1
|
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
|
2
|
Lacroix JJ, Wijerathne TD. PIEZO channels as multimodal mechanotransducers. Biochem Soc Trans 2025; 53:BST20240419. [PMID: 39936392 DOI: 10.1042/bst20240419] [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: 10/30/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
All living beings experience a wide range of endogenous and exogenous mechanical forces. The ability to detect these forces and rapidly convert them into specific biological signals is essential to a wide range of physiological processes. In vertebrates, these fundamental tasks are predominantly achieved by two related mechanosensitive ion channels called PIEZO1 and PIEZO2. PIEZO channels are thought to sense mechanical forces through flexible transmembrane blade-like domains. Structural studies indeed show that these mechanosensory domains adopt a curved conformation in a resting membrane but become flattened in a membrane under tension, promoting an open state. Yet, recent studies suggest the intriguing possibility that distinct mechanical stimuli activate PIEZO channels through discrete molecular rearrangements of these domains. In addition, biological signals downstream of PIEZO channel activation vary as a function of the mechanical stimulus and of the cellular context. These unique features could explain how PIEZOs confer cells the ability to differentially interpret a complex landscape of mechanical cues.
Collapse
Affiliation(s)
- Jérôme J Lacroix
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| | - Tharaka D Wijerathne
- Department of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91766, U.S.A
| |
Collapse
|
3
|
Goel VK, Barui AK, Nauli SM. Measuring Calcium Signaling at the Primary Cilia. Methods Mol Biol 2025; 2861:129-140. [PMID: 39395102 DOI: 10.1007/978-1-0716-4164-4_10] [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: 10/14/2024]
Abstract
Cellular signaling is nature's ingenious way for cells to perceive their surroundings and transmit external cues to internal compartments. Due to its critical role in cellular functions, the intricate machinery of molecular signaling has been intensively studied. A diverse arsenal of techniques exists to quantify the molecules involved in these processes. Among them, calcium stands out as a ubiquitous signaling molecule with roles in countless biological pathways. To elucidate its function as a second messenger, methods for measuring intracellular calcium have steadily evolved. This chapter introduces various methods for investigating calcium signaling cascades in cells as well as in cilia (thin hairlike projections) specifically, where calcium signaling is triggered by different cilial manipulation techniques.
Collapse
Affiliation(s)
- Vansh K Goel
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
| | - Ayan K Barui
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of California Irvine, Irvine, CA, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, USA.
- Department of Medicine, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Fakhari S, Campolina‐Silva G, Asayesh F, Girardet L, Scott‐Boyer M, Droit A, Soulet D, Greener J, Belleannée C. Shear stress effects on epididymal epithelial cell via primary cilia mechanosensory signaling. J Cell Physiol 2025; 240:e31475. [PMID: 39508588 PMCID: PMC11733861 DOI: 10.1002/jcp.31475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/24/2024] [Accepted: 10/17/2024] [Indexed: 11/15/2024]
Abstract
Shear stress, resulting from fluid flow, is a fundamental mechanical stimulus affecting various cellular functions. The epididymis, essential for sperm maturation, offers a compelling model to study the effects of shear stress on cellular behavior. This organ undergoes extensive proliferation and differentiation until puberty, achieving full functionality as spermatozoa commence their post-testicular maturation. Although the mechanical tension exerted by testicular fluid is hypothesized to drive epithelial proliferation and differentiation, the precise mechanisms remain unclear. Here we assessed whether the responsiveness of the epididymal cells to shear stress depends on functional primary cilia by combining microfluidic strategies on immortalized epididymal cells, calcium signaling assays, and high-throughput gene expression analysis. We identified 97 genes overexpressed in response to shear stress, including early growth response (Egr) 2/3, cellular communication network factor (Ccn) 1/2, and Fos proto-oncogene (Fos). While shear stress triggered a rapid increase of intracellular Ca2+, this response was abrogated following the impairment of primary ciliogenesis through pharmacological and siRNA approaches. Overall, our findings provide valuable insights into how mechanical forces influence the development of the male reproductive system, a requisite to sperm maturation.
Collapse
Affiliation(s)
- Sepideh Fakhari
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Gabriel Campolina‐Silva
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Farnaz Asayesh
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
| | - Laura Girardet
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| | - Marie‐Pier Scott‐Boyer
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Arnaud Droit
- Proteomics Platform, Québec Genomic Center, Université Laval, CHU de Québec Research Center (CHUL)Québec CityQuebecCanada
| | - Denis Soulet
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Faculté de pharmacieUniversité LavalQuébec CityQuebecCanada
| | - Jesse Greener
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
- Department of ChemistryFaculty of Science and EngineeringQuébec CityQuebecCanada
| | - Clémence Belleannée
- Department of Obstetrics, Gynecology, and Reproduction, Centre de recherche en Reproduction, Développement et Santé IntergénérationnelleFaculty of MedicineQuébec CityQuebecCanada
- Centre de recherche du centre hospitalier universitaire de Québec ‐ Université LavalQuébec CityQuebecCanada
| |
Collapse
|
5
|
Ding D, Tian R, Yang X, Ren Z, Jing ZC, Wu XT, Sun LW. The impact of ciliary length on the mechanical response of osteocytes to fluid shear stress. Nitric Oxide 2024; 155:1-11. [PMID: 39734011 DOI: 10.1016/j.niox.2024.12.003] [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: 11/19/2024] [Revised: 12/15/2024] [Accepted: 12/27/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Osteocytes are crucial for detecting mechanical stimuli and translating them into biochemical responses within the bone. The primary cilium, a cellular 'antenna,' plays a vital role in this process. However, there is a lack of direct correlation between cilium length changes and osteocyte mechanosensitivity changes. This study aims to reveal the relationship between ciliary length and nitric oxide (NO) release in osteocytes to show how primary cilia may be involved in reducing osteocyte mechanosensitivity caused by microgravity. MATERIALS AND METHODS We used the MLO-Y4 cell line and primary osteoblasts to adjust the ciliary length using chloral hydrate (CH) for shortening and lithium ions (Li+) for elongation. We then examined the impact of varied ciliary lengths on osteocyte response to fluid shear stress, focusing on the PC1/PC2-Ca2+-NO signaling pathway. Co-culture systems assessed downstream effects on osteoblast function, including collagen secretion and mineralization. RESULTS We observed a significant correlation between ciliary length and osteocyte mechanosensitivity, with longer primary cilia enhancing Ca2+ influx and NO release in response to fluid shear stress. However, contrary to expectations, calmodulin (CaM) expression did not increase with ciliary length, suggesting alternative pathways, such as PKC or Akt/PKB, may modulate p-eNOS activity. Co-cultured osteoblasts showed altered osteogenic functions regulated by osteocyte-derived signals influenced by primary cilia length. CONCLUSION Our findings clarify the role of primary cilia length in modulating osteocyte mechanosensitivity and their influence on osteoblast function, highlighting a complex regulatory network that may not solely rely on CaM for NO release. These insights contribute to a deeper understanding of bone mechanotransduction and could have implications for developing therapeutic targets for osteocyte-related disorders.
