstrate a causal role of Hh pathway activation in the pathogenesis of CC, we treated HCT 116, SW480 and SW620 CC cells lines with GDC-0449, a pharmacological inhibitor of Smoothened (SMO). Treatment with GDC-0449 markedly reduced expression of Hh target genes GLI1, PTCH1, HIP1, MUC5AC, thus indicating that this pathway is constitutively active in CC cell lines. Moreover, GDC-0449 partially reduced cell proliferation, which was associated with upregulation of p21 and downregulation of CycD1. Finally, treatment with the same drug reduced migration and three-dimensional invasion, which were associated with downregulation of Snail1, the EMT master gene, and with induction of the epithelial markers Cytokeratin-18 and E-cadherin. These results were confirmed by SMO genetic silencing. Notably, treatment with 5E1, a Sonic Hedgehog-specific mAb, markedly reduced the expression of Hedgehog target genes, as well as inhibited cell proliferation and mediated reversion toward an epithelial phenotype. This suggests the existence of a Hedgehog autocrine signaling loop affecting cell plasticity and fostering cell proliferation and migration/invasion in CC cell lines. These discoveries encourage future investigations to better characterize the role of Hedgehog in cellular plasticity and invasion during the different steps of CC pathogenesis.
Collapse
Affiliation(s)
- Paolo Magistri
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Raffaele Strippoli
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Niccolò Petrucciani
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Teijo Pellinen
- FIMM Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Lucia Rossi
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Livia Mangogna
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Paolo Aurello
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Francesco D'Angelo
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Marco Tripodi
- Molecular Genetics Section, Department of Cellular Biotechnology and Hematology, Sapienza—University of Rome, Rome, Italy
| | - Giovanni Ramacciato
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| | - Giuseppe Nigri
- Department of Medical and Surgical Sciences and Translational Medicine, Sapienza—University of Rome, Rome, Italy
| |
Collapse
|
42
|
Zhao G, Wang Q, Gu Q, Qiang W, Wei JJ, Dong P, Watari H, Li W, Yue J. Lentiviral CRISPR/Cas9 nickase vector mediated BIRC5 editing inhibits epithelial to mesenchymal transition in ovarian cancer cells. Oncotarget 2017; 8:94666-94680. [PMID: 29212257 PMCID: PMC5706903 DOI: 10.18632/oncotarget.21863] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 09/18/2017] [Indexed: 01/08/2023] Open
Abstract
BIRC5 encodes the protein survivin, a member of the inhibitor of apoptosis family. Survivin is highly expressed in a variety of cancers but has very low expression in the corresponding normal tissues, and its expression is often associated with tumor metastasis and chemoresistance. We report that survivin was highly expressed in ovarian cancer and strongly correlated with patient overall poor survival. For the first time, we provide experimental evidence that survivin is involved in epithelial to mesenchymal transition (EMT) in ovarian cancer cells. Lentiviral CRISPR/Cas9 nickase vector mediated BIRC5 gene editing led to the inhibition of EMT by upregulating epithelial cell marker, cytokeratin 7 and downregulating mesenchymal markers: snail2, β-catenin, and vimentin in both ovarian cancer SKOV3 and OVCAR3 cells. Consistent with this molecular approach, pharmacological treatment of ovarian cancer cells using a small molecule survivin inhibitor, YM155 also inhibited EMT in these ovarian cancer cell lines. Overexpression of BIRC5 promoted EMT in SKOV3 cells. Using molecular or pharmacological approaches, we found that cell proliferation, migration, and invasion were significantly inhibited following BIRC5 disruption in both cell lines. Inhibition of BIRC5 expression also sensitized cell responses to paclitaxel treatment. Moreover, loss of BIRC5 expression attenuated TGFβ signaling in both SKOV3 and OVCAR3 cells. Collectively, our studies demonstrated that disruption of BIRC5 expression inhibited EMT by attenuating the TGFβ pathway in ovarian cancer cells.
