1
|
Yang Y, Xu Z, He S, Wang C, Li R, Zhang R, Li J, Yang Z, Li H, Liu S, Guo Q. Developmental dynamics mimicking inversely engineered pericellular matrix for articular cartilage regeneration. Biomaterials 2025; 317:123066. [PMID: 39742841 DOI: 10.1016/j.biomaterials.2024.123066] [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: 09/03/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
The mechanical mismatch of scaffold matrix-mesenchymal stem cells (MSCs) has been a longstanding issue in the clinical application of MSC-based therapy for articular cartilage (AC) regeneration. Existing tissue-engineered scaffolds underestimate the importance of the natural chondrocyte pericellular matrix (PCM). Here, we reveal the temporal and spatial characteristics of collagen distribution around the chondrocytes. Next, we demonstrate a rationally designed layer-by-layer single-cell encapsulation system which can mimic PCM mechanical responses and enhance MSC chondrogenesis via reestablished the mechanical coupling of PCM-like primitive matrix and chondrocytes. This successfully simulates the temporal and spatial characteristics of collagen secretion. Through investigation of the micromechanical environment of the cells and full-atom simulation analysis of TRPV4, we determine the specific mechanisms by which cellular mechanical forces near the cell are converted into biological signals. The TRPV4-YAP/TAZ-PI3K-Akt signaling pathway is involved in MSC cartilage formation through a joint analysis of the mRNA sequencing and spatial transcriptome results. In a rat model of articular cartilage defects, our inversely engineered pericellular matrix-encapsulated MSC-loaded scaffolds show regenerative performance that are superior to those of scaffolds loaded with only MSCs. These results demonstrate the feasibility of using a PCM-mimicking system to improve MSC chondrogenesis and the efficacy of AC repair.
Collapse
Affiliation(s)
- Yongkang Yang
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Ziheng Xu
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Songlin He
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, 999077, PR China
| | - Chao Wang
- Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Runmeng Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Ruiyang Zhang
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Jianwei Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China
| | - Zhen Yang
- Arthritis Clinical and Research Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, PR China; Arthritis Institute, Peking University, Beijing, 100044, PR China
| | - Hao Li
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, 100853, PR China.
| | - Shuyun Liu
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China.
| | - Quanyi Guo
- School of Medicine, Nankai University, Tianjin, 300071, PR China; Institute of Orthopedics, The First Medical Center, Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 28 Fuxing Road, Haidian District, Beijing, 100853, PR China; National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, 100853, PR China.
| |
Collapse
|
2
|
Zhang FR, Tang J, Lai Y, Mo SQ, Lin ZM, Lei QQ, Han CC, Zhou AD, Lv XF, Wang C, Ou JS, Zhou JG, Pang RP. Smooth muscle cell Piezo1 is essential for phenotypic switch and neointimal hyperplasia. Br J Pharmacol 2025; 182:2031-2048. [PMID: 39900041 DOI: 10.1111/bph.17436] [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: 01/20/2024] [Revised: 10/17/2024] [Accepted: 11/10/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND AND PURPOSE Disturbed blood flow is a critical factor in activation of vascular smooth muscle cells (VSMCs) and initiation of neointimal hyperplasia. The Piezo1 channel is a recent yet well-characterised mechanosensor that plays a vital role in vascular development and homeostasis. However, the role of VSMC Piezo1 in neointima development remains largely unknown. The purpose of this study is to investigate the functional role of Piezo1 channel in neointimal hyperplasia. EXPERIMENTAL APPROACH We measured the expression of Piezo1 in VSMC-rich neointima from human coronary artery samples and two mouse neointimal hyperplasia models which were induced by cast implantation or guidewire injury. We utilised VSMC-specific Piezo1 knockout mice to explore its function and the underlying mechanism of neointimal hyperplasia, both in vivo and in vitro. KEY RESULTS In human and mouse neointima samples, we observed a significant up-regulation of Piezo1 expression in the VSMC-rich neointima compared to the medial layer. VSMC-specific knockout of Piezo1 significantly reduced neointimal hyperplasia in both animal models. Activation of Piezo1 facilitates, whereas Piezo1 deficiency inhibits disturbed flow-induced cell proliferation, migration and synthetic phenotype switch. Mechanistic studies suggest that Piezo1 activates YAP and TAZ through Ca2+ and its downstream effectors calmodulin kinase II and calcineurin, which in turn drive VSMC proliferation and migration, thereby facilitating neointimal hyperplasia. CONCLUSIONS AND IMPLICATIONS These findings demonstrate a critical role of mechanosensitive Piezo1 channel in neointimal hyperplasia via modulating VSMC phenotype. Piezo1 channels may represent a novel therapeutic target for maladaptive vascular remodelling and occlusive vascular diseases.
Collapse
Affiliation(s)
- Fei-Ran Zhang
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Tang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Lai
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shi-Qi Mo
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhuo-Miao Lin
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing-Qing Lei
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cong-Cong Han
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - An-Dong Zhou
- Department of Clinical Medicine, the Second Clinical Medical School, Guangdong Medical University, Dongguan, China
| | - Xiao-Fei Lv
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cheng Wang
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jia-Guo Zhou
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Program of Vascular Diseases, the Eighth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Cardiovascular diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Rui-Ping Pang
- Department of Physiology, Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
3
|
Mukherjee P, Mahanty M, Dutta B, Rahaman SG, Sankaran KR, Liu Z, Rahaman SO. Trpv4-mediated mechanotransduction regulates the differentiation of valvular interstitial cells to myofibroblasts: implications for aortic valve stenosis. Am J Physiol Cell Physiol 2025; 328:C1558-C1570. [PMID: 40203884 DOI: 10.1152/ajpcell.00977.2024] [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: 12/11/2024] [Revised: 01/10/2025] [Accepted: 03/26/2025] [Indexed: 04/11/2025]
Abstract
As aortic valve stenosis (AVS) progresses, the valve tissue also stiffens. This increase in tissue stiffness causes the valvular interstitial cells (VICs) to transform into myofibroblasts in response. VIC-to-myofibroblast differentiation is critically involved in the development of AVS. Herein, we investigated the role of mechanosensitive Ca2+-permeant transient receptor potential vanilloid 4 (Trpv4) channels in matrix stiffness- and transforming growth factor β1 (TGFβ1)-induced VIC-myofibroblast activation. We confirmed Trpv4 functionality in primary mouse wild-type VICs compared with Trpv4 null VICs using live Ca2+ influx detection during application of its selective agonist and antagonist. Using physiologically relevant hydrogels of varying stiffness that respectively mimic healthy or diseased aortic valve tissue stiffness, we found that genetic ablation of Trpv4 blocked matrix stiffness- and TGFβ1-induced VIC-myofibroblast activation as determined by changes in morphology, alterations of expression of α-smooth muscle actin, and modulations of F-actin generation. Our results showed that N-terminal residues 30-130 in Trpv4 were crucial for cellular force generation and VIC-myofibroblast activation, while deletion of residues 1-30 had no noticeable negative effect on these processes. Collectively, these data suggest a differential regulatory role for Trpv4 in stiffness/TGFβ1-induced VIC-myofibroblast activation. Our data further showed that Trpv4 regulates stiffness/TGFβ1-induced PI3K-AKT activity that is required for VIC-myofibroblast differentiation and cellular force generation, suggesting a mechanism by which Trpv4 activity regulates VIC-myofibroblast activation. Altogether, these data identify a novel role for Trpv4 mechanotransduction in regulating VIC-myofibroblast activation, implicating Trpv4 as a potential therapeutic target to slow and/or reverse AVS development.NEW & NOTEWORTHY Aortic valve stenosis (AVS) progression involves stiffened valve tissue, driving valvular interstitial cells (VICs) to transform into myofibroblasts. This study highlights the role of Trpv4 channels in VIC activation triggered by matrix stiffness and TGFß1. Using hydrogels mimicking healthy and diseased valves, researchers found that Trpv4 regulates cellular force generation and differentiation via PI3K-AKT activity. These findings identify Trpv4 as a potential therapeutic target to slow or reverse AVS progression.
Collapse
Affiliation(s)
- Pritha Mukherjee
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Manisha Mahanty
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Suneha G Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Karunakaran R Sankaran
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, Missouri, United States
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland, United States
| |
Collapse
|
4
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
5
|
Mahanty M, Dutta B, Ou W, Zhu X, Bromberg JS, He X, Rahaman SO. Macrophage microRNA-146a is a central regulator of the foreign body response to biomaterial implants. Biomaterials 2025; 314:122855. [PMID: 39362025 PMCID: PMC11560625 DOI: 10.1016/j.biomaterials.2024.122855] [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: 05/07/2024] [Revised: 09/20/2024] [Accepted: 09/26/2024] [Indexed: 10/05/2024]
Abstract
Host recognition and immune-mediated foreign body response (FBR) to biomaterials can adversely affect the functionality of implanted materials. FBR presents a complex bioengineering and medical challenge due to the lack of current treatments, making the detailed exploration of its molecular mechanisms crucial for developing new and effective therapies. To identify key molecular targets underlying the generation of FBR, here we perform analysis of microRNAs (miR) and mRNAs responses to implanted biomaterials. We found that (a) miR-146a levels inversely affect macrophage accumulation, foreign body giant cell (FBGC) formation, and fibrosis in a murine implant model; (b) macrophage-derived miR-146a is a crucial regulator of the FBR and FBGC formation, as confirmed by global and cell-specific knockout of miR-146a; (c) miR-146a modulates genes related to inflammation, fibrosis, and mechanosensing; (d) miR-146a modulates tissue stiffness near the implant during FBR as assessed by atomic force microscopy; and (e) miR-146a is linked to F-actin production and cellular traction force induction as determined by traction force microscopy, which are vital for FBGC formation. These novel findings suggest that targeting macrophage miR-146a could be a selective strategy to inhibit FBR, potentially improving the biocompatibility of biomaterials.
Collapse
Affiliation(s)
- Manisha Mahanty
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Bidisha Dutta
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA
| | - Wenquan Ou
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, 20742, USA
| | - Xiaoping Zhu
- University of Maryland, Department of Veterinary Medicine, College Park, MD, 20742, USA
| | | | - Xiaoming He
- University of Maryland, Fischell Department of Bioengineering, College Park, MD, 20742, USA
| | - Shaik O Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD, 20742, USA.
| |
Collapse
|
6
|
Townson J, Progida C. The emerging roles of the endoplasmic reticulum in mechanosensing and mechanotransduction. J Cell Sci 2025; 138:JCS263503. [PMID: 39976266 DOI: 10.1242/jcs.263503] [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] [Indexed: 02/21/2025] Open
Abstract
Cells are continuously subjected to physical and chemical cues from the extracellular environment, and sense and respond to mechanical cues via mechanosensation and mechanotransduction. Although the role of the cytoskeleton in these processes is well known, the contribution of intracellular membranes has been long neglected. Recently, it has become evident that various organelles play active roles in both mechanosensing and mechanotransduction. In this Review, we focus on mechanosensitive roles of the endoplasmic reticulum (ER), the functions of which are crucial for maintaining cell homeostasis. We discuss the effects of mechanical stimuli on interactions between the ER, the cytoskeleton and other organelles; the role of the ER in intracellular Ca2+ signalling via mechanosensitive channels; and how the unfolded protein response and lipid homeostasis contribute to mechanosensing. The expansive structure of the ER positions it as a key intracellular communication hub, and we additionally explore how this may be leveraged to transduce mechanical signals around the cell. By synthesising current knowledge, we aim to shed light on the emerging roles of the ER in cellular mechanosensing and mechanotransduction.