Collapse
Affiliation(s)
- Dong Ding
- National Infrastructures for Translational Medicine, Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China; Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Ran Tian
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xiao Yang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Zhe Ren
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China; Hospital of Beihang University, Beijing, 100191, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital Guangdong Academy of Medical Sciences, Southern Medical University. Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xin-Tong Wu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| | - Lian-Wen Sun
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology, National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering), School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
6
|
Hernández-Vega AM, García-Villegas R, Rosenbaum T. Roles for TRPV4 in disease: A discussion of possible mechanisms. Cell Calcium 2024; 124:102972. [PMID: 39609180 DOI: 10.1016/j.ceca.2024.102972] [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: 08/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
The transient receptor potential vanilloid 4 (TRPV4) ion channel is a ubiquitously expressed Ca2+-permeable ion channel that controls intracellular calcium ([Ca2+]i) homeostasis in various types of cells. The physiological roles for TRPV4 are tissue specific and the mechanisms behind this specificity remain mostly unclarified. It is noteworthy that mutations in the TRPV4 channel have been associated to a broad spectrum of congenital diseases, with most of these mutations mainly resulting in gain-of-function. Mutations have been identified in human patients showing a variety of phenotypes and symptoms, mostly related to skeletal and neuromuscular disorders. Since TRPV4 is so widely expressed throughout the body, it comes as no surprise that the literature is growing in evidence linking this protein to malfunction in systems other than the skeletal and neuromuscular. In this review, we summarize the expression patterns of TRPV4 in several tissues and highlight findings of recent studies that address critical structural and functional features of this channel, particularly focusing on its interactions and signaling pathways related to Ca2+ entry. Moreover, we discuss the roles of TRPV4 mutations in some diseases and pinpoint some of the mechanisms underlying pathological states where TRPV4's malfunction is prominent.
Collapse
Affiliation(s)
- Ana M Hernández-Vega
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Refugio García-Villegas
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Av. Instituto Politécnico Nacional 2508, Ciudad de México, 07360, México
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
7
|
Moore ER, Konermann A. Investigating the Role of Primary Cilia and Bone Morphogenetic Protein Signaling in Periodontal Ligament Response to Orthodontic Strain In Vivo and In Vitro: A Pilot Study. Int J Mol Sci 2024; 25:12648. [PMID: 39684361 DOI: 10.3390/ijms252312648] [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: 10/09/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Periodontal ligament (PDL) cells are crucial for mechanosensation and mechanotransduction within the PDL, yet the role of primary cilia in orthodontic force transmission has not been examined. While bone morphogenetic protein (BMP) signaling significantly influences ciliary function, its effect on cellular responses to mechanical stress has not been investigated. This study aims to investigate whether primary cilia and BMP signaling are involved in the periodontal ligament's response to orthodontic tooth movement and the resultant mechanical strain. To visualize primary cilia, human PDL cells were cultured on glass-bottom dishes for five days, with a subset fixed daily, followed by immunostaining with anti-acetylated α-tubulin and Alexa Fluor 568 and imaging using a fluorescence microscope under 405 nm and 561 nm laser excitation. Human PDL cells were grown on Bioflex® culture plates and subsequently exposed to static tensile strains of 2.5%, 5%, 10%, 20%, on a FX-6000T™ Tension System for 24 h. RT-qPCR was performed to evaluate changes in expression of primary cilia via Ift88 expression, mechanotransduction via Cox2 expression, and BMP signaling-related genes. Histological specimens from orthodontically loaded and control human premolars were investigated for primary cilia and BMP signaling using immunohistochemistry and confocal microscopy. Primary cilia were observed in PDL cells from day one, with their incidence and length increasing over time alongside cell density. BMP signaling components, including upregulated genes such as Bmp7 (10.99-14.97 fold), Alk2 (3.19-5.45 fold), and Bmpr2 (1.64-8.40 fold), consistently responded to strain, while Cox2 and Ift88 showed differential regulation depending on strain intensity. In vivo, orthodontic movement activated BMP signaling and increased primary cilium incidence in the PDL. These findings indicate the potential role of primary cilia and BMP signaling in the mechanosensitivity of PDL cells under orthodontic forces. Further studies are required to understand the complex mechanotransduction mechanisms and role of these components in cellular adaptation during orthodontic tooth movement.
Collapse
Affiliation(s)
- Emily R Moore
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Anna Konermann
- Department of Orthodontics, University of Bonn, 53111 Bonn, Germany
| |
Collapse
|
8
|
Yang C, Cai W, Xiang P, Liu Y, Xu H, Zhang W, Han F, Luo Z, Liang T. Viscoelastic hydrogel combined with dynamic compression promotes osteogenic differentiation of bone marrow mesenchymal stem cells and bone repair in rats. Regen Biomater 2024; 12:rbae136. [PMID: 39845143 PMCID: PMC11751691 DOI: 10.1093/rb/rbae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 01/24/2025] Open
Abstract
A biomechanical environment constructed exploiting the mechanical property of the extracellular matrix and external loading is essential for cell behaviour. Building suitable mechanical stimuli using feasible scaffold material and moderate mechanical loading is critical in bone tissue engineering for bone repair. However, the detailed mechanism of the mechanical regulation remains ambiguous. In addition, TRPV4 is involved in bone development. Therefore, this study aims to construct a viscoelastic hydrogel combined with dynamic compressive loading and investigate the effect of the dynamic mechanical environment on the osteogenic differentiation of stem cells and bone repair in vivo. The role of TRPV4 in the mechanobiology process was also assessed. A sodium alginate-gelatine hydrogel with adjustable viscoelasticity and good cell adhesion ability was obtained. The osteogenic differentiation of BMSCs was obtained using the fast stress relaxation hydrogel and a smaller compression strain of 1.5%. TRPV4 was activated in the hydrogel with fast stress relaxation time, followed by the increase in intracellular Ca2+ level and the activation of the Wnt/β-catenin pathway. The inhibition of TRPV4 induced a decrease in the intracellular Ca2+ level, down-regulation of β-catenin and reduced osteogenesis differentiation of BMSCs, suggesting that TRPV4 might be the key mechanism in the regulation of BMSC osteogenic differentiation in the viscoelastic dynamic mechanical environment. The fast stress relaxation hydrogel also showed a good osteogenic promotion effect in the rat femoral defect model. The dynamic viscoelastic mechanical environment significantly induced the osteogenic differentiation of BMSCs and bone regeneration, which TRPV4 being involved in this mechanobiological process. Our study not only provided important guidance for the mechanical design of new biomaterials, but also provided a new perspective for the understanding of the interaction between cells and materials, the role of mechanical loading in tissue regeneration and the use of mechanical regulation in tissue engineering.