Collapse
Affiliation(s)
- Guannan Zhao
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, USA
| | - Qinghui Wang
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, USA
| | - Qingqing Gu
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, USA
| | - Wenan Qiang
- Department of Pathology, Department of Obstetrics and Gynecology, Northwestern University School of Medicine, Chicago, USA.,Center for Developmental Therapeutics, Chemistry of Life Processes Institute, Northwestern University, Evanston, USA
| | - Jian-Jun Wei
- Department of Pathology, Department of Obstetrics and Gynecology, Northwestern University School of Medicine, Chicago, USA
| | - Peixin Dong
- Department of Women's Health Educational System, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Hidemichi Watari
- Department of Women's Health Educational System, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Gynecology, Hokkaido University School of Medicine, Hokkaido University, Sapporo, Japan
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, USA
| | - Junming Yue
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Science Center, Memphis, USA.,Center for Cancer Research, University of Tennessee Health Science Center, Memphis, USA
| |
Collapse
|
43
|
Shihata WA, Putra MRA, Chin-Dusting JPF. Is There a Potential Therapeutic Role for Caveolin-1 in Fibrosis? Front Pharmacol 2017; 8:567. [PMID: 28970796 PMCID: PMC5609631 DOI: 10.3389/fphar.2017.00567] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/09/2017] [Indexed: 01/06/2023] Open
Abstract
Fibrosis is a process of dysfunctional wound repair, described by a failure of tissue regeneration and excessive deposition of extracellular matrix, resulting in tissue scarring and subsequent organ deterioration. There are a broad range of stimuli that may trigger, and exacerbate the process of fibrosis, which can contribute to the growing rates of morbidity and mortality. Whilst the process of fibrosis is widely described and understood, there are no current standard treatments that can reduce or reverse the process effectively, likely due to the continuing knowledge gaps surrounding the cellular mechanisms involved. Several cellular targets have been implicated in the regulation of the fibrotic process including membrane domains, ion channels and more recently mechanosensors, specifically caveolae, particularly since these latter contain various signaling components, such as members of the TGFβ and MAPK/ERK signaling pathways, all of which are key players in the process of fibrosis. This review explores the anti-fibrotic influences of the caveola, and in particular the key underpinning protein, caveolin-1, and its potential as a novel therapeutic target.
Collapse
Affiliation(s)
- Waled A Shihata
- Vascular Pharmacology Laboratory, Cardiovascular Disease Program, Department of Pharmacology, Biomedical Discovery Institute, Monash UniversityClayton, VIC, Australia.,Department of Medicine, Monash UniversityClayton, VIC, Australia.,Baker Heart and Diabetes InstituteMelbourne, VIC, Australia
| | - Mohammad R A Putra
- Vascular Pharmacology Laboratory, Cardiovascular Disease Program, Department of Pharmacology, Biomedical Discovery Institute, Monash UniversityClayton, VIC, Australia
| | - Jaye P F Chin-Dusting
- Vascular Pharmacology Laboratory, Cardiovascular Disease Program, Department of Pharmacology, Biomedical Discovery Institute, Monash UniversityClayton, VIC, Australia.,Department of Medicine, Monash UniversityClayton, VIC, Australia.,Baker Heart and Diabetes InstituteMelbourne, VIC, Australia
| |
Collapse
|
44
|
Biomarker research to improve clinical outcomes of peritoneal dialysis: consensus of the European Training and Research in Peritoneal Dialysis (EuTRiPD) network. Kidney Int 2017; 92:824-835. [PMID: 28797473 DOI: 10.1016/j.kint.2017.02.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 02/16/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022]
Abstract
Peritoneal dialysis (PD) therapy substantially requires biomarkers as tools to identify patients who are at the highest risk for PD-related complications and to guide personalized interventions that may improve clinical outcome in the individual patient. In this consensus article, members of the European Training and Research in Peritoneal Dialysis Network (EuTRiPD) review the current status of biomarker research in PD and suggest a selection of biomarkers that can be relevant to the care of PD patients and that are directly accessible in PD effluents. Currently used biomarkers such as interleukin-6, interleukin-8, ex vivo-stimulated interleukin-6 release, cancer antigen-125, and advanced oxidation protein products that were collected through a Delphi procedure were first triaged for inclusion as surrogate endpoints in a clinical trial. Next, novel biomarkers were selected as promising candidates for proof-of-concept studies and were differentiated into inflammation signatures (including interleukin-17, M1/M2 macrophages, and regulatory T cell/T helper 17), mesothelial-to-mesenchymal transition signatures (including microRNA-21 and microRNA-31), and signatures for senescence and inadequate cellular stress responses. Finally, the need for defining pathogen-specific immune fingerprints and phenotype-associated molecular signatures utilizing effluents from the clinical cohorts of PD patients and "omics" technologies and bioinformatics-biostatistics in future joint-research efforts was expressed. Biomarker research in PD offers the potential to develop valuable tools for improving patient management. However, for all biomarkers discussed in this consensus article, the association of biological rationales with relevant clinical outcomes remains to be rigorously validated in adequately powered, prospective, independent clinical studies.