Collapse
Affiliation(s)
- Jonathan Townson
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| | - Cinzia Progida
- Department of Biosciences, University of Oslo, Blindernveien 31, 0316 Oslo, Norway
| |
Collapse
|
7
|
Zhang P, Yang D, Li K, Zhang J, Wang Z, Ma F, Liao X, Ma S, Zeng X, Zhang X. Matrix stiffness regulates NPC invasiveness by modulating a mechanoresponsive TRPV4-Nox4-IL-8 signaling axis. J Cancer 2025; 16:1324-1334. [PMID: 39895789 PMCID: PMC11786026 DOI: 10.7150/jca.104235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/01/2025] [Indexed: 02/04/2025] Open
Abstract
Matrix stiffness is a critical determinant of tumorigenesis and cancer progression. Transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive calcium channel, regulates angiogenesis and stromal stiffness in various tumors. However, it is unclear whether matrix stiffness regulates the invasiveness of nasopharyngeal carcinoma (NPC) cells through TRPV4. In this study, we found that increased matrix stiffness of NPC tissues correlated with advanced tumor stages. Furthermore, simulation of high matrix stiffness in vitro upregulated TRPV4, and increased the migration, invasion, and epithelial mesenchymal transition (EMT) of NPC cells. Knockdown or pharmacological inhibition of TRPV4 significantly suppressed the calcium influx in NPC cells, and inhibited their invasiveness and EMT under high-stiffness conditions. Mechanistically, TRPV4 modulated the invasiveness of NPC cells in response to matrix stiffness via the NOX4/IL-8 axis. Notably, TRPV4 and IL-8 levels were significantly increased in the high-stiffness NPC tissues, and showed a positive correlation. Taken together, matrix stiffness promotes the malignant progression of NPC cells through the activation of the TRPV4/NOX4/IL-8 axis, which could be explored further as a potential target for NPC therapy.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Dunhui Yang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Kang Li
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Jin Zhang
- Department of Otolaryngology, The Second People's Hospital of Yibin, Yibin, Sichuan, China
| | - Zhen Wang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Fang Ma
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xianqin Liao
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Shibo Ma
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xianhai Zeng
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| | - Xiangmin Zhang
- Department of Otolaryngology, Longgang Otolaryngology hospital & Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen, Guangdong, China
| |
Collapse
|
8
|
Dutta B, Rahaman SG, Mukherjee P, Rahaman SO. Transient Receptor Potential Vanilloid 4 Calcium-Permeable Channel Contributes to Valve Stiffening in Aortic Stenosis. J Am Heart Assoc 2025; 14:e037931. [PMID: 39719407 DOI: 10.1161/jaha.124.037931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Aortic valve stenosis (AVS) is a progressive disease characterized by fibrosis, inflammation, calcification, and stiffening of the aortic valve leaflets, leading to disrupted blood flow. If untreated, AVS can progress to heart failure and death within 2 to 5 years. Uncovering the molecular mechanisms behind AVS is key for developing effective noninvasive therapies. Emerging evidence highlights that matrix stiffness affect gene expression, inflammation, and cell differentiation. Activation of valvular interstitial cells into myofibroblasts, along with excessive extracellular matrix accumulation and remodeling, are major contributors to AVS progression. Inflammation further exacerbates the disease, as macrophages infiltrate valve leaflets, enhancing inflammation, activating valvular interstitial cells, and driving extracellular matrix remodeling. Our lab and others have shown that the activities of macrophages and fibroblasts are sensitive to matrix stiffness. Previously, we identified mechanosensitive transient receptor potential vanilloid 4 (TRPV4) channels as key regulators of fibrosis and macrophage activation, implicating TRPV4 in AVS as a potential stiffness sensor. METHODS AND RESULTS Herein, we found elevated levels of TRPV4, α-smooth muscle actin, and cluster of differentiation 68 proteins in human AVS tissues compared with controls. Furthermore, the stiffening of human aortic valve tissue is associated with the levels of myofibroblasts, macrophages, and TRPV4 protein expression. In a mouse model, TRPV4 promoted valve stiffening during hypercholesterolemia-induced AVS. Additionally, TRPV4 mediated intracellular stiffness in valvular interstitial cells in response to transforming growth factor β1, which was blocked by the TRPV4 antagonist GSK2193874. CONCLUSIONS These findings reveal a novel mechanism linking TRPV4 to valve stiffening, providing insights into how extracellular matrix mechanical properties drive inflammation and fibrosis in AVS.
Collapse
Affiliation(s)
- Bidisha Dutta
- Department of Nutrition and Food Science University of Maryland College Park MD USA
| | - Suneha G Rahaman
- Department of Nutrition and Food Science University of Maryland College Park MD USA
| | - Pritha Mukherjee
- Department of Nutrition and Food Science University of Maryland College Park MD USA
| | - Shaik O Rahaman
- Department of Nutrition and Food Science University of Maryland College Park MD USA
| |
Collapse
|
9
|
Sánchez-Hernández R, Benítez-Angeles M, Hernández-Vega AM, Rosenbaum T. Recent advances on the structure and the function relationships of the TRPV4 ion channel. Channels (Austin) 2024; 18:2313323. [PMID: 38354101 PMCID: PMC10868539 DOI: 10.1080/19336950.2024.2313323] [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: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.
Collapse
Affiliation(s)
- Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Ana M. Hernández-Vega
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
10
|
Mukherjee P, Mahanty M, Dutta B, Rahaman SG, Sankaran KR, Rahaman SO. TRPV4-mediated Mechanotransduction Regulates the Differentiation of Valvular Interstitial Cells to Myofibroblasts: Implications for Aortic Stenosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622116. [PMID: 39574752 PMCID: PMC11580895 DOI: 10.1101/2024.11.05.622116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
As aortic valve stenosis (AVS) progresses, the valve tissue also stiffens. This increase in tissue stiffness causes the valvular interstitial cells (VICs) to transform into myofibroblasts in response. VIC-to-myofibroblast differentiation is critically involved in the development of AVS. Herein, we investigated the role of mechanosensitive Ca2+-permeant transient receptor potential vanilloid 4 (Trpv4) channels in matrix stiffness- and transforming growth factor β1 (TGFβ1)-induced VIC-myofibroblast activation. We confirmed Trpv4 functionality in primary mouse wild-type VICs compared to Trpv4 null VICs using live Ca2+ influx detection during application of its selective agonist and antagonist. Using physiologically relevant hydrogels of varying stiffness that respectively mimic healthy or diseased aortic valve tissue stiffness, we found that genetic ablation of Trpv4 blocked matrix stiffness- and TGFβ1-induced VIC-myofibroblast activation as determined by changes in morphology, alterations of expression of α-smooth muscle actin, and modulations of F-actin generation. Our results showed that N-terminal residues 30-130 in Trpv4 were crucial for cellular force generation and VIC-myofibroblast activation, while deletion of residues 1-30 had no noticeable negative effect on these processes. Collectively, these data suggest a differential regulatory role for Trpv4 in stiffness/TGFβ1-induced VIC-myofibroblast activation. Our data further showed that Trpv4 regulates stiffness/TGFβ1-induced PI3K-AKT activity that is required for VIC-myofibroblast differentiation and cellular force generation, suggesting a mechanism by which Trpv4 activity regulates VIC-myofibroblast activation. Altogether, these data identify a novel role for Trpv4 mechanotransduction in regulating VIC-myofibroblast activation, implicating Trpv4 as a potential therapeutic target to slow and/or reverse AVS development.
Collapse
Affiliation(s)
- Pritha Mukherjee
- University of Maryland, Department of Nutrition and Food Science, College Park, MD 20742
| | - Manisha Mahanty
- University of Maryland, Department of Nutrition and Food Science, College Park, MD 20742
| | - Bidisha Dutta
- University of Maryland, Department of Nutrition and Food Science, College Park, MD 20742
| | - Suneha G. Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD 20742
| | | | - Shaik O. Rahaman
- University of Maryland, Department of Nutrition and Food Science, College Park, MD 20742
| |
Collapse
|
11
|
Lan BQ, Wang YJ, Yu SX, Liu W, Liu YJ. Physical effects of 3-D microenvironments on confined cell behaviors. Am J Physiol Cell Physiol 2024; 327:C1192-C1201. [PMID: 39246142 DOI: 10.1152/ajpcell.00288.2024] [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: 04/30/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/10/2024]
Abstract
Cell migration is a fundamental and functional cellular process, influenced by a complex microenvironment consisting of different cells and extracellular matrix. Recent research has highlighted that, besides biochemical cues from the microenvironment, physical cues can also greatly alter cellular behavior. However, due to the complexity of the microenvironment, little is known about how the physical interactions between migrating cells and surrounding microenvironment instructs cell movement. Here, we explore various examples of three-dimensional microenvironment reconstruction models in vitro and describe how the physical interplay between migrating cells and the neighboring microenvironment controls cell behavior. Understanding this mechanical cooperation will provide key insights into organ development, regeneration, and tumor metastasis.
Collapse
Affiliation(s)
- Bao-Qiong Lan
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Ya-Jun Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Sai-Xi Yu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Wei Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, the People's Republic of China
| |
Collapse
|
12
|
Koskimäki S, Tojkander S. TRPV4-A Multifunctional Cellular Sensor Protein with Therapeutic Potential. SENSORS (BASEL, SWITZERLAND) 2024; 24:6923. [PMID: 39517820 PMCID: PMC11548305 DOI: 10.3390/s24216923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/02/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Transient receptor potential vanilloid (TRPV) channel proteins belong to the superfamily of TRP proteins that form cationic channels in the animal cell membranes. These proteins have various subtype-specific functions, serving, for example, as sensors for pain, pressure, pH, and mechanical extracellular stimuli. The sensing of extracellular cues by TRPV4 triggers Ca2+-influx through the channel, subsequently coordinating numerous intracellular signaling cascades in a spatio-temporal manner. As TRPV channels play such a wide role in various cellular and physiological functions, loss or impaired TRPV protein activity naturally contributes to many pathophysiological processes. This review concentrates on the known functions of TRPV4 sensor proteins and their potential as a therapeutic target.
Collapse
Affiliation(s)
- Sanna Koskimäki
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | | |
Collapse
|
13
|
Sheth M, Sharma M, Lehn M, Reza H, Takebe T, Takiar V, Wise-Draper T, Esfandiari L. Three-dimensional matrix stiffness modulates mechanosensitive and phenotypic alterations in oral squamous cell carcinoma spheroids. APL Bioeng 2024; 8:036106. [PMID: 39092008 PMCID: PMC11293878 DOI: 10.1063/5.0210134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
Extracellular biophysical cues such as matrix stiffness are key stimuli tuning cell fate and affecting tumor progression in vivo. However, it remains unclear how cancer spheroids in a 3D microenvironment perceive matrix mechanical stiffness stimuli and translate them into intracellular signals driving progression. Mechanosensitive Piezo1 and TRPV4 ion channels, upregulated in many malignancies, are major transducers of such physical stimuli into biochemical responses. Most mechanotransduction studies probing the reception of changing stiffness cues by cells are, however, still limited to 2D culture systems or cell-extracellular matrix models, which lack the major cell-cell interactions prevalent in 3D cancer tumors. Here, we engineered a 3D spheroid culture environment with varying mechanobiological properties to study the effect of static matrix stiffness stimuli on mechanosensitive and malignant phenotypes in oral squamous cell carcinoma spheroids. We find that spheroid growth is enhanced when cultured in stiff extracellular matrix. We show that the protein expression of mechanoreceptor Piezo1 and stemness marker CD44 is upregulated in stiff matrix. We also report the upregulation of a selection of genes with associations to mechanoreception, ion channel transport, extracellular matrix organization, and tumorigenic phenotypes in stiff matrix spheroids. Together, our results indicate that cancer cells in 3D spheroids utilize mechanosensitive ion channels Piezo1 and TRPV4 as means to sense changes in static extracellular matrix stiffness, and that stiffness drives pro-tumorigenic phenotypes in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Maulee Sheth
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio 45221, USA
| | - Manju Sharma
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio 45221, USA
| | - Maria Lehn
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio 45219, USA
| | - HasanAl Reza
- Division of Gastroenterology, Hepatology and Nutrition and Division of Developmental Biology, and Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| | | | | | | | | |
Collapse
|
14
|
Abdul Halim MS, Dyson JM, Gong MM, O'Bryan MK, Nosrati R. Fallopian tube rheology regulates epithelial cell differentiation and function to enhance cilia formation and coordination. Nat Commun 2024; 15:7411. [PMID: 39198453 PMCID: PMC11358425 DOI: 10.1038/s41467-024-51481-9] [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: 06/14/2023] [Accepted: 08/08/2024] [Indexed: 09/01/2024] Open
Abstract
The rheological properties of the extracellular fluid in the female reproductive tract vary spatiotemporally, however, the effect on the behaviour of epithelial cells that line the tract is unexplored. Here, we reveal that epithelial cells respond to the elevated viscosity of culture media by modulating their development and functionality to enhance cilia formation and coordination. Specifically, ciliation increases by 4-fold and cilia beating frequency decreases by 30% when cells are cultured at 100 mPa·s. Further, cilia manifest a coordinated beating pattern that can facilitate the formation of metachronal waves. At the cellular level, viscous loading activates the TRPV4 channel in the epithelial cells to increase intracellular Ca2+, subsequently decreasing the mitochondrial membrane potential level for ATP production to maintain cell viability and function. Our findings provide additional insights into the role of elevated tubal fluid viscosity in promoting ciliation and coordinating their beating-a potential mechanism to facilitate the transport of egg and embryo, suggesting possible therapeutic opportunities for infertility treatment.