Collapse
Affiliation(s)
- Chao Yang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Wenbin Cai
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Pan Xiang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Yu Liu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Hao Xu
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Wen Zhang
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Fengxuan Han
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Zongping Luo
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, PR China
| | - Ting Liang
- Medical 3D Printing Center, Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu 215000, PR China
| |
Collapse
|
9
|
Fan Q, Hadla M, Peterson Z, Nelson G, Ye H, Wang X, Mardirossian JM, Harris PC, Alper SL, Prakash YS, Beyder A, Torres VE, Chebib FT. Activation of PIEZO1 Attenuates Kidney Cystogenesis In Vitro and Ex Vivo. KIDNEY360 2024; 5:1601-1612. [PMID: 39356563 DOI: 10.34067/kid.0000000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Key Points
PIEZO1 activation reduces cystogenesis: Yoda1 activates PIEZO1, raising calcium and lowering cAMP, reducing cyst growth in autosomal dominant polycystic kidney disease models.Context-dependent role of PIEZO1: PIEZO1 knockout in mice with or without polycystic kidneys does not affect cyst formation, suggesting redundancy in mechanosensitive pathways.Therapeutic potential: Findings support PIEZO1 activation as a part of combination therapy to slow cyst growth in autosomal dominant polycystic kidney disease, needing more in vivo studies.
Background
The disruption of calcium signaling associated with polycystin deficiency is a key factor in abnormal epithelial growth in autosomal dominant polycystic kidney disease. Calcium homeostasis can be influenced by mechanotransduction. The mechanosensitive cation channel PIEZO1 has been implicated in sensing intrarenal pressure and regulating urinary osmoregulation, but its role in kidney cystogenesis is unclear.
Methods
We hypothesized that altered mechanotransduction contributes to cystogenesis in autosomal dominant polycystic kidney disease and that activation of mechanosensitive cation channels could be a therapeutic strategy.
Results
We demonstrate that Yoda1, a PIEZO1 activator, increases intracellular calcium and reduces forskolin-induced cAMP levels in mouse inner medullary collecting duct (mIMCD3) cells. Notably, knockout of polycystin-2 attenuated the efficacy of Yoda1 in reducing cAMP levels in mIMCD3 cells. Yoda1 also reduced forskolin-induced mIMCD3 cyst surface area in vitro and cystic index in mouse metanephros ex vivo in a dose-dependent manner. However, collecting duct–specific PIEZO1 knockout neither induced cystogenesis in wild-type mice nor altered cystogenesis in the Pkd1RC/RC mouse model.
Conclusions
These findings support the potential role of PIEZO1 agonists in mitigating cystogenesis by increasing intracellular calcium and reducing cAMP levels, but the unaltered in vivo cystic phenotype after PIEZO1 knockout in the collecting duct suggests possible redundancy in mechanotransductive pathways.
Collapse
Affiliation(s)
- Qingfeng Fan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mohamad Hadla
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Zack Peterson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Grace Nelson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hong Ye
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular biology, Mayo Clinic, Rochester, Minnesota
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
10
|
Jahn J, Ehlen QT, Kaplan L, Best TM, Meng Z, Huang CY. Interplay of Glucose Metabolism and Hippo Pathway in Chondrocytes: Pathophysiology and Therapeutic Targets. Bioengineering (Basel) 2024; 11:972. [PMID: 39451348 PMCID: PMC11505586 DOI: 10.3390/bioengineering11100972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
In this review, we explore the intricate relationship between glucose metabolism and mechanotransduction pathways, with a specific focus on the role of the Hippo signaling pathway in chondrocyte pathophysiology. Glucose metabolism is a vital element in maintaining proper chondrocyte function, but it has also been implicated in the pathogenesis of osteoarthritis (OA) via the induction of pro-inflammatory signaling pathways and the establishment of an intracellular environment conducive to OA. Alternatively, mechanotransduction pathways such as the Hippo pathway possess the capacity to respond to mechanical stimuli and have an integral role in maintaining chondrocyte homeostasis. However, these mechanotransduction pathways can be dysregulated and potentially contribute to the progression of OA. We discussed how alterations in glucose levels may modulate the Hippo pathway components via a variety of mechanisms. Characterizing the interaction between glucose metabolism and the Hippo pathway highlights the necessity of balancing both metabolic and mechanical signaling to maintain chondrocyte health and optimal functionality. Furthermore, this review demonstrates the scarcity of the literature on the relationship between glucose metabolism and mechanotransduction and provides a summary of current research dedicated to this specific area of study. Ultimately, increased research into this topic may elucidate novel mechanisms and relationships integrating mechanotransduction and glucose metabolism. Through this review we hope to inspire future research into this topic to develop innovative treatments for addressing the clinical challenges of OA.
Collapse
Affiliation(s)
- Jacob Jahn
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
| | - Lee Kaplan
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Thomas M. Best
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Orthopedics, University of Miami, Miami, FL 33136, USA
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
| | - Zhipeng Meng
- University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.J.); (Q.T.E.); (L.K.); (T.M.B.); (Z.M.)
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chun-Yuh Huang
- UHealth Sports Medicine Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
11
|
Cai J, Deng Y, Min Z, Li C, Zhao Z, Jing D. Deciphering the dynamics: Exploring the impact of mechanical forces on histone acetylation. FASEB J 2024; 38:e23849. [PMID: 39096133 DOI: 10.1096/fj.202400907rr] [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: 04/22/2024] [Revised: 07/01/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Abstract
Living cells navigate a complex landscape of mechanical cues that influence their behavior and fate, originating from both internal and external sources. At the molecular level, the translation of these physical stimuli into cellular responses relies on the intricate coordination of mechanosensors and transducers, ultimately impacting chromatin compaction and gene expression. Notably, epigenetic modifications on histone tails govern the accessibility of gene-regulatory sites, thereby regulating gene expression. Among these modifications, histone acetylation emerges as particularly responsive to the mechanical microenvironment, exerting significant control over cellular activities. However, the precise role of histone acetylation in mechanosensing and transduction remains elusive due to the complexity of the acetylation network. To address this gap, our aim is to systematically explore the key regulators of histone acetylation and their multifaceted roles in response to biomechanical stimuli. In this review, we initially introduce the ubiquitous force experienced by cells and then explore the dynamic alterations in histone acetylation and its associated co-factors, including HDACs, HATs, and acetyl-CoA, in response to these biomechanical cues. Furthermore, we delve into the intricate interactions between histone acetylation and mechanosensors/mechanotransducers, offering a comprehensive analysis. Ultimately, this review aims to provide a holistic understanding of the nuanced interplay between histone acetylation and mechanical forces within an academic framework.