Collapse
|
45
|
Martin S, Dudek-Peric AM, Garg AD, Roose H, Demirsoy S, Van Eygen S, Mertens F, Vangheluwe P, Vankelecom H, Agostinis P. An autophagy-driven pathway of ATP secretion supports the aggressive phenotype of BRAF V600E inhibitor-resistant metastatic melanoma cells. Autophagy 2017; 13:1512-1527. [PMID: 28722539 DOI: 10.1080/15548627.2017.1332550] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The ingrained capacity of melanoma cells to rapidly evolve toward an aggressive phenotype is manifested by their increased ability to develop drug-resistance, evident in the case of vemurafenib, a therapeutic-agent targeting BRAFV600E. Previous studies indicated a tight correlation between heightened melanoma-associated macroautophagy/autophagy and acquired Vemurafenib resistance. However, how this vesicular trafficking pathway supports Vemurafenib resistance remains unclear. Here, using isogenic human and murine melanoma cell lines of Vemurafenib-resistant and patient-derived melanoma cells with primary resistance to the BRAFV600E inhibitor, we found that the enhanced migration and invasion of the resistant melanoma cells correlated with an enhanced autophagic capacity and autophagosome-mediated secretion of ATP. Extracellular ATP (eATP) was instrumental for the invasive phenotype and the expansion of a subset of Vemurafenib-resistant melanoma cells. Compromising the heightened autophagy in these BRAFV600E inhibitor-resistant melanoma cells through the knockdown of different autophagy genes (ATG5, ATG7, ULK1), reduced their invasive and eATP-secreting capacity. Furthermore, eATP promoted the aggressive nature of the BRAFV600E inhibitor-resistant melanoma cells by signaling through the purinergic receptor P2RX7. This autophagy-propelled eATP-dependent autocrine-paracrine pathway supported the maintenance and expansion of a drug-resistant melanoma phenotype. In conclusion, we have identified an autophagy-driven response that relies on the secretion of ATP to drive P2RX7-based migration and expansion of the Vemurafenib-resistant phenotype. This emphasizes the potential of targeting autophagy in the treatment and management of metastatic melanoma.
Collapse
Affiliation(s)
- Shaun Martin
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium.,b Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg, Leuven , Belgium
| | - Aleksandra M Dudek-Peric
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium
| | - Abhishek D Garg
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium
| | - Heleen Roose
- c Department of Development and Regeneration, Cluster Stem Cell Biology and Embryology , University of Leuven (KU Leuven) , Campus Gasthuisberg, Leuven , Belgium
| | - Seyma Demirsoy
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium
| | - Sofie Van Eygen
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium
| | - Freya Mertens
- c Department of Development and Regeneration, Cluster Stem Cell Biology and Embryology , University of Leuven (KU Leuven) , Campus Gasthuisberg, Leuven , Belgium
| | - Peter Vangheluwe
- b Laboratory of Cellular Transport Systems, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg, Leuven , Belgium
| | - Hugo Vankelecom
- c Department of Development and Regeneration, Cluster Stem Cell Biology and Embryology , University of Leuven (KU Leuven) , Campus Gasthuisberg, Leuven , Belgium
| | - Patrizia Agostinis
- a Laboratory for Cell Death Research and Therapy, Department of Cellular and Molecular Medicine , University of Leuven (KU Leuven) , Campus Gasthuisberg , Leuven , Belgium
| |
Collapse
|
46
|
Abstract
Over the past decade, interest in caveolae biology has peaked. These small bulb-shaped plasma membrane invaginations of 50-80nm diameter present in most cell types have been upgraded from simple membrane structures to a more complex bona fide organelle. However, although caveolae are involved in several essential cellular functions and pathologies, the underlying molecular mechanisms remain poorly defined. Following the identification of caveolins and cavins as the main caveolae constituents, recent studies have brought new insight into their structural organization as a coat. In this review, we discuss how these new data on caveolae can be integrated in the context of their role in signaling and pathophysiology.