Collapse
Affiliation(s)
- Melati S Abdul Halim
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia
| | - Jennifer M Dyson
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Max M Gong
- Department of Biomedical Engineering, Trine University, Angola, IN, USA
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Molecular Science and Biotechnology Institute, Faculty of Science, University of Melbourne, Parkville, Victoria, Australia
| | - Reza Nosrati
- Department of Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
15
|
Mancini A, Gentile MT, Pentimalli F, Cortellino S, Grieco M, Giordano A. Multiple aspects of matrix stiffness in cancer progression. Front Oncol 2024; 14:1406644. [PMID: 39015505 PMCID: PMC11249764 DOI: 10.3389/fonc.2024.1406644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 05/27/2024] [Indexed: 07/18/2024] Open
Abstract
The biophysical and biomechanical properties of the extracellular matrix (ECM) are crucial in the processes of cell differentiation and proliferation. However, it is unclear to what extent tumor cells are influenced by biomechanical and biophysical changes of the surrounding microenvironment and how this response varies between different tumor forms, and over the course of tumor progression. The entire ensemble of genes encoding the ECM associated proteins is called matrisome. In cancer, the ECM evolves to become highly dysregulated, rigid, and fibrotic, serving both pro-tumorigenic and anti-tumorigenic roles. Tumor desmoplasia is characterized by a dramatic increase of α-smooth muscle actin expressing fibroblast and the deposition of hard ECM containing collagen, fibronectin, proteoglycans, and hyaluronic acid and is common in many solid tumors. In this review, we described the role of inflammation and inflammatory cytokines, in desmoplastic matrix remodeling, tumor state transition driven by microenvironment forces and the signaling pathways in mechanotransduction as potential targeted therapies, focusing on the impact of qualitative and quantitative variations of the ECM on the regulation of tumor development, hypothesizing the presence of matrisome drivers, acting alongside the cell-intrinsic oncogenic drivers, in some stages of neoplastic progression and in some tumor contexts, such as pancreatic carcinoma, breast cancer, lung cancer and mesothelioma.
Collapse
Affiliation(s)
- Alessandro Mancini
- Department of Translational Medical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- BioUp Sagl, Lugano, Switzerland
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University “Giuseppe De Gennaro,” Casamassima, Bari, Italy
| | - Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Naples, NA, Italy
- Sbarro Health Research Organization (S.H.R.O.) Italia Foundation ETS, Candiolo, TO, Italy
| | - Michele Grieco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, United States
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
16
|
Mahanty M, Dutta B, Ou W, Zhu X, Bromberg JS, He X, Rahaman SO. Macrophage microRNA-146a is a central regulator of the foreign body response to biomaterial implants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.588018. [PMID: 38617341 PMCID: PMC11014630 DOI: 10.1101/2024.04.03.588018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Host recognition and immune-mediated foreign body response (FBR) to biomaterials can adversely affect the functionality of implanted materials. To identify key targets underlying the generation of FBR, here we perform analysis of microRNAs (miR) and mRNAs responses to implanted biomaterials. We found that (a) miR-146a levels inversely affect macrophage accumulation, foreign body giant cell (FBGC) formation, and fibrosis in a murine implant model; (b) macrophage-derived miR-146a is a crucial regulator of the FBR and FBGC formation, as confirmed by global and cell-specific knockout of miR-146a; (c) miR-146a modulates genes related to inflammation, fibrosis, and mechanosensing; (d) miR-146a modulates tissue stiffness near the implant during FBR; and (e) miR-146a is linked to F-actin production and cellular traction force induction, which are vital for FBGC formation. These novel findings suggest that targeting macrophage miR-146a could be a selective strategy to inhibit FBR, potentially improving the biocompatibility of biomaterials.
Collapse
|
17
|
Sacco JL, Vaneman ZT, Gomez EW. Extracellular matrix viscoelasticity regulates TGFβ1-induced epithelial-mesenchymal transition and apoptosis via integrin linked kinase. J Cell Physiol 2024; 239:e31165. [PMID: 38149820 DOI: 10.1002/jcp.31165] [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: 06/21/2023] [Revised: 10/06/2023] [Accepted: 11/17/2023] [Indexed: 12/28/2023]
Abstract
Transforming growth factor (TGF)-β1 is a multifunctional cytokine that plays important roles in health and disease. Previous studies have revealed that TGFβ1 activation, signaling, and downstream cell responses including epithelial-mesenchymal transition (EMT) and apoptosis are regulated by the elasticity or stiffness of the extracellular matrix. However, tissues within the body are not purely elastic, rather they are viscoelastic. How matrix viscoelasticity impacts cell fate decisions downstream of TGFβ1 remains unknown. Here, we synthesized polyacrylamide hydrogels that mimic the viscoelastic properties of breast tumor tissue. We found that increasing matrix viscous dissipation reduces TGFβ1-induced cell spreading, F-actin stress fiber formation, and EMT-associated gene expression changes, and promotes TGFβ1-induced apoptosis in mammary epithelial cells. Furthermore, TGFβ1-induced expression of integrin linked kinase (ILK) and colocalization of ILK with vinculin at cell adhesions is attenuated in mammary epithelial cells cultured on viscoelastic substrata in comparison to cells cultured on nearly elastic substrata. Overexpression of ILK promotes TGFβ1-induced EMT and reduces apoptosis in cells cultured on viscoelastic substrata, suggesting that ILK plays an important role in regulating cell fate downstream of TGFβ1 in response to matrix viscoelasticity.
Collapse
Affiliation(s)
- Jessica L Sacco
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Zachary T Vaneman
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Esther W Gomez
- Department of Chemical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, Pennsylvania, USA
| |
Collapse
|
18
|
Jiang D, Guo R, Dai R, Knoedler S, Tao J, Machens HG, Rinkevich Y. The Multifaceted Functions of TRPV4 and Calcium Oscillations in Tissue Repair. Int J Mol Sci 2024; 25:1179. [PMID: 38256251 PMCID: PMC10816018 DOI: 10.3390/ijms25021179] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
The transient receptor potential vanilloid 4 (TRPV4) specifically functions as a mechanosensitive ion channel and is responsible for conveying changes in physical stimuli such as mechanical stress, osmotic pressure, and temperature. TRPV4 enables the entry of cation ions, particularly calcium ions, into the cell. Activation of TRPV4 channels initiates calcium oscillations, which trigger intracellular signaling pathways involved in a plethora of cellular processes, including tissue repair. Widely expressed throughout the body, TRPV4 can be activated by a wide array of physicochemical stimuli, thus contributing to sensory and physiological functions in multiple organs. This review focuses on how TRPV4 senses environmental cues and thereby initiates and maintains calcium oscillations, critical for responses to organ injury, tissue repair, and fibrosis. We provide a summary of TRPV4-induced calcium oscillations in distinct organ systems, along with the upstream and downstream signaling pathways involved. In addition, we delineate current animal and disease models supporting TRPV4 research and shed light on potential therapeutic targets for modulating TRPV4-induced calcium oscillation to promote tissue repair while reducing tissue fibrosis.
Collapse
Affiliation(s)
- Dongsheng Jiang
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Ruiji Guo
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Ruoxuan Dai
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| | - Samuel Knoedler
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Division of Plastic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02152, USA
| | - Jin Tao
- Department of Physiology and Neurobiology and Centre for Ion Channelopathy, Medical College of Soochow University, Suzhou 215123, China;
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou 215123, China
| | - Hans-Günther Machens
- Department of Plastic and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
| | - Yuval Rinkevich
- Institute of Regenerative Biology and Medicine, Helmholtz Center Munich, 81377 Munich, Germany; (R.G.); (R.D.); (S.K.)
| |
Collapse
|
19
|
Mierke CT. Extracellular Matrix Cues Regulate Mechanosensing and Mechanotransduction of Cancer Cells. Cells 2024; 13:96. [PMID: 38201302 PMCID: PMC10777970 DOI: 10.3390/cells13010096] [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: 11/12/2023] [Revised: 12/29/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Extracellular biophysical properties have particular implications for a wide spectrum of cellular behaviors and functions, including growth, motility, differentiation, apoptosis, gene expression, cell-matrix and cell-cell adhesion, and signal transduction including mechanotransduction. Cells not only react to unambiguously mechanical cues from the extracellular matrix (ECM), but can occasionally manipulate the mechanical features of the matrix in parallel with biological characteristics, thus interfering with downstream matrix-based cues in both physiological and pathological processes. Bidirectional interactions between cells and (bio)materials in vitro can alter cell phenotype and mechanotransduction, as well as ECM structure, intentionally or unintentionally. Interactions between cell and matrix mechanics in vivo are of particular importance in a variety of diseases, including primarily cancer. Stiffness values between normal and cancerous tissue can range between 500 Pa (soft) and 48 kPa (stiff), respectively. Even the shear flow can increase from 0.1-1 dyn/cm2 (normal tissue) to 1-10 dyn/cm2 (cancerous tissue). There are currently many new areas of activity in tumor research on various biological length scales, which are highlighted in this review. Moreover, the complexity of interactions between ECM and cancer cells is reduced to common features of different tumors and the characteristics are highlighted to identify the main pathways of interaction. This all contributes to the standardization of mechanotransduction models and approaches, which, ultimately, increases the understanding of the complex interaction. Finally, both the in vitro and in vivo effects of this mechanics-biology pairing have key insights and implications for clinical practice in tumor treatment and, consequently, clinical translation.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Biological Physics Division, Peter Debye Institute of Soft Matter Physics, Faculty of Physics and Earth Science, Leipzig University, Linnéstraße 5, 04103 Leipzig, Germany
| |
Collapse
|
20
|
Zhao Q, Wang J, Qu S, Gong Z, Duan Y, Han L, Wang J, Wang C, Tan J, Yuan Q, Zhang Y. Neuro-Inspired Biomimetic Microreactor for Sensory Recovery and Hair Follicle Neogenesis under Skin Burns. ACS NANO 2023; 17:23115-23131. [PMID: 37934769 DOI: 10.1021/acsnano.3c09107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Deep burns are one of the most severe skin wounds, with typical symptoms being a contradiction between initial severe pain and a subsequent loss of sensation. Although it has long been known that sensory nerves promote skin regeneration and modulate skin function, no proven burn management strategies target sensory nerves. Here, a neuro-inspired biomimetic microreactor is designed based on the immune escape outer membrane of neuroblastoma cells and neural-associated intracellular proteins. The microreactor is constructed on a metal-organic framework (MOF) with a neuroblastoma membrane coating the surface and intracellular proteins loaded inside, called Neuro-MOF. It is loaded into a therapeutic hydrogel and triggers the release of its content proteins upon excitation by near-infrared light. The proteins compensate the skin microenvironment for permanent neurological damage after burns to initiate peripheral nerve regeneration and hair follicle niche formation. In addition, the neuroblastoma cell membrane is displayed on the surface of the Neuro-MOF microreactor, decreasing its immunogenicity and suppressing local inflammation. In a mouse model of deep skin burns, the Neuro-MOF microreactor exhibited significant functional skin regeneration effects, particularly sensory recovery and hair follicle neogenesis.