Collapse
Affiliation(s)
- Jingyi Cai
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yudi Deng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ziyang Min
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chaoyuan Li
- Department of Implantology, School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dian Jing
- Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
- National Center for Stomatology, Shanghai, China
- National Clinical Research Center for Oral Diseases, Shanghai, China
- Shanghai Key Laboratory of Stomatology, Shanghai, China
| |
Collapse
|
12
|
Tiberio F, Coda ARD, Tosi DD, Luzi D, Polito L, Liso A, Lattanzi W. Mechanobiology and Primary Cilium in the Pathophysiology of Bone Marrow Myeloproliferative Diseases. Int J Mol Sci 2024; 25:8860. [PMID: 39201546 PMCID: PMC11354938 DOI: 10.3390/ijms25168860] [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: 06/21/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
Philadelphia-Negative Myeloproliferative neoplasms (MPNs) are a diverse group of blood cancers leading to excessive production of mature blood cells. These chronic diseases, including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), can significantly impact patient quality of life and are still incurable in the vast majority of the cases. This review examines the mechanobiology within a bone marrow niche, emphasizing the role of mechanical cues and the primary cilium in the pathophysiology of MPNs. It discusses the influence of extracellular matrix components, cell-cell and cell-matrix interactions, and mechanosensitive structures on hematopoietic stem cell (HSC) behavior and disease progression. Additionally, the potential implications of the primary cilium as a chemo- and mechanosensory organelle in bone marrow cells are explored, highlighting its involvement in signaling pathways crucial for hematopoietic regulation. This review proposes future research directions to better understand the dysregulated bone marrow niche in MPNs and to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Federica Tiberio
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Domiziano Dario Tosi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Debora Luzi
- S.C. Oncoematologia, Azienda Ospedaliera di Terni, 05100 Terni, Italy;
| | - Luca Polito
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
| | - Arcangelo Liso
- Department of Medicine and Surgery, University of Perugia, 06129 Perugia, Italy
| | - Wanda Lattanzi
- Department of Life Science and Public Health, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.T.); (D.D.T.); (L.P.)
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
13
|
Wu S, Zhou H, Ling H, Sun Y, Luo Z, Ngo T, Fu Y, Wang W, Kong Y. LIPUS regulates the progression of knee osteoarthritis in mice through primary cilia-mediated TRPV4 channels. Apoptosis 2024; 29:785-798. [PMID: 38517601 PMCID: PMC11055729 DOI: 10.1007/s10495-024-01950-9] [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] [Accepted: 02/24/2024] [Indexed: 03/24/2024]
Abstract
Osteoarthritis (OA) is a common disease in middle-aged and elderly people. An imbalance in calcium ion homeostasis will contribute to chondrocyte apoptosis and ultimately lead to the progression of OA. Transient receptor potential channel 4 (TRPV4) is involved in the regulation of intracellular calcium homeostasis. TRPV4 is expressed in primary cilia, which can sense mechanical stimuli from outside the cell, and its abnormal expression is closely related to the development of OA. Low-intensity pulsed ultrasound (LIPUS) can alleviate chondrocyte apoptosis while the exact mechanism is unclear. In this project, with the aim of revealing the mechanism of action of LIPUS, we proposed to use OA chondrocytes and animal models, LIPUS intervention, inhibition of primary cilia, use TRPV4 inhibitors or TRPV4 agonist, and use Immunofluorescence (IF), Immunohistochemistry (IHC), Western Blot (WB), Quantitative Real-time PCR (QP) to detect the expression of cartilage synthetic matrix and endoplasmic reticulum stress markers. The results revealed that LIPUS altered primary cilia expression, promoted synthetic matrix metabolism in articular chondrocytes and was associated with primary cilia. In addition, LIPUS exerted a active effect on OA by activating TRPV4, inducing calcium inward flow, and facilitating the entry of NF-κB into the nucleus to regulate synthetic matrix gene transcription. Inhibition of TRPV4 altered primary cilia expression in response to LIPUS stimulation, and knockdown of primary cilia similarly inhibited TRPV4 function. These results suggest that LIPUS mediates TRPV4 channels through primary cilia to regulate the process of knee osteoarthritis in mice.
Collapse
Affiliation(s)
- Sha Wu
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Haiqi Zhou
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Huixian Ling
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuyan Sun
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ziyu Luo
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - ThaiNamanh Ngo
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yuanyuan Fu
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Wen Wang
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
14
|
Fan Q, Hadla M, Peterson Z, Nelson G, Ye H, Wang X, Mardirossian JM, Harris PC, Alper SL, Prakash YS, Beyder A, Torres VE, Chebib FT. Activation of Piezo1 Inhibits Kidney Cystogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593717. [PMID: 38766249 PMCID: PMC11101129 DOI: 10.1101/2024.05.11.593717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The disruption of calcium signaling associated with polycystin deficiency has been proposed as the primary event underlying the increased abnormally patterned epithelial cell growth characteristic of Polycystic Kidney Disease. Calcium can be regulated through mechanotransduction, and the mechanosensitive cation channel Piezo1 has been implicated in sensing of intrarenal pressure and in urinary osmoregulation. However, a possible role for PIEZO1 in kidney cystogenesis remains undefined. We hypothesized that cystogenesis in ADPKD reflects altered mechanotransduction, suggesting activation of mechanosensitive cation channels as a therapeutic strategy for ADPKD. Here, we show that Yoda-1 activation of PIEZO1 increases intracellular Ca 2+ and reduces forskolin-induced cAMP levels in mIMCD3 cells. Yoda-1 reduced forskolin-induced IMCD cyst surface area in vitro and in mouse metanephros ex vivo in a dose-dependent manner. Knockout of polycystin-2 dampened the efficacy of PIEZO1 activation in reducing both cAMP levels and cyst surface area in IMCD3 cells. However, collecting duct-specific Piezo1 knockout neither induced cystogenesis in wild-type mice nor affected cystogenesis in the Pkd1 RC/RC model of ADPKD. Our study suggests that polycystin-2 and PIEZO1 play a role in mechanotransduction during cystogenesis in vitro , and ex vivo , but that in vivo cyst expansion may require inactivation or repression of additional suppressors of cystogenesis and/or growth. Our study provides a preliminary proof of concept for PIEZO1 activation as a possible component of combination chemotherapy to retard or halt cystogenesis and/or cyst growth.