Collapse
|
47
|
Horejs CM, St-Pierre JP, Ojala JRM, Steele JAM, da Silva PB, Rynne-Vidal A, Maynard SA, Hansel CS, Rodríguez-Fernández C, Mazo MM, You AYF, Wang AJ, von Erlach T, Tryggvason K, López-Cabrera M, Stevens MM. Preventing tissue fibrosis by local biomaterials interfacing of specific cryptic extracellular matrix information. Nat Commun 2017; 8:15509. [PMID: 28593951 PMCID: PMC5472175 DOI: 10.1038/ncomms15509] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 04/04/2017] [Indexed: 12/22/2022] Open
Abstract
Matrix metalloproteinases (MMPs) contribute to the breakdown of tissue structures such as the basement membrane, promoting tissue fibrosis. Here we developed an electrospun membrane biofunctionalized with a fragment of the laminin β1-chain to modulate the expression of MMP2 in this context. We demonstrate that interfacing of the β1-fragment with the mesothelium of the peritoneal membrane via a biomaterial abrogates the release of active MMP2 in response to transforming growth factor β1 and rescues tissue integrity ex vivo and in vivo in a mouse model of peritoneal fibrosis. Importantly, our data demonstrate that the membrane inhibits MMP2 expression. Changes in the expression of epithelial-to-mesenchymal transition (EMT)-related molecules further point towards a contribution of the modulation of EMT. Biomaterial-based presentation of regulatory basement membrane signals directly addresses limitations of current therapeutic approaches by enabling a localized and specific method to counteract MMP2 release applicable to a broad range of therapeutic targets.
Collapse
Affiliation(s)
- Christine-Maria Horejs
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
| | - Jean-Philippe St-Pierre
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Juha R. M. Ojala
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
| | - Joseph A. M. Steele
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
| | - Patricia Barros da Silva
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
| | - Angela Rynne-Vidal
- Centro de Biología Molecular Severo Ochoa, CSIC, Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Stephanie A. Maynard
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Catherine S. Hansel
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Chemistry, Imperial College London, Imperial College Road, London SW7 2AZ, UK
| | - Clara Rodríguez-Fernández
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Manuel M. Mazo
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Amanda Y. F. You
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Alex J. Wang
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Thomas von Erlach
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
| | - Karl Tryggvason
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
- Cardiovascular and Metabolic Disorders Program, Duke-NUS, 8 College Road, Singapore 169857, Singapore
| | - Manuel López-Cabrera
- Centro de Biología Molecular Severo Ochoa, CSIC, Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Molly M. Stevens
- Department of Materials, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Bioengineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Institute of Biomedical Engineering, Imperial College London, Exhibition Road, London SW7 2AZ, UK
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, Stockholm 17177, Sweden
| |
Collapse
|
48
|
Che M, Shi T, Feng S, Li H, Zhang X, Feng N, Lou W, Dou J, Tang G, Huang C, Xu G, Qian Q, Sun S, He L, Wang H. The MicroRNA-199a/214 Cluster Targets E-Cadherin and Claudin-2 and Promotes High Glucose-Induced Peritoneal Fibrosis. J Am Soc Nephrol 2017; 28:2459-2471. [PMID: 28428333 DOI: 10.1681/asn.2016060663] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 02/15/2017] [Indexed: 12/16/2022] Open
Abstract
Serum response factor (SRF) was found to be involved in the phenotypic transition and fibrosis of the peritoneal membrane during treatment with peritoneal dialysis (PD), but the exact mechanism remains unclear. SRF regulates microRNAs (miRNAs) that contain the SRF-binding consensus (CArG) element in the promoter region. Therefore, we investigated whether the miR-199a/214 gene cluster, which contains a CArG element in its promoter, is directly regulated by SRF. High-glucose (HG) treatment significantly unregulated the expression of the miR-199a-5p/214-3p gene cluster in human peritoneal mesothelial cells (HPMCs). By chromatin immunoprecipitation and reporter assays, we found that SRF binds to the miR-199a-5p/214-3p gene cluster promoter after HG stimulation. In vitro, in HPMCs, silencing of miR-199a-5p or miR-214-3p inhibited the HG-induced phenotypic transition and cell migration but enhanced cell adhesion, whereas ectopic expression of mimic oligonucleotides had the opposite effects. Both miR-199a-5p and miR-214-3p targeted claudin-2 and E-cadherin mRNAs. In a PD rat model, treatment with an SRF inhibitor silenced miR-199a-5p and miR-214-3p and alleviated HG-PD fluid-induced damage and fibrosis. Overall, this study reveals a novel SRF-miR-199a/miR-214-E-cadherin/claudin-2 axis that mediates damage and fibrosis in PD.