Collapse
Affiliation(s)
- Qin Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Jinyang Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Shuyuan Qu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Zijian Gong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Yiling Duan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Litian Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Jiaolong Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Can Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Jie Tan
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Quan Yuan
- Institute of Chemical Biology and Nanomedicine, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha 410082, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan 430079, China
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, School of Medicine, Wuhan University, Wuhan 430071, China
| |
Collapse
|
21
|
Sayedyahossein S, Thines L, Sacks DB. Ca 2+ signaling and the Hippo pathway: Intersections in cellular regulation. Cell Signal 2023; 110:110846. [PMID: 37549859 PMCID: PMC10529277 DOI: 10.1016/j.cellsig.2023.110846] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/02/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
The Hippo signaling pathway is a master regulator of organ size and tissue homeostasis. Hippo integrates a broad range of cellular signals to regulate numerous processes, such as cell proliferation, differentiation, migration and mechanosensation. Ca2+ is a fundamental second messenger that modulates signaling cascades involved in diverse cellular functions, some of which are also regulated by the Hippo pathway. Studies published over the last five years indicate that Ca2+ can influence core Hippo pathway components. Nevertheless, comprehensive understanding of the crosstalk between Ca2+ signaling and the Hippo pathway, and possible mechanisms through which Ca2+ regulates Hippo, remain to be elucidated. In this review, we summarize the multiple intersections between Ca2+ and the Hippo pathway and address the biological consequences.
Collapse
Affiliation(s)
- Samar Sayedyahossein
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Louise Thines
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| | - David B Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
22
|
Niu L, Lu YJ, Zu XW, Yang W, Shen FK, Xu YY, Jiang M, Xie Y, Li SY, Gao J, Bai G. Magnolol alleviates pulmonary fibrosis inchronic obstructive pulmonary disease by targeting transient receptor potential vanilloid 4-ankyrin repeat domain. Phytother Res 2023; 37:4282-4297. [PMID: 37282760 DOI: 10.1002/ptr.7907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/08/2023]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) plays a role in regulating pulmonary fibrosis (PF). While several TRPV4 antagonists including magnolol (MAG), have been discovered, the mechanism of action is not fully understood. This study aimed to investigate the effect of MAG on alleviating fibrosis in chronic obstructive pulmonary disease (COPD) based on TRPV4, and to further analyze its mechanism of action on TRPV4. COPD was induced using cigarette smoke and LPS. The therapeutic effect of MAG on COPD-induced fibrosis was evaluated. TRPV4 was identified as the main target protein of MAG using target protein capture with MAG probe and drug affinity response target stability assay. The binding sites of MAG at TRPV4 were analyzed using molecular docking and small molecule interaction with TRPV4-ankyrin repeat domain (ARD). The effects of MAG on TRPV4 membrane distribution and channel activity were analyzed by co-immunoprecipitation, fluorescence co-localization, and living cell assay of calcium levels. By targeting TRPV4-ARD, MAG disrupted the binding between phosphatidylinositol 3 kinase γ and TRPV4, leading to hampered membrane distribution on fibroblasts. Additionally, MAG competitively impaired ATP binding to TRPV4-ARD, inhibiting TRPV4 channel opening activity. MAG effectively blocked the fibrotic process caused by mechanical or inflammatory signals, thus alleviating PF in COPD. Targeting TRPV4-ARD presents a novel treatment strategy for PF in COPD.
Collapse
Affiliation(s)
- Lin Niu
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu-Jie Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Xing-Wang Zu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Wen Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Fu-Kui Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yan-Yan Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Min Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yang Xie
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Su-Yun Li
- The Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
- Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases co-constructed by Henan Province and Education Ministry of China, Henan University of Chinese Medicine, Zhengzhou, China
| | - Jie Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Gang Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
23
|
Xu HQ, Guo ZX, Yan JF, Wang SY, Gao JL, Han XX, Qin WP, Lu WC, Gao CH, Zhu WW, Fu YT, Jiao K. Fibrotic Matrix Induces Mesenchymal Transformation of Epithelial Cells in Oral Submucous Fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1208-1222. [PMID: 37328100 DOI: 10.1016/j.ajpath.2023.05.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/17/2023] [Accepted: 05/30/2023] [Indexed: 06/18/2023]
Abstract
Oral submucous fibrosis (OSF) is a potentially malignant disorder of the oral mucosa; however, whether and how the fibrotic matrix of OSF is involved in the malignant transformation of epithelial cells remains unknown. Herein, oral mucosa tissue from patients with OSF, OSF rat models, and their controls were used to observe the extracellular matrix changes and epithelial-mesenchymal transformation (EMT) in fibrotic lesions. Compared with controls, oral mucous tissues from patients with OSF showed an increased number of myofibroblasts, a decreased number of blood vessels, and increased type I and type III collagen levels. In addition, the oral mucous tissues from humans and OSF rats showed increased stiffness, accompanied by increased EMT activities of epithelial cells. The EMT activities of stiff construct-cultured epithelial cells were increased significantly by exogenous piezo-type mechanosensitive ion channel component 1 (Piezo1) activation, and decreased by yes-associated protein (YAP) inhibition. During ex vivo implantation, oral mucosal epithelial cells of the stiff group showed increased EMT activities and increased levels of Piezo1 and YAP compared with those in the sham and soft groups. These results indicate that increased stiffness of the fibrotic matrix in OSF led to increased proliferation and EMT of mucosal epithelial cells, in which the Piezo1-YAP signal transduction is important.
Collapse
Affiliation(s)
- Hao-Qing Xu
- The College of Life Science, Northwest University, Xi'an, China; Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Zhen-Xing Guo
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jian-Fei Yan
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Shu-Yan Wang
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Jia-Lu Gao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Xiao-Xiao Han
- The College of Life Science, Northwest University, Xi'an, China; Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Wen-Pin Qin
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Wei-Cheng Lu
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Chang-He Gao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China; The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Wei-Wei Zhu
- The College of Life Science, Northwest University, Xi'an, China; Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Yu-Tong Fu
- The College of Life Science, Northwest University, Xi'an, China; Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; State Key Laboratory of Stomatognathic Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
24
|
Zheng Y, Huang Q, Zhang Y, Geng L, Wang W, Zhang H, He X, Li Q. Multimodal roles of transient receptor potential channel activation in inducing pathological tissue scarification. Front Immunol 2023; 14:1237992. [PMID: 37705977 PMCID: PMC10497121 DOI: 10.3389/fimmu.2023.1237992] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/15/2023] [Indexed: 09/15/2023] Open
Abstract
Transient receptor potential (TRP) channels are a class of transmembrane proteins that can sense a variety of physical/chemical stimuli, participate in the pathological processes of various diseases and have attracted increasing attention from researchers. Recent studies have shown that some TRP channels are involved in the development of pathological scarification (PS) and directly participate in PS fibrosis and re-epithelialization or indirectly activate immune cells to release cytokines and neuropeptides, which is subdivided into immune inflammation, fibrosis, pruritus and mechanical forces increased. This review elaborates on the characteristics of TRP channels, the mechanism of PS and how TRP channels mediate the development of PS, summarizes the important role of TRP channels in the different pathogenesis of PS and proposes that therapeutic strategies targeting TRP will be important for the prevention and treatment of PS. TRP channels are expected to become new targets for PS, which will make further breakthroughs and provide potential pharmacological targets and directions for the in-depth study of PS.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiang He
- Department of Dermatology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiannan Li
- Department of Dermatology, Shuguang Hospital Affiliated with Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Verma BK, Chatterjee A, Kondaiah P, Gundiah N. Substrate Stiffness Modulates TGF-β Activation and ECM-Associated Gene Expression in Fibroblasts. Bioengineering (Basel) 2023; 10:998. [PMID: 37760100 PMCID: PMC10525202 DOI: 10.3390/bioengineering10090998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 06/19/2023] [Accepted: 07/06/2023] [Indexed: 09/29/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional cytokine that regulates the expression of ECM-associated genes during early injury. Tissue fibrosis development is driven by synergistic cues between the evolving biochemical and mechanical milieu. Few studies have addressed the role of substrate stiffness on TGF-β activity and extracellular matrix (ECM)-associated genes. We used a commercial formulation of polydimethylsiloxane (PDMS) to fabricate substrates of 40 kPa, 300 kPa, and 1.5 MPa stiffness, and cultured the HMF3S fibroblasts on substrates. We quantified TGF-β protein secreted by HMF3S cells on different substrates using a TGF-β responsive promoter reporter assay. We also tested for variations in gene expression levels on the substrates using RT-PCR and Western blotting and determined the MMP-2 and MMP-9 activities with gelatin zymography. The results showed that TGF-β protein activation was significantly compromised at lower stiffnesses. The expression of integrin α5 decreased on lower stiffness substrates and correlated with inefficient TGF-β protein activation. Collagen I, collagen III, and MMP-2 expression levels were lower on softer substrates; there was little MMP-9 activity on all substrates. Cell and nuclear morphologies were more rounded on compliant substrates, correlating with increased tubulin expression. Proliferations were higher on stiffer substrates, whereas cells on softer substrates showed cell cycle arrest. These results demonstrated critical feedback mechanisms between substrate stiffness and ECM regulation by fibroblasts, relevant in fibrosis.
Collapse
Affiliation(s)
- Brijesh Kumar Verma
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Aritra Chatterjee
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Paturu Kondaiah
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bengaluru 560012, India
| | - Namrata Gundiah
- Department of Mechanical Engineering, Indian Institute of Science, Bengaluru 560012, India
| |
Collapse
|
26
|
Moon SY, de Campos PS, Matte BF, Placone JK, Zanella VG, Martins MD, Lamers ML, Engler AJ. Cell contractility drives mechanical memory of oral squamous cell carcinoma. Mol Biol Cell 2023; 34:ar89. [PMID: 37342880 PMCID: PMC10398896 DOI: 10.1091/mbc.e22-07-0266] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 06/06/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023] Open
Abstract
Matrix stiffening is ubiquitous in solid tumors and can direct epithelial-mesenchymal transition (EMT) and cancer cell migration. Stiffened niche can even cause poorly invasive oral squamous cell carcinoma (OSCC) cell lines to acquire a less adherent, more migratory phenotype, but mechanisms and durability of this acquired "mechanical memory" are unclear. Here, we observed that contractility and its downstream signals could underlie memory acquisition; invasive SSC25 cells overexpress myosin II (vs. noninvasive Cal27 cells) consistent with OSCC. However, prolonged exposure of Cal27 cells to a stiff niche or contractile agonists up-regulated myosin and EMT markers and enabled them to migrate as fast as SCC25 cells, which persisted even when the niche softened and indicated "memory" of their prior niche. Stiffness-mediated mesenchymal phenotype acquisition required AKT signaling and was also observed in patient samples, whereas phenotype recall on soft substrates required focal adhesion kinase (FAK) activity. Phenotype durability was further observed in transcriptomic differences between preconditioned Cal27 cells cultured without or with FAK or AKT antagonists, and such transcriptional differences corresponded to discrepant patient outcomes. These data suggest that mechanical memory, mediated by contractility via distinct kinase signaling, may be necessary for OSCC to disseminate.
Collapse
Affiliation(s)
- So Youn Moon
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| | | | | | - Jesse K. Placone
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093
- Department of Physics and Engineering, West Chester University of Pennsylvania, West Chester, PA 19383
| | - Virgı´lio G. Zanella
- Department of Oral Pathology, Federal University of Rio Grande do Sul
- Department of Head and Neck Surgery, Santa Rita Hospital, Santa Casa de Misericórdia de Porto, Alegre
| | | | - Marcelo Lazzaron Lamers
- Department of Oral Pathology, Federal University of Rio Grande do Sul
- Deparment of Morphological Sciences, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, RS 90035, Brazil
| | - Adam J. Engler
- Department of Bioengineering, University of California San Diego, La Jolla, CA 92093
- Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037
| |
Collapse
|
27
|
Di X, Gao X, Peng L, Ai J, Jin X, Qi S, Li H, Wang K, Luo D. Cellular mechanotransduction in health and diseases: from molecular mechanism to therapeutic targets. Signal Transduct Target Ther 2023; 8:282. [PMID: 37518181 PMCID: PMC10387486 DOI: 10.1038/s41392-023-01501-9] [Citation(s) in RCA: 110] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 08/01/2023] Open
Abstract
Cellular mechanotransduction, a critical regulator of numerous biological processes, is the conversion from mechanical signals to biochemical signals regarding cell activities and metabolism. Typical mechanical cues in organisms include hydrostatic pressure, fluid shear stress, tensile force, extracellular matrix stiffness or tissue elasticity, and extracellular fluid viscosity. Mechanotransduction has been expected to trigger multiple biological processes, such as embryonic development, tissue repair and regeneration. However, prolonged excessive mechanical stimulation can result in pathological processes, such as multi-organ fibrosis, tumorigenesis, and cancer immunotherapy resistance. Although the associations between mechanical cues and normal tissue homeostasis or diseases have been identified, the regulatory mechanisms among different mechanical cues are not yet comprehensively illustrated, and no effective therapies are currently available targeting mechanical cue-related signaling. This review systematically summarizes the characteristics and regulatory mechanisms of typical mechanical cues in normal conditions and diseases with the updated evidence. The key effectors responding to mechanical stimulations are listed, such as Piezo channels, integrins, Yes-associated protein (YAP) /transcriptional coactivator with PDZ-binding motif (TAZ), and transient receptor potential vanilloid 4 (TRPV4). We also reviewed the key signaling pathways, therapeutic targets and cutting-edge clinical applications of diseases related to mechanical cues.