Collapse
|
15
|
Mascharak S, Guo JL, Griffin M, Berry CE, Wan DC, Longaker MT. Modelling and targeting mechanical forces in organ fibrosis. NATURE REVIEWS BIOENGINEERING 2024; 2:305-323. [PMID: 39552705 PMCID: PMC11567675 DOI: 10.1038/s44222-023-00144-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 11/19/2024]
Abstract
Few efficacious therapies exist for the treatment of fibrotic diseases, such as skin scarring, liver cirrhosis and pulmonary fibrosis, which is related to our limited understanding of the fundamental causes and mechanisms of fibrosis. Mechanical forces from cell-matrix interactions, cell-cell contact, fluid flow and other physical stimuli may play a central role in the initiation and propagation of fibrosis. In this Review, we highlight the mechanotransduction mechanisms by which various sources of physical force drive fibrotic disease processes, with an emphasis on central pathways that may be therapeutically targeted to prevent and reverse fibrosis. We then discuss engineered models of mechanotransduction in fibrosis, as well as molecular and biomaterials-based therapeutic approaches for limiting fibrosis and promoting regenerative healing phenotypes in various organs. Finally, we discuss challenges within fibrosis research that remain to be addressed and that may greatly benefit from next-generation bioengineered model systems.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Jason L. Guo
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Shamik Mascharak, Jason L. Guo, Michelle Griffin
| | - Charlotte E. Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Derrick C. Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael T. Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
16
|
Buck HV, Stains JP. Osteocyte-mediated mechanical response controls osteoblast differentiation and function. Front Physiol 2024; 15:1364694. [PMID: 38529481 PMCID: PMC10961341 DOI: 10.3389/fphys.2024.1364694] [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: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Low bone mass is a pervasive global health concern, with implications for osteoporosis, frailty, disability, and mortality. Lifestyle factors, including sedentary habits, metabolic dysfunction, and an aging population, contribute to the escalating prevalence of osteopenia and osteoporosis. The application of mechanical load to bone through physical activity and exercise prevents bone loss, while sufficient mechanical load stimulates new bone mass acquisition. Osteocytes, cells embedded within the bone, receive mechanical signals and translate these mechanical cues into biological signals, termed mechano-transduction. Mechano-transduction signals regulate other bone resident cells, such as osteoblasts and osteoclasts, to orchestrate changes in bone mass. This review explores the mechanisms through which osteocyte-mediated response to mechanical loading regulates osteoblast differentiation and bone formation. An overview of bone cell biology and the impact of mechanical load will be provided, with emphasis on the mechanical cues, mechano-transduction pathways, and factors that direct progenitor cells toward the osteoblast lineage. While there are a wide range of clinically available treatments for osteoporosis, the majority act through manipulation of the osteoclast and may have significant disadvantages. Despite the central role of osteoblasts to the deposition of new bone, few therapies directly target osteoblasts for the preservation of bone mass. Improved understanding of the mechanisms leading to osteoblastogenesis may reveal novel targets for translational investigation.
Collapse
Affiliation(s)
| | - Joseph Paul Stains
- School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a background on osteocytes and the primary cilium, discussing the role it plays in osteocyte mechanosensing. RECENT FINDINGS Osteocytes are thought to be the primary mechanosensing cells in bone tissue, regulating bone adaptation in response to exercise, with the primary cilium suggested to be a key mechanosensing mechanism in bone. More recent work has suggested that, rather than being direct mechanosensors themselves, primary cilia in bone may instead form a key chemo-signalling nexus for processing mechanoregulated signalling pathways. Recent evidence suggests that pharmacologically induced lengthening of the primary cilium in osteocytes may potentiate greater mechanotransduction, rather than greater mechanosensing. While more research is required to delineate the specific osteocyte mechanobiological molecular mechanisms governed by the primary cilium, it is clear from the literature that the primary cilium has significant potential as a therapeutic target to treat mechanoregulated bone diseases, such as osteoporosis.
Collapse
Affiliation(s)
- Stefaan W Verbruggen
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK.
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK.
- Centre for Predictive in vitro Models, Centre for Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, UK.
| | - Anuphan Sittichokechaiwut
- Department of Preventive Dentistry, Faculty of Dentistry, Naresuan University, Phitsanulok, Thailand
- Center of Excellence in Biomaterials, Naresuan University, Phitsanulok, Thailand
| | - Gwendolen C Reilly
- INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
- Kroto Research Institute, Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| |
Collapse
|
18
|
Mohseni M, Vahidi B, Azizi H. Computational simulation of applying mechanical vibration to mesenchymal stem cell for mechanical modulation toward bone tissue engineering. Proc Inst Mech Eng H 2023; 237:1377-1389. [PMID: 37982187 DOI: 10.1177/09544119231208223] [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] [Indexed: 11/21/2023]
Abstract
Evaluation of cell response to mechanical stimuli at in vitro conditions is known as one of the important issues for modulating cell behavior. Mechanical stimuli, including mechanical vibration and oscillatory fluid flow, act as important biophysical signals for the mechanical modulation of stem cells. In the present study, mesenchymal stem cell (MSC) consists of cytoplasm, nucleus, actin, and microtubule. Also, integrin and primary cilium were considered as mechanoreceptors. In this study, the combined effect of vibration and oscillatory fluid flow on the cell and its components were investigated using numerical modeling. The results of the FEM and FSI model showed that the cell response (stress and strain values) at the frequency of 30 H z mechanical vibration has the highest value. The achieved results on shear stress caused by the fluid flow on the cell showed that the cell experiences shear stress in the range of 0 . 1 - 10 Pa . Mechanoreceptors that bind separately to the cell surface, can be highly stimulated by hydrodynamic pressure and, therefore, can play a role in the mechanical modulation of MSCs at in vitro conditions. The results of this research can be effective in future studies to optimize the conditions of mechanical stimuli applied to the cell culture medium and to determine the mechanisms involved in mechanotransduction.