Collapse
Affiliation(s)
- Mingwen Che
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Nephrology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China.,Department of Medicine, Hospital of PLA, Korla, Xinjiang, China
| | - Tiantian Shi
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Department of Medicine, Yangling Demonstration Zone Hospital, Yangling, Shaanxi, China; and
| | - Shidong Feng
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huan Li
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Xiaomin Zhang
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ning Feng
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Weijuan Lou
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianhua Dou
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guangbo Tang
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chen Huang
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guoshuang Xu
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Qi Qian
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China.,Mayo Clinic Department of Medicine, Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Mayo Graduate School, Rochester, Minnesota
| | - Shiren Sun
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lijie He
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China; .,Department of Nephrology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hanmin Wang
- Department of Nephrology, State Key Laboratory of Cancer Biology and Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi, China;
| |
Collapse
|
49
|
Rynne-Vidal A, Au-Yeung CL, Jiménez-Heffernan JA, Pérez-Lozano ML, Cremades-Jimeno L, Bárcena C, Cristóbal-García I, Fernández-Chacón C, Yeung TL, Mok SC, Sandoval P, López-Cabrera M. Mesothelial-to-mesenchymal transition as a possible therapeutic target in peritoneal metastasis of ovarian cancer. J Pathol 2017; 242:140-151. [PMID: 28247413 PMCID: PMC5468005 DOI: 10.1002/path.4889] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 12/28/2016] [Accepted: 02/17/2017] [Indexed: 12/12/2022]
Abstract
Peritoneal dissemination is the primary metastatic route of ovarian cancer (OvCa), and is often accompanied by the accumulation of ascitic fluid. The peritoneal cavity is lined by mesothelial cells (MCs), which can be converted into carcinoma‐associated fibroblasts (CAFs) through mesothelial‐to‐mesenchymal transition (MMT). Here, we demonstrate that MCs isolated from ascitic fluid (AFMCs) of OvCa patients with peritoneal implants also undergo MMT and promote subcutaneous tumour growth in mice. RNA sequencing of AFMCs revealed that MMT‐related pathways – including transforming growth factor (TGF)‐β signalling – are differentially regulated, and a gene signature was verified in peritoneal implants from OvCa patients. In a mouse model, pre‐induction of MMT resulted in increased peritoneal tumour growth, whereas interfering with the TGF‐β receptor reduced metastasis. MC‐derived CAFs showed activation of Smad‐dependent TGF‐β signalling, which was disrupted in OvCa cells, despite their elevated TGF‐β production. Accordingly, targeting Smad‐dependent signalling in the peritoneal pre‐metastatic niche in mice reduced tumour colonization, suggesting that Smad‐dependent MMT could be crucial in peritoneal carcinomatosis. Together, these results indicate that bidirectional communication between OvCa cells and MC‐derived CAFs, via TGF‐β‐mediated MMT, seems to be crucial to form a suitable metastatic niche. We suggest MMT as a possible target for therapeutic intervention and a potential source of biomarkers for improving OvCa diagnosis and/or prognosis. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Angela Rynne-Vidal
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain.,Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chi Lam Au-Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - José A Jiménez-Heffernan
- Departamento de Anatomía Patológica, Hospital Universitario La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
| | - María Luisa Pérez-Lozano
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Lucía Cremades-Jimeno
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Carmen Bárcena
- Departamento de Anatomía Patológica, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | - Tsz Lun Yeung
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Samuel C Mok
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Pilar Sandoval
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| | - Manuel López-Cabrera
- Centro de Biología Molecular-Severo Ochoa (CBMSO), Departamento de Biología Celular e Inmunología, Madrid, Spain
| |
Collapse
|
50
|
Genomic reprograming analysis of the Mesothelial to Mesenchymal Transition identifies biomarkers in peritoneal dialysis patients. Sci Rep 2017; 7:44941. [PMID: 28327551 PMCID: PMC5361179 DOI: 10.1038/srep44941] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 02/15/2017] [Indexed: 12/21/2022] Open
Abstract
Peritoneal dialysis (PD) is an effective renal replacement therapy, but a significant proportion of patients suffer PD-related complications, which limit the treatment duration. Mesothelial-to-mesenchymal transition (MMT) contributes to the PD-related peritoneal dysfunction. We analyzed the genetic reprograming of MMT to identify new biomarkers that may be tested in PD-patients. Microarray analysis revealed a partial overlapping between MMT induced in vitro and ex vivo in effluent-derived mesothelial cells, and that MMT is mainly a repression process being higher the number of genes that are down-regulated than those that are induced. Cellular morphology and number of altered genes showed that MMT ex vivo could be subdivided into two stages: early/epithelioid and advanced/non-epithelioid. RT-PCR array analysis demonstrated that a number of genes differentially expressed in effluent-derived non-epithelioid cells also showed significant differential expression when comparing standard versus low-GDP PD fluids. Thrombospondin-1 (TSP1), collagen-13 (COL13), vascular endothelial growth factor A (VEGFA), and gremlin-1 (GREM1) were measured in PD effluents, and except GREM1, showed significant differences between early and advanced stages of MMT, and their expression was associated with a high peritoneal transport status. The results establish a proof of concept about the feasibility of measuring MMT-associated secreted protein levels as potential biomarkers in PD.
Collapse
|