Collapse
Affiliation(s)
- Xingpeng Di
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xiaoshuai Gao
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Liao Peng
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Xi Jin
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Shiqian Qi
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Hong Li
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China
| | - Kunjie Wang
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| | - Deyi Luo
- Department of Urology and Institute of Urology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, P.R. China.
| |
Collapse
|
28
|
Liao X, Li X, Liu R. Extracellular-matrix mechanics regulate cellular metabolism: A ninja warrior behind mechano-chemo signaling crosstalk. Rev Endocr Metab Disord 2023; 24:207-220. [PMID: 36385696 DOI: 10.1007/s11154-022-09768-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2022] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the indispensable constituent of environmental cues, such as gravity, barometric pressure, vibration, and contact with bodies, which are involved in pattern and organogenesis, providing mechanical input to tissues and determining the ultimate fate of cells. Extracellular matrix (ECM) stiffness, the slow elastic force, carries the external physical force load onto the cell or outputs the internal force exerted by the cell and its neighbors into the environment. Accumulating evidence illustrates the pivotal role of ECM stiffness in the regulation of organogenesis, maintenance of tissue homeostasis, and the development of multiple diseases, which is largely fulfilled through its systematical impact on cellular metabolism. This review summarizes the establishment and regulation of ECM stiffness, the mechanisms underlying how ECM stiffness is sensed by cells and signals to modulate diverse cell metabolic pathways, and the physiological and pathological significance of the ECM stiffness-cell metabolism axis.
Collapse
Affiliation(s)
- Xiaoyu Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Xin Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, 14, Section 3, Renminnan Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
29
|
Zeng ML, Kong S, Chen TX, Peng BW. Transient Receptor Potential Vanilloid 4: a Double-Edged Sword in the Central Nervous System. Mol Neurobiol 2023; 60:1232-1249. [PMID: 36434370 DOI: 10.1007/s12035-022-03141-6] [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: 08/09/2022] [Accepted: 11/17/2022] [Indexed: 11/26/2022]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a nonselective cation channel that can be activated by diverse stimuli, such as heat, mechanical force, hypo-osmolarity, and arachidonic acid metabolites. TRPV4 is widely expressed in the central nervous system (CNS) and participates in many significant physiological processes. However, accumulative evidence has suggested that deficiency, abnormal expression or distribution, and overactivation of TRPV4 are involved in pathological processes of multiple neurological diseases. Here, we review the latest studies concerning the known features of this channel, including its expression, structure, and its physiological and pathological roles in the CNS, proposing an emerging therapeutic strategy for CNS diseases.
Collapse
Affiliation(s)
- Meng-Liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.,Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Tao-Xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, Taikang Medical School (School of Basic Medical Sciences), Wuhan University, Donghu Rd185#, Wuhan, 430071, Hubei, China.
| |
Collapse
|
30
|
Albrektsson T, Tengvall P, Amengual L, Coli P, Kotsakis GA, Cochran D. Osteoimmune regulation underlies oral implant osseointegration and its perturbation. Front Immunol 2023; 13:1056914. [PMID: 36761175 PMCID: PMC9902598 DOI: 10.3389/fimmu.2022.1056914] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/20/2022] [Indexed: 01/26/2023] Open
Abstract
In the field of biomaterials, an endosseous implant is now recognized as an osteoimmunomodulatory but not bioinert biomaterial. Scientific advances in bone cell biology and in immunology have revealed a close relationship between the bone and immune systems resulting in a field of science called osteoimmunology. These discoveries have allowed for a novel interpretation of osseointegration as representing an osteoimmune reaction rather than a classic bone healing response, in which the activation state of macrophages ((M1-M2 polarization) appears to play a critical role. Through this viewpoint, the immune system is responsible for isolating the implant biomaterial foreign body by forming bone around the oral implant effectively shielding off the implant from the host bone system, i.e. osseointegration becomes a continuous and dynamic host defense reaction. At the same time, this has led to the proposal of a new model of osseointegration, the foreign body equilibrium (FBE). In addition, as an oral wound, the soft tissues are involved with all their innate immune characteristics. When implant integration is viewed as an osteoimmune reaction, this has implications for how marginal bone is regulated. For example, while bacteria are constitutive components of the soft tissue sulcus, if the inflammatory front and immune reaction is at some distance from the marginal bone, an equilibrium is established. If however, this inflammation approaches the marginal bone, an immune osteoclastic reaction occurs and marginal bone is removed. A number of clinical scenarios can be envisioned whereby the osteoimmune equilibrium is disturbed and marginal bone loss occurs, such as complications of aseptic nature and the synergistic activation of pro-inflammatory pathways (implant/wear debris, DAMPs, and PAMPs). Understanding that an implant is a foreign body and that the host reacts osteoimmunologically to shield off the implant allows for a distinction to be drawn between osteoimmunological conditions and peri-implant bone loss. This review will examine dental implant placement as an osteoimmune reaction and its implications for marginal bone loss.
Collapse
Affiliation(s)
- T. Albrektsson
- Department of Biomaterials, University of Gothenburg, Gothenburg, Sweden
| | - P. Tengvall
- Department of Biomaterials, University of Gothenburg, Gothenburg, Sweden,*Correspondence: P. Tengvall,
| | - L. Amengual
- Dental Implantology Unit, Hospital Leonardo Guzmán, Antofagasta, Chile
| | - P. Coli
- Edinburgh Dental Specialists, Edinburgh, United Kingdom,Department of Prosthetic Dentistry and Dental Material Science, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden,Department of Dental Material Science, The Sahlgrenska Academy at Gothenburg University, Gothenburg, Sweden
| | - G. A. Kotsakis
- Department of Periodontology, University of Texas, San Antonio, TX, United States
| | - D. Cochran
- Department of Periodontology, University of Texas, San Antonio, TX, United States
| |
Collapse
|
31
|
Deng B, Zhao Z, Kong W, Han C, Shen X, Zhou C. Biological role of matrix stiffness in tumor growth and treatment. J Transl Med 2022; 20:540. [PMID: 36419159 PMCID: PMC9682678 DOI: 10.1186/s12967-022-03768-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/09/2022] [Indexed: 11/24/2022] Open
Abstract
In recent years, the biological role of changes in physical factors in carcinogenesis and progression has attracted increasing attention. Matrix stiffness, also known as ECM stress, is a critical physical factor of tumor microenvironment and remains alternating during carcinogenesis as a result of ECM remodeling through activation of cancer-associated fibroblasts and extracellular collagen accumulation, crosslinking and fibrosis. Different content and density of extracellular collagen in ECM endows matrix with varying stiffness. Physical signals induced by matrix stiffness are transmitted to tumor cells primarily by the integrins receptor family and trigger a series of mechanotransduction that result in changes in tumor cell morphology, proliferative capacity, and invasive ability. Importantly, accumulating evidence revealed that changes in matrix stiffness in tumor tissues greatly control the sensitivity of tumor cells in response to chemotherapy, radiotherapy, and immunotherapy through integrin signaling, YAP signaling, and related signaling pathways. Here, the present review analyzes the current research advances on matrix stiffness and tumor cell behavior with a view to contributing to tumor cell growth and treatment, with the hope of improving the understanding of the biological role of matrix stiffness in tumors.
Collapse
Affiliation(s)
- Boer Deng
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Ziyi Zhao
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Weimin Kong
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Chao Han
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China
| | - Xiaochang Shen
- grid.24696.3f0000 0004 0369 153XDepartment of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People’s Republic of China ,grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Chunxiao Zhou
- grid.10698.360000000122483208Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ,grid.10698.360000000122483208Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| |
Collapse
|
32
|
Zhu Z, Yu S, Niu K, Wang P. LGR5 promotes invasion and migration by regulating YAP activity in hypopharyngeal squamous cell carcinoma cells under inflammatory condition. PLoS One 2022; 17:e0275679. [PMID: 36288272 PMCID: PMC9604011 DOI: 10.1371/journal.pone.0275679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 09/21/2022] [Indexed: 11/09/2022] Open
Abstract
High leucine-rich repeat-containing G-protein coupled receptor 5 (LGR5) expression caused by an inflammatory condition was reported to promote tumor proliferation and the epithelial-mesenchymal transition (EMT) in various malignant tumors, but those effects have not been studied in hypopharyngeal squamous cell carcinoma (HSCC) and the molecular mechanism remains unclear. This study was aimed to determine whether YAP/TAZ is involved in the regulation of LGR5 expression in the inflammatory condition. Human hypopharyngeal carcinoma FaDu cells were stimulated with inflammatory medium. The cell invasion ability were evaluated through wound healing assay and transwell invasion assay. The expression levels of EMT-related proteins, LGR5, and p-YAP were detected by real time PCR, western blotting, and immunofluorescence. The results showed that LGR5 expression and the EMT process were significantly enhanced under inflammatory condition. The expression of EMT-related proteins was up-regulated, while that of p-YAP was decreased. After inhibiting the high LGR5 expression with short interfering RNA, the expression of EMT-related proteins was also down-regulated, while that of p-YAP was significantly increased. The use of verteporfin (VP), an inhibitor of YAP activity that promotes YAP phosphorylation, did not affect LGR5 expression. In conclusion, we suggest that the inflammatory condition leads to high LGR5 expression, which up-regulating the expression of EMT-related proteins by inhibiting the YAP phosphorylation.
Collapse
Affiliation(s)
- Zijia Zhu
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
- Department of Breast Surgery, Jilin University, Changchun, Jilin, China
| | - Shuyuan Yu
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Kai Niu
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ping Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
- * E-mail:
| |
Collapse
|
33
|
Miyano T, Suzuki A, Sakamoto N. Calcium influx through TRPV4 channels involve in hyperosmotic stress-induced epithelial-mesenchymal transition in tubular epithelial cells. Biochem Biophys Res Commun 2022; 617:48-54. [PMID: 35689842 DOI: 10.1016/j.bbrc.2022.06.003] [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: 04/28/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is a biological process that occurs in the pathogenesis of kidney diseases in which injured tubular epithelial cells transform into myofibroblasts. We previously showed that mannitol-mediated hyperosmotic stress induces EMT of tubular epithelial cells. Although Ca2+ signaling is essential for the induction of EMT in tubular epithelial cells, the role of specific calcium channels is unknown. In this study, we assessed the transient receptor potential vanilloid 4 (TRPV4)-mediated Ca2+ influx in the hyperosmolarity-induced EMT. The Fluo-4 assay was used to examine the effect of hyperosmotic stress on the intracellular Ca2+ level of normal rat kidney (NRK)-52E cells. Expression of a mesenchymal marker α-smooth muscle actin (α-SMA) and an epithelial marker E-cadherin was also observed by fluorescence microscopy. The hyperosmotic stress caused a transient increase in intracellular Ca2+ concentration as well as a decrease in E-cadherin and an increase in α-SMA expressions in tubular epithelial cells, indicating the induction of EMT. A TRPV4 channel antagonist inhibited hyperosmotic stress-induced Ca2+ influx and the EMT, whereas, a TRPV4 channel agonist increased Ca2+ influx and EMT induction in tubular epithelial cells without the hyperosmotic stress. These findings suggest that Ca2+ influx through TRPV4 channels contributes to the hyperosmotic stress-induced EMT of tubular epithelial cells.