Collapse
Affiliation(s)
- Mohammadreza Mohseni
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Bahman Vahidi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Hamidreza Azizi
- Division of Biomedical Engineering, Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
19
|
Jia Y, Le H, Wang X, Zhang J, Liu Y, Ding J, Zheng C, Chang F. Double-edged role of mechanical stimuli and underlying mechanisms in cartilage tissue engineering. Front Bioeng Biotechnol 2023; 11:1271762. [PMID: 38053849 PMCID: PMC10694366 DOI: 10.3389/fbioe.2023.1271762] [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: 08/02/2023] [Accepted: 10/11/2023] [Indexed: 12/07/2023] Open
Abstract
Mechanical stimuli regulate the chondrogenic differentiation of mesenchymal stem cells and the homeostasis of chondrocytes, thus affecting implant success in cartilage tissue engineering. The mechanical microenvironment plays fundamental roles in the maturation and maintenance of natural articular cartilage, and the progression of osteoarthritis Hence, cartilage tissue engineering attempts to mimic this environment in vivo to obtain implants that enable a superior regeneration process. However, the specific type of mechanical loading, its optimal regime, and the underlying molecular mechanisms are still under investigation. First, this review delineates the composition and structure of articular cartilage, indicating that the morphology of chondrocytes and components of the extracellular matrix differ from each other to resist forces in three top-to-bottom overlapping zones. Moreover, results from research experiments and clinical trials focusing on the effect of compression, fluid shear stress, hydrostatic pressure, and osmotic pressure are presented and critically evaluated. As a key direction, the latest advances in mechanisms involved in the transduction of external mechanical signals into biological signals are discussed. These mechanical signals are sensed by receptors in the cell membrane, such as primary cilia, integrins, and ion channels, which next activate downstream pathways. Finally, biomaterials with various modifications to mimic the mechanical properties of natural cartilage and the self-designed bioreactors for experiment in vitro are outlined. An improved understanding of biomechanically driven cartilage tissue engineering and the underlying mechanisms is expected to lead to efficient articular cartilage repair for cartilage degeneration and disease.
Collapse
Affiliation(s)
- Yao Jia
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Hanxiang Le
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
- The Fourth Treatment Area of Trauma Hip Joint Surgery Department, Tianjin Hospital, Tianjin, China
| | - Xianggang Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Jiaxin Zhang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Yan Liu
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Jiacheng Ding
- The Second Bethune Clinical Medical College of Jilin University, Jilin, China
| | - Changjun Zheng
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| | - Fei Chang
- Department of Orthopedics, The Second Hospital of Jilin University, Jilin, China
| |
Collapse
|
20
|
LaGuardia JS, Shariati K, Bedar M, Ren X, Moghadam S, Huang KX, Chen W, Kang Y, Yamaguchi DT, Lee JC. Convergence of Calcium Channel Regulation and Mechanotransduction in Skeletal Regenerative Biomaterial Design. Adv Healthc Mater 2023; 12:e2301081. [PMID: 37380172 PMCID: PMC10615747 DOI: 10.1002/adhm.202301081] [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: 04/05/2023] [Revised: 06/20/2023] [Indexed: 06/30/2023]
Abstract
Cells are known to perceive their microenvironment through extracellular and intracellular mechanical signals. Upon sensing mechanical stimuli, cells can initiate various downstream signaling pathways that are vital to regulating proliferation, growth, and homeostasis. One such physiologic activity modulated by mechanical stimuli is osteogenic differentiation. The process of osteogenic mechanotransduction is regulated by numerous calcium ion channels-including channels coupled to cilia, mechanosensitive and voltage-sensitive channels, and channels associated with the endoplasmic reticulum. Evidence suggests these channels are implicated in osteogenic pathways such as the YAP/TAZ and canonical Wnt pathways. This review aims to describe the involvement of calcium channels in regulating osteogenic differentiation in response to mechanical loading and characterize the fashion in which those channels directly or indirectly mediate this process. The mechanotransduction pathway is a promising target for the development of regenerative materials for clinical applications due to its independence from exogenous growth factor supplementation. As such, also described are examples of osteogenic biomaterial strategies that involve the discussed calcium ion channels, calcium-dependent cellular structures, or calcium ion-regulating cellular features. Understanding the distinct ways calcium channels and signaling regulate these processes may uncover potential targets for advancing biomaterials with regenerative osteogenic capabilities.
Collapse
Affiliation(s)
- Jonnby S. LaGuardia
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kaavian Shariati
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Meiwand Bedar
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Xiaoyan Ren
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Shahrzad Moghadam
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Kelly X. Huang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Wei Chen
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Youngnam Kang
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
| | - Dean T. Yamaguchi
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
| | - Justine C. Lee
- Division of Plastic & Reconstructive Surgery, University of California, Los Angeles David Geffen School of Medicine, Los Angeles, CA, 90095, USA
- Research Service, Greater Los Angeles VA Healthcare System, Los Angeles, CA, 91343, USA
- Department of Orthopaedic Surgery, Los Angeles, CA, 90095, USA
- UCLA Molecular Biology Institute, Los Angeles, CA, 90095, USA
| |
Collapse
|
21
|
Quadri N, Upadhyai P. Primary cilia in skeletal development and disease. Exp Cell Res 2023; 431:113751. [PMID: 37574037 DOI: 10.1016/j.yexcr.2023.113751] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023]
Abstract
Primary cilia are non-motile, microtubule-based sensory organelle present in most vertebrate cells with a fundamental role in the modulation of organismal development, morphogenesis, and repair. Here we focus on the role of primary cilia in embryonic and postnatal skeletal development. We examine evidence supporting its involvement in physiochemical and developmental signaling that regulates proliferation, patterning, differentiation and homeostasis of osteoblasts, chondrocytes, and their progenitor cells in the skeleton. We discuss how signaling effectors in mechanotransduction and bone development, such as Hedgehog, Wnt, Fibroblast growth factor and second messenger pathways operate at least in part at the primary cilium. The relevance of primary cilia in bone formation and maintenance is underscored by a growing list of rare genetic skeletal ciliopathies. We collate these findings and summarize the current understanding of molecular factors and mechanisms governing primary ciliogenesis and ciliary function in skeletal development and disease.