Collapse
Affiliation(s)
- Takashi Miyano
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan.
| | - Atsushi Suzuki
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan.
| |
Collapse
|
34
|
Abstract
Transient receptor potential vanilloid 4 (TRPV4) channels are multi-modally activated cation permeable channels that are expressed most organ tissues including the skin. TRPV4 is highly expressed in the skin and functions in skin resident cells such as epidermal keratinocytes, melanocytes, immune mast cells and macrophages, and cutaneous neurons. TRPV4 plays many crucial roles in skin homeostasis to affect an extensive range of processes such as temperature sensation, osmo-sensation, hair growth, cell apoptosis, skin barrier integrity, differentiation, nociception and itch. Since TRPV4 functions in a plenitude of pathological states, TRPV4 can become a versatile therapeutic target for diseases such as chronic pain, itch and skin cancer.
Collapse
Affiliation(s)
- Carlene Moore
- Division of Headache and Division of Translational Brain Sciences, Department of Neurology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
35
|
Kuronuma K, Otsuka M, Wakabayashi M, Yoshioka T, Kobayashi T, Kameda M, Morioka Y, Chiba H, Takahashi H. Role of transient receptor potential vanilloid 4 in therapeutic anti-fibrotic effects of pirfenidone. Am J Physiol Lung Cell Mol Physiol 2022; 323:L193-L205. [PMID: 35787697 DOI: 10.1152/ajplung.00565.2020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, fatal lung disorder characterized by aberrant extracellular matrix deposition in the interstitium. Pirfenidone is an anti-fibrotic agent used to treat patients with IPF. Pirfenidone shows a pleiotropic mode of action, but its underlying anti-fibrotic mechanism is unclear. Transient receptor potential vanilloid 4 (TRPV4), which is a mechanosensitive calcium channel, was recently shown to be related to pulmonary fibrosis. To clarify the anti-fibrotic mechanisms of pirfenidone, we investigated whether TRPV4 blockade has a pharmacological effect in a murine model of pulmonary fibrosis and whether pirfenidone contributes to suppression of TRPV4. Our synthetic TRPV4 antagonist and pirfenidone treatment attenuated lung injury in the bleomycin mouse model. TRPV4-mediated increases in intracellular calcium were inhibited by pirfenidone. Additionally, TRPV4-stimulated interleukin-8 release from cells was reduced and a delay in cell migration was abolished by pirfenidone. Furthermore, pirfenidone decreased TRPV4 endogenous ligands in bleomycin-administered mouse lungs and their production by microsomes of human lungs. We found TRPV4 expression in the bronchiolar and alveolar epithelium and activated fibroblasts of the lungs in patients with IPF. Finally, we showed that changes in forced vital capacity of patients with IPF treated with pirfenidone were significantly correlated with metabolite levels of TRPV4 endogenous ligands in bronchoalveolar lavage fluid. These results suggest that the anti-fibrotic action of pirfenidone is partly mediated by TRPV4 and that TRPV4 endogenous ligands in bronchoalveolar lavage fluid may be biomarkers for distinguishing responders to pirfenidone.
Collapse
Affiliation(s)
- Koji Kuronuma
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuo Otsuka
- Department of Respiratory Medicine, Sapporo-Kosei General Hospital, Sapporo, Japan
| | - Masato Wakabayashi
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Takeshi Yoshioka
- Translational Research Unit, Biomarker R&D Department, Shionogi Co., Ltd., Osaka, Japan
| | - Tomofumi Kobayashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masami Kameda
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuhide Morioka
- Drug Discovery and Disease Research Laboratory, Shionogi Co., Ltd., Osaka, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroki Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
36
|
Chen Y, Su Y, Wang F. The Piezo1 ion channel in glaucoma: a new perspective on mechanical stress. Hum Cell 2022; 35:1307-1322. [PMID: 35767143 DOI: 10.1007/s13577-022-00738-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
Abstract
Glaucomatous optic nerve damage caused by pathological intraocular pressure elevation is irreversible, and its course is often difficult to control. This group of eye diseases is closely related to biomechanics, and the correlation between glaucoma pathogenesis and mechanical stimulation has been studied in recent decades. The nonselective cation channel Piezo1, the most important known mechanical stress sensor, is a transmembrane protein widely expressed in various cell types. Piezo1 has been detected throughout the eye, and the close relationship between Piezo1 and glaucoma is being confirmed. Pathological changes in glaucoma occur in both the anterior and posterior segments of the eye, and it is of great interest for researchers to determine whether Piezo1 plays a role in these changes and how it functions. The elucidation of the mechanisms of Piezo1 action in nonocular tissues and the reported roles of similar mechanically activated ion channels in glaucoma will provide an appropriate basis for further investigation. From a new perspective, this review provides a detailed description of the current progress in elucidating the role of Piezo1 in glaucoma, including relevant questions and assumptions, the remaining challenging research directions and mechanism-related therapeutic potential.
Collapse
Affiliation(s)
- Yidan Chen
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Yiyuan Road, Harbin, 150001, China
| | - Ying Su
- Eye Hospital, First Affiliated Hospital, Harbin Medical University, Yiman Road, Harbin, 150007, China.
| | - Feng Wang
- Department of Ophthalmology, Fourth Affiliated Hospital, Harbin Medical University, Yiyuan Road, Harbin, 150001, China.
| |
Collapse
|
37
|
Liang H, Li L, Zhu S, Tan J, Yang B, Wang X, Wu G, Xie C, Li L, Liu Z, Li Y, Song H, Chen G, Lin L. MicroRNA-744-5p suppresses tumorigenesis and metastasis of osteosarcoma through the p38 mitogen-activated protein kinases pathway by targeting transforming growth factor-beta 1. Bioengineered 2022; 13:12309-12325. [PMID: 35593122 PMCID: PMC9276001 DOI: 10.1080/21655979.2022.2072619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. Accumulating evidence has revealed that microRNAs (miRNAs) play a crucial role in the progression of OS. In this study, we found that miR-744-5p was the least expressed miRNA in patients with OS by analyzing GSE65071 from the GENE EXPRESSION OMNIBUS (GEO) database. Through real-time quantitative PCR (qRT-PCR), western blotting, colony formation assay, 5-Ethynyl-2-Deoxyuridine (EdU) incorporation assay, transwell migration, and invasion assays, we demonstrated its ability to inhibit the proliferation, migration, and invasion of OS cells in vitro. According to the luciferase reporter assay, transforming growth factor-β1 (TGFB1) was negatively regulated by miR-744-5p and reversed the effects of miR-744-5p on OS. Subcutaneous tumor-forming animal models and tail vein injection lung metastatic models were used in animal experiments, and it was found that miR-744-5p negatively regulated tumor growth and metastasis in vivo. Furthermore, rescue assays verified that miR-744-5p regulates TGFB1 expression in OS. Further experiments revealed that the p38 MAPK signaling pathway is involved in the miR-744-5p/TGFB1 axis. Generally, this study suggests that miR-744-5p is a negative regulator of TGFB1 and suppresses OS progression and metastasis via the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Haofeng Liang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,Department of orthopedics, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Lin Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shuang Zhu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jianye Tan
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Bingsheng Yang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoping Wang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guofeng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Xie
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lutao Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhengwei Liu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yucong Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Haoqiang Song
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guoli Chen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
38
|
Yimin YM, Huang X, Meng XC, Gu SC, Zhang ZW, Liu YH, Luo SY, Zan T. [Effects of transient receptor potential vanilloid type 4-specific activator on human vascular endothelial cell functions and blood supply of rat perforator flap and its mechanism]. ZHONGHUA SHAO SHANG YU CHUANG MIAN XIU FU ZA ZHI 2022; 38:434-446. [PMID: 35599419 DOI: 10.3760/cma.j.cn501120-20210419-00138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Objective: To analyze the effects of transient receptor potential vanilloid type 4 (TRPV4) activation on the function and endothelial-to-mesenchymal transition (EndMT) of human umbilical vein endothelial cells (HUVECs), as well as to explore the effects of TRPV4 activation on blood perfusion and survival of rat perforator flap and the mechanism. Methods: The experimental research methods were used. The 3rd to 6th passages of HUVECs were used for experiments and divided into 0.5 μmol/L 4α-phorbol 12, 13-didecanoate (4αPDD) group, 1.0 μmol/L 4αPDD group, 3.0 μmol/L 4αPDD group, 10.0 μmol/L 4αPDD group, and phosphate buffer solution (PBS) group, which were cultivated in corresponding final molarity of 4αPDD and PBS, respectively. The cell proliferation activity at 6 and 12 h of culture was detected using cell counting kit-8 (CCK-8). Another batch of cells was acquired and divided into PBS group, 1 μmol/L 4αPDD group, and 3 μmol/L 4αPDD group, which were treated similarly as described before and then detected for cell proliferation activity at 6, 12, 24, and 48 h of culture. The residual scratch area of cells at post scratch hour (PSH) 12, 24, and 48 was detected by scratch test, and the percentage of the residual scratch area was calculated. The number of migrated cells at 24 and 48 h of culture was detected by Transwell experiment. The tube-formation assay was used to measure the number of tubular structures at 4 and 8 h of culture. The protein expressions of E-cadherin, N-cadherin, Slug, and Snail at 24 h of culture were detected by Western blotting. All the sample numbers in each group at each time point in vitro experiments were 3. A total of 36 male Sprague-Dawley rats aged 8 to 10 weeks were divided into delayed flap group, 4αPDD group, and normal saline group according to the random number table, with 12 rats in each group, and iliolumbar artery perforator flap models on the back were constructed. The flap surgical delay procedure was only performed in the rats in delayed flap group one week before the flap transfer surgery. Neither rats in 4αPDD group nor normal saline group had flap surgical delay; instead, they were intraperitoneally injected with 4αPDD and an equivalent mass of normal saline, respectively, at 10 min before, 24 h after, and 48 h after the surgery. The general state of flap was observed on post surgery day (PSD) 0 (immediately), 1, 4, and 7. The flap survival rates were assessed on PSD 7. The flap blood perfusion was detected by laser speckle contrast imaging technique on PSD 1, 4, and 7. The microvascular density in the flap's choke vessel zone was detected by immunohistochemical staining. All the sample numbers in each group at each time point in vivo experiments were 12. Data were statistically analyzed with analysis of variance for factorial design, analysis of variance for repeated measurement, one-way analysis of variance, least significant difference t test, and Bonferroni correction. Results: At 6 and 12 h of culture, there were no statistically significant differences in cell proliferation activity in the overall comparison among PBS group, 0.5 μmol/L 4αPDD group, 1.0 μmol/L 4αPDD group, 3.0 μmol/L 4αPDD group, and 10.0 μmol/L 4αPDD group (P>0.05). At 6, 12, 24, and 48 h of culture, there were no statistically significant differences in cell proliferation activity in the overall comparison among PBS group, 1 μmol/L 4αPDD group, and 3 μmol/L 4αPDD group (P>0.05). At PSH 12, the percentages of the residual scratch area of cells in 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD group were close to that in PBS group (P>0.05). At PSH 24 and 48, compared with those in PBS group, the percentages of the residual scratch area of cells in 3 μmol/L 4αPDD group were significantly decreased (with t values of 2.83 and 2.79, respectively, P<0.05), while the percentages of the residual scratch area of cells in 1 μmol/L 4αPDD group showed no significant differences (P>0.05). At 24 h of culture, the number of migrated cells in 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD group were close to that in PBS group (P>0.05). At 48 h of culture, the number of migrated cells in 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD groups were significantly greater than that in PBS group (with t values of 6.20 and 9.59, respectively, P<0.01). At 4 h of culture, the numbers of tubular structures of cells in 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD group were significantly greater than that in PBS group (with t values of 4.68 and 4.95, respectively, P<0.05 or <0.01). At 8 h of culture, the numbers of tubular structures of cells in 1 μmol/L 4αPDD and 3 μmol/L 4αPDD groups were similar to that in PBS group (P>0.05). At 24 h of culture, compared with those in PBS group, the protein expression level of E-cadherin of cells in 3 μmol/L 4αPDD group was significantly decreased (t=5.13, P<0.01), whereas there was no statistically significant difference in the protein expression level of E-cadherin of cells in 1 μmol/L 4αPDD group (P>0.05); the protein expression level of N-cadherin of cells in 3 μmol/L 4αPDD group was significantly increased (t=4.93, P<0.01), whereas there was no statistically significant difference in the protein expression level of N-cadherin of cells in 1 μmol/L 4αPDD group (P>0.05); the protein expression levels of Slug of cells in 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD group were significantly increased (with t values of 3.85 and 6.52, respectively, P<0.05 or P<0.01); and the protein expression level of Snail of cells in 3 μmol/L 4αPDD group was significantly increased (t=4.08, P<0.05), whereas there was no statistically significant difference in the protein expression level of Snail of cells in 1 μmol/L 4αPDD group (P>0.05). There were no statistically significant differences in the protein expression levels of E-cadherin, N-cadherin, Slug, or Snail of cells between 1 μmol/L 4αPDD group and 3 μmol/L 4αPDD group (P>0.05). The general condition of flaps of rats in the three groups was good on PSD 0. On PSD 1, the flaps of rats in the three groups were basically similar, with bruising and swelling at the distal end. On PSD 4, the swelling of flaps of rats in the three groups subsided, and the distal end turned dark brown and necrosis occurred, with the area of necrosis in flaps of rats in normal saline group being larger than the areas in 4αPDD group and delayed flap group. On PSD 7, the necrotic areas of flaps of rats in the 3 groups were fairly stable, with the area of necrosis at the distal end of flap of rats in delayed flap group being the smallest. On PSD 7, the flap survival rates of rats in 4αPDD group ((80±13)%) and delayed flap group ((87±9)%) were similar (P>0.05), and both were significantly higher than (70±11)% in normal saline group (with t values of 2.24 and 3.65, respectively, P<0.05 or P<0.01). On PSD 1, the overall blood perfusion signals of rats in the 3 groups were basically the same, and the blood perfusion signals in the choke vessel zone were relatively strong, with a certain degree of underperfusion at the distal end. On PSD 4, the boundary between the surviving and necrotic areas of flaps of rats in the 3 groups became evident, and the blood perfusion signals in the choke vessel zone were improved, with the normal saline group's distal hypoperfused area of flap being larger than the areas in delayed flap group and 4αPDD group. On PSD 7, the blood perfusion signals of overall flap of rats had generally stabilized in the 3 groups, with the intensity of blood perfusion signal in the choke vessel zone and overall flap of rats in delayed flap group and 4αPDD group being significantly greater than that in normal saline group. On PSD 7, the microvascular density in the choke vessel zone of flap of rats in 4αPDD group and delayed flap group were similar (P>0.05), and both were significantly higher than that in normal saline group (with t values of 4.11 and 5.38, respectively, P<0.01). Conclusions: After activation, TRPV4 may promote the migration and tubular formation of human vascular endothelial cells via the EndMT pathway, leading to the enhanced blood perfusion of perforator flap and microvascular density in the choke vessel zone, and therefore increase the flap survival rate.