Collapse
Affiliation(s)
- Neha Quadri
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
22
|
Zhang Y, Tawiah GK, Wu X, Zhang Y, Wang X, Wei X, Qiao X, Zhang Q. Primary cilium-mediated mechanotransduction in cartilage chondrocytes. Exp Biol Med (Maywood) 2023; 248:1279-1287. [PMID: 37897221 PMCID: PMC10625344 DOI: 10.1177/15353702231199079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Osteoarthritis (OA) is one of the most prevalent joint disorders associated with the degradation of articular cartilage and an abnormal mechanical microenvironment. Mechanical stimuli, including compression, shear stress, stretching strain, osmotic challenge, and the physical properties of the matrix microenvironment, play pivotal roles in the tissue homeostasis of articular cartilage. The primary cilium, as a mechanosensory and chemosensory organelle, is important for detecting and transmitting both mechanical and biochemical signals in chondrocytes within the matrix microenvironment. Growing evidence indicates that primary cilia are critical for chondrocytes signaling transduction and the matrix homeostasis of articular cartilage. Furthermore, the ability of primary cilium to regulate cellular signaling is dynamic and dependent on the cellular matrix microenvironment. In the current review, we aim to elucidate the key mechanisms by which primary cilia mediate chondrocytes sensing and responding to the matrix mechanical microenvironment. This might have potential therapeutic applications in injuries and OA-associated degeneration of articular cartilage.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Godfred K Tawiah
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaoan Wu
- Department of Physiology and Biophysics, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Yanjun Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xiaohong Qiao
- Department of Histology and Embryology, Shanxi Medical University, Jinzhong 030604, Shanxi, China
- Department of Orthopaedics, Lvliang Hospital Affiliated to Shanxi Medical University, Lvliang 033099, Shanxi, China
| | - Quanyou Zhang
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| |
Collapse
|
23
|
Sutton MM, Duffy MP, Verbruggen SW, Jacobs CR. Osteoclastogenesis Requires Primary Cilia Disassembly and Can Be Inhibited by Promoting Primary Cilia Formation Pharmacologically. Cells Tissues Organs 2023; 213:235-244. [PMID: 37231815 PMCID: PMC10863750 DOI: 10.1159/000531098] [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: 04/25/2022] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
The primary cilium is a solitary, sensory organelle with many roles in bone development, maintenance, and function. In the osteogenic cell lineage, including skeletal stem cells, osteoblasts, and osteocytes, the primary cilium plays a vital role in the regulation of bone formation, and this has made it a promising pharmaceutical target to maintain bone health. While the role of the primary cilium in the osteogenic cell lineage has been increasingly characterized, little is known about the potential impact of targeting the cilium in relation to osteoclasts, a hematopoietic cell responsible for bone resorption. The objective of this study was to determine whether osteoclasts have a primary cilium and to investigate whether or not the primary cilium of macrophages, osteoclast precursors, serves a functional role in osteoclast formation. Using immunocytochemistry, we showed the macrophages have a primary cilium, while osteoclasts lack this organelle. Furthermore, we increased macrophage primary cilia incidence and length using fenoldopam mesylate and found that cells undergoing such treatment showed a significant decrease in the expression of osteoclast markers tartrate-resistant acid phosphatase, cathepsin K, and c-Fos, as well as decreased osteoclast formation. This work is the first to show that macrophage primary cilia resorption may be a necessary step for osteoclast differentiation. Since primary cilia and preosteoclasts are responsive to fluid flow, we applied fluid flow at magnitudes present in the bone marrow to differentiating cells and found that osteoclastic gene expression by macrophages was not affected by fluid flow mechanical stimulation, suggesting that the role of the primary cilium in osteoclastogenesis is not a mechanosensory one. The primary cilium has been suggested to play a role in bone formation, and our findings indicate that it may also present a means to regulate bone resorption, presenting a dual benefit of developing ciliary-targeted pharmaceuticals for bone disease.
Collapse
Affiliation(s)
- Michael M. Sutton
- Department of Biomedical Engineering, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, USA
| | - Michael P. Duffy
- Department of Biomedical Engineering, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, USA
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stefaan W. Verbruggen
- Department of Biomedical Engineering, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, USA
- Centre for Predictive in vitro Models, School of Engineering and Materials Science, Queen Mary University of London, London, UK
- Department of Mechanical Engineering and INSIGNEO Institute for in silico Medicine, University of Sheffield, Sheffield, UK
| | - Christopher R. Jacobs
- Department of Biomedical Engineering, Fu Foundation School of Engineering and Applied Science, Columbia University, New York, NY, USA
| |
Collapse
|
24
|
The Impact of Plasma Membrane Ion Channels on Bone Remodeling in Response to Mechanical Stress, Oxidative Imbalance, and Acidosis. Antioxidants (Basel) 2023; 12:antiox12030689. [PMID: 36978936 PMCID: PMC10045377 DOI: 10.3390/antiox12030689] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
The extracellular milieu is a rich source of different stimuli and stressors. Some of them depend on the chemical–physical features of the matrix, while others may come from the ‘outer’ environment, as in the case of mechanical loading applied on the bones. In addition to these forces, a plethora of chemical signals drives cell physiology and fate, possibly leading to dysfunctions when the homeostasis is disrupted. This variety of stimuli triggers different responses among the tissues: bones represent a particular milieu in which a fragile balance between mechanical and metabolic demands should be tuned and maintained by the concerted activity of cell biomolecules located at the interface between external and internal environments. Plasma membrane ion channels can be viewed as multifunctional protein machines that act as rapid and selective dual-nature hubs, sensors, and transducers. Here we focus on some multisensory ion channels (belonging to Piezo, TRP, ASIC/EnaC, P2XR, Connexin, and Pannexin families) actually or potentially playing a significant role in bone adaptation to three main stressors, mechanical forces, oxidative stress, and acidosis, through their effects on bone cells including mesenchymal stem cells, osteoblasts, osteoclasts, and osteocytes. Ion channel-mediated bone remodeling occurs in physiological processes, aging, and human diseases such as osteoporosis, cancer, and traumatic events.
Collapse
|
25
|
Liu P, Liu Y, Zhou J. Ciliary mechanosensation - roles of polycystins and mastigonemes. J Cell Sci 2023; 136:286945. [PMID: 36752106 DOI: 10.1242/jcs.260565] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Cilia are surface-exposed organelles that provide motility and sensory functions for cells, and it is widely believed that mechanosensation can be mediated through cilia. Polycystin-1 and -2 (PC-1 and PC-2, respectively) are transmembrane proteins that can localize to cilia; however, the molecular mechanisms by which polycystins contribute to mechanosensation are still controversial. Studies detail two prevailing models for the molecular roles of polycystins on cilia; one stresses the mechanosensation capabilities and the other unveils their ligand-receptor nature. The discovery that polycystins interact with mastigonemes, the 'hair-like' protrusions of flagella, is a novel finding in identifying the interactors of polycystins in cilia. While the functions of polycystins proposed by both models may coexist in cilia, it is hoped that a precise understanding of the mechanism of action of polycystins can be achieved by uncovering their distribution and interacting factors inside cilia. This will hopefully provide a satisfying answer to the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD), which is caused by mutations in PC-1 and PC-2. In this Review, we discuss the characteristics of polycystins in the context of cilia and summarize the functions of mastigonemes in unicellular ciliates. Finally, we compare flagella and molecular features of PC-2 between unicellular and multicellular organisms, with the aim of providing new insights into the ciliary roles of polycystins in general.