Collapse
Affiliation(s)
- Y M Yimin
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - X Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - X C Meng
- Department of Plastic and Cosmetic Surgery, the First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - S C Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Z W Zhang
- Department of Plastic and Reconstructive Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Y H Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - S Y Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - T Zan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
39
|
He J, Shan S, Li Q, Fang B, Xie Y. Mechanical Stretch Triggers Epithelial-Mesenchymal Transition in Keratinocytes Through Piezo1 Channel. Front Physiol 2022; 13:745572. [PMID: 35615675 PMCID: PMC9124769 DOI: 10.3389/fphys.2022.745572] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) process has emerged as a central regulator of embryonic development, tissue repair and tumor malignancy. In recent years, researchers have specifically focused on how mechanical signals drive the EMT program in epithelial cells. However, how epithelial cells specifically leverage mechanical force to control the EMT process remains unclear. Here, we show that the bona fide mechanically activated cation channel Piezo1 plays a critical role in the EMT. The Piezo1 is expressed in human primary epidermal keratinocytes (HEKs) and is responsible for the mechanical stretch-induced Ca2+ concentration. Inhibition of Piezo1 activation by the inhibitor GsMTx4 or by siRNA-mediated Piezo1 knockdown influenced the morphology and migration of HEKs. Moreover, Piezo1 activity also altered EMT-correlated markers expression in response to mechanical stretch. We propose that the mechanically activated cation channel Piezo1 is an important determinant of mechanical force-induced EMT in keratinocytes and might play similar roles in other epithelial cells.
Collapse
Affiliation(s)
| | | | | | - Bin Fang
- *Correspondence: Yun Xie, ; Bin Fang,
| | - Yun Xie
- *Correspondence: Yun Xie, ; Bin Fang,
| |
Collapse
|
40
|
Mukherjee P, Rahaman SG, Goswami R, Dutta B, Mahanty M, Rahaman SO. Role of mechanosensitive channels/receptors in atherosclerosis. Am J Physiol Cell Physiol 2022; 322:C927-C938. [PMID: 35353635 PMCID: PMC9109792 DOI: 10.1152/ajpcell.00396.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/09/2022] [Accepted: 03/22/2022] [Indexed: 11/22/2022]
Abstract
Mechanical forces are critical physical cues that can affect numerous cellular processes regulating the development, tissue maintenance, and functionality of cells. The contribution of mechanical forces is especially crucial in the vascular system where it is required for embryogenesis and for maintenance of physiological function in vascular cells including aortic endothelial cells, resident macrophages, and smooth muscle cells. Emerging evidence has also identified a role of these mechanical cues in pathological conditions of the vascular system such as atherosclerosis and associated diseases like hypertension. Of the different mechanotransducers, mechanosensitive ion channels/receptors are gaining prominence due to their involvement in numerous physiological and pathological conditions. However, only a handful of potential mechanosensory ion channels/receptors have been shown to be involved in atherosclerosis, and their precise role in disease development and progression remains poorly understood. Here, we provide a comprehensive account of recent studies investigating the role of mechanosensitive ion channels/receptors in atherosclerosis. We discuss the different groups of mechanosensitive proteins and their specific roles in inflammation, endothelial dysfunction, macrophage foam cell formation, and lesion development, which are crucial for the development and progression of atherosclerosis. Results of the studies discussed here will help in developing an understanding of the current state of mechanobiology in vascular diseases, specifically in atherosclerosis, which may be important for the development of innovative and targeted therapeutics for this disease.
Collapse
Affiliation(s)
- Pritha Mukherjee
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | | | - Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Manisha Mahanty
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| | - Shaik O Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, Maryland
| |
Collapse
|
41
|
Batan D, Peters DK, Schroeder ME, Aguado BA, Young MW, Weiss RM, Anseth KS. Hydrogel cultures reveal Transient Receptor Potential Vanilloid 4 regulation of myofibroblast activation and proliferation in valvular interstitial cells. FASEB J 2022; 36:e22306. [PMID: 35385164 PMCID: PMC9009405 DOI: 10.1096/fj.202101863r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/16/2022] [Accepted: 03/28/2022] [Indexed: 12/23/2022]
Abstract
As aortic valve stenosis develops, valve tissue becomes stiffer. In response to this change in environmental mechanical stiffness, valvular interstitial cells (VICs) activate into myofibroblasts. We aimed to investigate the role of mechanosensitive calcium channel Transient Receptor Potential Vanilloid type 4 (TRPV4) in stiffness induced myofibroblast activation. We verified TRPV4 functionality in VICs using live calcium imaging during application of small molecule modulators of TRPV4 activity. We designed hydrogel biomaterials that mimic mechanical features of healthy or diseased valve tissue microenvironments, respectively, to investigate the role of TRPV4 in myofibroblast activation and proliferation. Our results show that TRPV4 regulates VIC proliferation in a microenvironment stiffness-independent manner. While there was a trend toward inhibiting myofibroblast activation on soft microenvironments during TRPV4 inhibition, we observed near complete deactivation of myofibroblasts on stiff microenvironments. We further identified Yes-activated protein (YAP) as a downstream target for TRPV4 activity on stiff microenvironments. Mechanosensitive TRPV4 channels regulate VIC myofibroblast activation, whereas proliferation regulation is independent of the microenvironmental stiffness. Collectively, the data suggests differential regulation of stiffness-induced proliferation and myofibroblast activation. Our data further suggest a regulatory role for TRPV4 regarding YAP nuclear localization. TRPV4 is an important regulator for VIC myofibroblast activation, which is linked to the initiation of valve fibrosis. Although more validation studies are necessary, we suggest TRPV4 as a promising pharmaceutical target to slow aortic valve stenosis progression.
Collapse
Affiliation(s)
- Dilara Batan
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80303 USA
| | - Douglas K. Peters
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Megan E. Schroeder
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Brian A. Aguado
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Mark W. Young
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Robert M. Weiss
- Division of Cardiovascular Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kristi S. Anseth
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
42
|
Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol 2022; 15:34. [PMID: 35331296 PMCID: PMC8943941 DOI: 10.1186/s13045-022-01252-0] [Citation(s) in RCA: 219] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer microenvironment is critical for tumorigenesis and cancer progression. The extracellular matrix (ECM) interacts with tumor and stromal cells to promote cancer cells proliferation, migration, invasion, angiogenesis and immune evasion. Both ECM itself and ECM stiffening-induced mechanical stimuli may activate cell membrane receptors and mechanosensors such as integrin, Piezo1 and TRPV4, thereby modulating the malignant phenotype of tumor and stromal cells. A better understanding of how ECM stiffness regulates tumor progression will contribute to the development of new therapeutics. The rapidly expanding evidence in this research area suggests that the regulators and effectors of ECM stiffness represent potential therapeutic targets for cancer. This review summarizes recent work on the regulation of ECM stiffness in cancer, the effects of ECM stiffness on tumor progression, cancer immunity and drug resistance. We also discuss the potential targets that may be druggable to intervene ECM stiffness and tumor progression. Based on these advances, future efforts can be made to develop more effective and safe drugs to interrupt ECM stiffness-induced oncogenic signaling, cancer progression and drug resistance.
Collapse
|
43
|
Zeng ML, Cheng JJ, Kong S, Yang XL, Jia XL, Cheng XL, Chen L, He FG, Liu YM, Fan YT, Gongga L, Chen TX, Liu WH, He XH, Peng BW. Inhibition of Transient Receptor Potential Vanilloid 4 (TRPV4) Mitigates Seizures. Neurotherapeutics 2022; 19:660-681. [PMID: 35182379 PMCID: PMC9226259 DOI: 10.1007/s13311-022-01198-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2022] [Indexed: 02/08/2023] Open
Abstract
Astrocytes are critical regulators of the immune/inflammatory response in several human central nervous system (CNS) diseases. Emerging evidence suggests that dysfunctional astrocytes are crucial players in seizures. The objective of this study was to investigate the role of transient receptor potential vanilloid 4 (TRPV4) in 4-aminopyridine (4-AP)-induced seizures and the underlying mechanism. We also provide evidence for the role of Yes-associated protein (YAP) in seizures. 4-AP was administered to mice or primary cultured astrocytes. YAP-specific small interfering RNA (siRNA) was administered to primary cultured astrocytes. Mouse brain tissue and surgical specimens from epileptic patient brains were examined, and the results showed that TRPV4 was upregulated, while astrocytes were activated and polarized to the A1 phenotype. The levels of glial fibrillary acidic protein (GFAP), cytokine production, YAP, signal transducer activator of transcription 3 (STAT3), intracellular Ca2+([Ca2+]i) and the third component of complement (C3) were increased in 4-AP-induced mice and astrocytes. Perturbations in the immune microenvironment in the brain were balanced by TRPV4 inhibition or the manipulation of [Ca2+]i in astrocytes. Knocking down YAP with siRNA significantly inhibited 4-AP-induced pathological changes in astrocytes. Our study demonstrated that astrocytic TRPV4 activation promoted neuroinflammation through the TRPV4/Ca2+/YAP/STAT3 signaling pathway in mice with seizures. Astrocyte TRPV4 inhibition attenuated neuroinflammation, reduced neuronal injury, and improved neurobehavioral function. Targeting astrocytic TRPV4 activation may provide a promising therapeutic approach for managing epilepsy.
Collapse
Affiliation(s)
- Meng-liu Zeng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Jing-jing Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Shuo Kong
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xing-liang Yang
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xiang-lei Jia
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Xue-lei Cheng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Ling Chen
- Institute of Forensic Medicine, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Fang-gang He
- Institute of Forensic Medicine, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Yu-min Liu
- Department of Neurology, Zhongnan Hospital, Wuhan University, Donghu Road 169#, 430071 Wuhan, Hubei China
| | - Yuan-teng Fan
- Department of Neurology, Zhongnan Hospital, Wuhan University, Donghu Road 169#, 430071 Wuhan, Hubei China
| | - Lanzi Gongga
- Tibet University Medical College, 850000 Lhasa, Tibet China
| | - Tao-xiang Chen
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| | - Wan-hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Xiao-hua He
- Department of Pathophysiology, School of Basic Medical Sciences, Wuhan University, 430071 Wuhan, Hubei China
| | - Bi-wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disease, School of Basic Medical Sciences, Wuhan University, Donghu Rd185#, 430071 Wuhan, Hubei China
| |
Collapse
|
44
|
Oghbaei F, Zarezadeh R, Jafari-Gharabaghlou D, Ranjbar M, Nouri M, Fattahi A, Imakawa K. Epithelial-mesenchymal transition process during embryo implantation. Cell Tissue Res 2022; 388:1-17. [PMID: 35024964 DOI: 10.1007/s00441-021-03574-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/29/2021] [Indexed: 03/01/2023]
Abstract
The epithelial to mesenchymal transition (EMT) in endometrial epithelial and trophectoderm cells is essential for the progression of embryo implantation and its impairment could cause implantation failure. Therefore, EMT should be tightly regulated in both embryonic and endometrial cells during implantation. Studies reported the involvement of numerous factors in EMT regulation, including hormones, growth factors, transcription factors, microRNAs, aquaporins (AQPs), and ion channels. These factors act through different signaling pathways to affect the expression of epithelial and mesenchymal markers as well as the cellular cytoskeleton. Although the mechanisms involved in cancer cell EMT have been well studied, little is known about EMT during embryo implantation. Therefore, we comprehensively reviewed different factors that regulate the EMT, a key event required for the conceptus implantation to the endometrium.Summary sentence: Abnormal epithelial-mesenchymal transition (EMT) process within endometrial epithelial cells (EECs) or trophoblast cells can cause implantation failure. This process is regulated by various factors. Thus, the objective of this review was to summarize the effective factors on the EMT process during implantation.
Collapse
Affiliation(s)
- Farnaz Oghbaei
- Department of Basic Medical Sciences, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davoud Jafari-Gharabaghlou
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Minoo Ranjbar
- Department of Midwifery, Bonab Branch, Islamic Azad University, Bonab, Iran
| | - Mohammad Nouri
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kazuhiko Imakawa
- Laboratory of Molecular Reproduction, Research Institute of Agriculture, Tokai University, Kumamoto, 862-8652, Japan
| |
Collapse
|
45
|
Subbalakshmi AR, Ashraf B, Jolly MK. Biophysical and biochemical attributes of hybrid epithelial/mesenchymal phenotypes. Phys Biol 2022; 19. [PMID: 34986465 DOI: 10.1088/1478-3975/ac482c] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 01/05/2022] [Indexed: 11/11/2022]
Abstract
The Epithelial-Mesenchymal Transition (EMT) is a biological phenomenon associated with explicit phenotypic and molecular changes in cellular traits. Unlike the earlier-held popular belief of it being a binary process, EMT is now thought of as a landscape including diverse hybrid E/M phenotypes manifested by varying degrees of the transition. These hybrid cells can co-express both epithelial and mesenchymal markers and/or functional traits, and can possess the property of collective cell migration, enhanced tumor-initiating ability, and immune/targeted therapy-evasive features, all of which are often associated with worse patient outcomes. These characteristics of the hybrid E/M cells have led to a surge in studies that map their biophysical and biochemical hallmarks that can be helpful in exploiting their therapeutic vulnerabilities. This review discusses recent advances made in investigating hybrid E/M phenotype(s) from diverse biophysical and biochemical aspects by integrating live cell-imaging, cellular morphology quantification and mathematical modelling, and highlights a set of questions that remain unanswered about the dynamics of hybrid E/M states.
Collapse
Affiliation(s)
- Ayalur Raghu Subbalakshmi
- Indian Institute of Science, Centre for BioSystems Science and Engineering, Bangalore, 560012, INDIA
| | - Bazella Ashraf
- Central University of Kashmir, Department of Biotechnology, Ganderbal, Jammu and Kashmir, 191201, INDIA
| | - Mohit Kumar Jolly
- Indian Institute of Science, Centre for BioSystems Science and Engineering, Bangalore, 560012, INDIA
| |
Collapse
|
46
|
Gargini R, Segura-Collar B, Garranzo-Asensio M, Hortigüela R, Iglesias-Hernández P, Lobato-Alonso D, Moreno-Raja M, Esteban-Martin S, Sepúlveda-Sánchez JM, Nevola L, Sánchez-Gómez P. IDP-410: a Novel Therapeutic Peptide that Alters N-MYC Stability and Reduces Angiogenesis and Tumor Progression in Glioblastomas. Neurotherapeutics 2022; 19:408-420. [PMID: 35099769 PMCID: PMC9130446 DOI: 10.1007/s13311-021-01176-6] [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] [Accepted: 12/14/2021] [Indexed: 01/03/2023] Open
Abstract
Glioblastomas (GBMs) are the most frequent and highly aggressive brain tumors, being resistant to all cytotoxic and molecularly targeted agents tested so far. There is, therefore, an urgent need to find novel therapeutic approaches and/or alternative targets to bring treatment options to patients. Here, we first show that GBMs express high levels of N-MYC protein, a transcription factor involved in normal brain development. A novel stapled peptide designed to specifically target N-MYC protein monomer, IDP-410, is able to impair the formation of N-MYC/MAX complex and reduce the stability of N-MYC itself. As a result, the viability of GBM cells is compromised. Moreover, the efficacy is found dependent on the levels of expression of N-MYC. Finally, we demonstrate that IDP-410 reduces GBM growth in vivo when administered systemically, both in subcutaneous and intracranial xenografts, reducing the vascularization of the tumors, highlighting a potential relationship between the function of N-MYC and the expression of mesenchymal/angiogenic genes. Overall, our results strengthen the view of N-MYC as a therapeutic target in GBM and strongly suggest that IDP-410 could be further developed to become a first-in-class inhibitor of N-MYC protein, affecting not only tumor cell proliferation and survival, but also the interplay between GBM cells and their microenvironment.
Collapse
Affiliation(s)
- Ricardo Gargini
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain.
| | | | | | - Rafael Hortigüela
- Neurooncology Unit, Instituto de Salud Carlos III-UFIEC, Madrid, Spain
- Centro de Investigaciones, Biológicas Margarita Salas-CSIC, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Orsini EM, Perelas A, Southern BD, Grove LM, Olman MA, Scheraga RG. Stretching the Function of Innate Immune Cells. Front Immunol 2021; 12:767319. [PMID: 34795674 PMCID: PMC8593101 DOI: 10.3389/fimmu.2021.767319] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022] Open
Abstract
The importance of innate immune cells to sense and respond to their physical environment is becoming increasingly recognized. Innate immune cells (e.g. macrophages and neutrophils) are able to receive mechanical signals through several mechanisms. In this review, we discuss the role of mechanosensitive ion channels, such as Piezo1 and transient receptor potential vanilloid 4 (TRPV4), and cell adhesion molecules, such as integrins, selectins, and cadherins in biology and human disease. Furthermore, we explain that these mechanical stimuli activate intracellular signaling pathways, such as MAPK (p38, JNK), YAP/TAZ, EDN1, NF-kB, and HIF-1α, to induce protein conformation changes and modulate gene expression to drive cellular function. Understanding the mechanisms by which immune cells interpret mechanosensitive information presents potential targets to treat human disease. Important areas of future study in this area include autoimmune, allergic, infectious, and malignant conditions.
Collapse
Affiliation(s)
- Erica M Orsini
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Apostolos Perelas
- Department of Pulmonary and Critical Care, Virginia Commonwealth University, Richmond, VA, United States
| | - Brian D Southern
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Lisa M Grove
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Mitchell A Olman
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Rachel G Scheraga
- Respiratory Institute, Cleveland Clinic, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
48
|
Goswami R, Arya RK, Sharma S, Dutta B, Stamov DR, Zhu X, Rahaman SO. Mechanosensing by TRPV4 mediates stiffness-induced foreign body response and giant cell formation. Sci Signal 2021; 14:eabd4077. [PMID: 34726952 PMCID: PMC9976933 DOI: 10.1126/scisignal.abd4077] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Implantation of biomaterials or devices into soft tissue often leads to the development of the foreign body response (FBR), an inflammatory condition that can cause implant failure, tissue injury, and death of the patient. Macrophages accumulate and fuse to generate destructive foreign body giant cells (FBGCs) at the tissue-implant interface, leading to the development of fibrous scar tissue around the implant that is generated by myofibroblasts. We previously showed that the FBR in vivo and FBGC formation in vitro require transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel. Here, we report that TRPV4 was required specifically for the FBR induced by implant stiffness independently of biochemical cues and for intracellular stiffening that promotes FBGC formation in vitro. TRPV4 deficiency reduced collagen deposition and the accumulation of macrophages, FBGCs, and myofibroblasts at stiff, but not soft, implants in vivo and inhibited macrophage-induced differentiation of wild-type fibroblasts into myofibroblasts in vitro. Atomic force microscopy demonstrated that TRPV4 was required for implant-adjacent tissue stiffening in vivo and for cytoskeletal remodeling and intracellular stiffening induced by fusogenic cytokines in vitro. Together, these data suggest a mechanism whereby a reciprocal functional interaction between TRPV4 and substrate stiffness leads to cytoskeletal remodeling and cellular force generation to promote FBGC formation during the FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Rakesh K. Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Dimitar R. Stamov
- JPK BioAFM Business, Nano Surfaces Division, Bruker Nano GmbH, Am Studio 2D, 12489 Berlin, Germany
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.,Corresponding author.:
| |
Collapse
|
49
|
Canales Coutiño B, Mayor R. Reprint of: Mechanosensitive ion channels in cell migration. Cells Dev 2021; 168:203730. [PMID: 34456177 DOI: 10.1016/j.cdev.2021.203730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/18/2021] [Accepted: 04/19/2021] [Indexed: 11/18/2022]
Abstract
Cellular processes are initiated and regulated by different stimuli, including mechanical forces. Cell membrane mechanosensors represent the first step towards the conversion of mechanical stimuli to a biochemical or electrical response. Mechanosensitive (MS) ion channels form a growing family of ion gating channels that respond to direct physical force or plasma membrane deformations. A number of calcium (Ca2+) permeable MS channels are known to regulate the initiation, direction, and persistence of cell migration during development and tumour progression. While the evidence that links individual MS ion channels to cell migration is growing, a unified analysis of the molecular mechanisms regulated downstream of MS ion channel activation is lacking. In this review, we describe the MS ion channel families known to regulate cell migration. We discuss the molecular mechanisms that act downstream of MS ion channels with an emphasis on Ca2+ mediated processes. Finally, we propose the future directions and impact of MS ion channel activity in the field of cell migration.
Collapse
Affiliation(s)
- Brenda Canales Coutiño
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Roberto Mayor
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
50
|
Transient Receptor Potential Channels in the Epithelial-to-Mesenchymal Transition. Int J Mol Sci 2021; 22:ijms22158188. [PMID: 34360952 PMCID: PMC8348042 DOI: 10.3390/ijms22158188] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/19/2021] [Accepted: 07/27/2021] [Indexed: 12/14/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a strictly regulated process that is indispensable for normal development, but it can result in fibrosis and cancer progression. It encompasses a complete alteration of the cellular transcriptomic profile, promoting the expression of genes involved in cellular migration, invasion and proliferation. Extracellular signaling factors driving the EMT process require secondary messengers to convey their effects to their targets. Due to its remarkable properties, calcium represents an ideal candidate to translate molecular messages from receptor to effector. Therefore, calcium-permeable ion channels that facilitate the influx of extracellular calcium into the cytosol can exert major influences on cellular phenotype. Transient receptor potential (TRP) channels represent a superfamily of non-selective cation channels that decode physical and chemical stimuli into cellular behavior. Their role as cellular sensors renders them interesting proteins to study in the context of phenotypic transitions, such as EMT. In this review, we elaborate on the current knowledge regarding TRP channel expression and activity in cellular phenotype and EMT.
Collapse
|