Collapse
Affiliation(s)
- Peiwei Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China
| | - Ying Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology , College of Life Sciences in Shandong Normal University, Jinan 250358, China.,College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
27
|
Meslier QA, DiMauro N, Somanchi P, Nano S, Shefelbine SJ. Manipulating load-induced fluid flow in vivo to promote bone adaptation. Bone 2022; 165:116547. [PMID: 36113842 DOI: 10.1016/j.bone.2022.116547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/01/2022] [Accepted: 09/12/2022] [Indexed: 11/02/2022]
Abstract
Mechanical stimulation is critical to maintaining bone mass and strength. Strain has been commonly thought of as the mechanical stimulus driving bone adaptation. However, numerous studies have hypothesized that fluid flow in the lacunar-canalicular system plays a role in mechanoadaptation. The role of fluid flow compared to strain magnitude on bone remodeling has yet to be characterized. This study aimed to determine the contribution of fluid flow velocity compared to strain on bone adaptation. We used finite element modeling to design in vivo experiments, manipulating strain and fluid flow contributions. Using a uniaxial compression tibia model in mice, we demonstrated that high fluid flow velocity results in significant bone adaptation even under low strain magnitude. In contrast, high strain magnitude paired with low fluid velocity does not trigger a bone response. These findings support previous hypotheses stating that fluid flow is the principal mechanical stimulus driving bone adaptation. Moreover, they give new insights regarding bone adaptative response and provide new pathways toward treatment against age-related mechanosensitivity loss in bone.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Nicole DiMauro
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Priya Somanchi
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Sarah Nano
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA, USA; Department of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
28
|
Melena I, Hughes JW. Islet cilia and glucose homeostasis. Front Cell Dev Biol 2022; 10:1082193. [PMID: 36531945 PMCID: PMC9751591 DOI: 10.3389/fcell.2022.1082193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/22/2022] [Indexed: 09/05/2023] Open
Abstract
Diabetes is a growing pandemic affecting over ten percent of the U.S. population. Individuals with all types of diabetes exhibit glucose dysregulation due to altered function and coordination of pancreatic islets. Within the critical intercellular space in pancreatic islets, the primary cilium emerges as an important physical structure mediating cell-cell crosstalk and signal transduction. Many events leading to hormone secretion, including GPCR and second-messenger signaling, are spatiotemporally regulated at the level of the cilium. In this review, we summarize current knowledge of cilia action in islet hormone regulation and glucose homeostasis, focusing on newly implicated ciliary pathways that regulate insulin exocytosis and intercellular communication. We present evidence of key signaling proteins on islet cilia and discuss ways in which cilia might functionally connect islet endocrine cells with the non-endocrine compartments. These discussions aim to stimulate conversations regarding the extent of cilia-controlled glucose homeostasis in health and in metabolic diseases.
Collapse
Affiliation(s)
| | - Jing W. Hughes
- Division of Endocrinology, Metabolism and Lipid Research, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, United States
| |
Collapse
|
29
|
Moore ER. Primary Cilia: The New Face of Craniofacial Research. Biomolecules 2022; 12:biom12121724. [PMID: 36551151 PMCID: PMC9776107 DOI: 10.3390/biom12121724] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The primary cilium is a solitary, sensory organelle that extends from the surface of nearly every vertebrate cell, including craniofacial cells. This organelle converts chemical and physical external stimuli into intracellular signaling cascades and mediates several well-known signaling pathways simultaneously. Thus, the primary cilium is considered a cellular signaling nexus and amplifier. Primary cilia dysfunction directly results in a collection of diseases and syndromes that typically affect multiple organ systems, including the face and teeth. Despite this direct connection, primary cilia are largely unexplored in craniofacial research. In this review, I briefly summarize craniofacial abnormalities tied to the primary cilium and examine the existing information on primary cilia in craniofacial development and repair. I close with a discussion on preliminary studies that motivate future areas of exploration that are further supported by studies performed in long bone and kidney cells.
Collapse
Affiliation(s)
- Emily R Moore
- Harvard School of Dental Medicine, Department of Developmental Biology, 188 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|
30
|
Eisa-Beygi S, Burrows PE, Link BA. Endothelial cilia dysfunction in pathogenesis of hereditary hemorrhagic telangiectasia. Front Cell Dev Biol 2022; 10:1037453. [PMID: 36438574 PMCID: PMC9686338 DOI: 10.3389/fcell.2022.1037453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/21/2022] [Indexed: 09/09/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is associated with defective capillary network, leading to dilated superficial vessels and arteriovenous malformations (AVMs) in which arteries connect directly to the veins. Loss or haploinsufficiency of components of TGF-β signaling, ALK1, ENG, SMAD4, and BMP9, have been implicated in the pathogenesis AVMs. Emerging evidence suggests that the inability of endothelial cells to detect, transduce and respond to blood flow, during early development, is an underpinning of AVM pathogenesis. Therefore, components of endothelial flow detection may be instrumental in potentiating TGF-β signaling in perfused blood vessels. Here, we argue that endothelial cilium, a microtubule-based and flow-sensitive organelle, serves as a signaling hub by coupling early flow detection with potentiation of the canonical TGF-β signaling in nascent endothelial cells. Emerging evidence from animal models suggest a role for primary cilia in mediating vascular development. We reason, on recent observations, that endothelial cilia are crucial for vascular development and that embryonic loss of endothelial cilia will curtail TGF-β signaling, leading to associated defects in arteriovenous development and impaired vascular stability. Loss or dysfunction of endothelial primary cilia may be implicated in the genesis of AVMs due, in part, to inhibition of ALK1/SMAD4 signaling. We speculate that AVMs constitute part of the increasing spectrum of ciliopathy-associated vascular defects.
Collapse
Affiliation(s)
- Shahram Eisa-Beygi
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Patricia E. Burrows
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Brian A. Link
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
31
|
Stoufflet J, Caillé I. The Primary Cilium and Neuronal Migration. Cells 2022; 11:3384. [PMID: 36359777 PMCID: PMC9658458 DOI: 10.3390/cells11213384] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 09/29/2023] Open
Abstract
The primary cilium (PC) is a microtubule-based tiny sensory organelle emanating from the centrosome and protruding from the surface of most eukaryotic cells, including neurons. The extremely severe phenotypes of ciliopathies have suggested their paramount importance for multiple developmental events, including brain formation. Neuronal migration is an essential step of neural development, with all neurons traveling from their site of birth to their site of integration. Neurons perform a unique type of cellular migration called cyclic saltatory migration, where their soma periodically jumps along with the stereotyped movement of their centrosome. We will review here how the role of the PC on cell motility was first described in non-neuronal cells as a guide pointing to the direction of migration. We will see then how these findings are extended to neuronal migration. In neurons, the PC appears to regulate the rhythm of cyclic saltatory neuronal migration in multiple systems. Finally, we will review recent findings starting to elucidate how extracellular cues sensed by the PC could be intracellularly transduced to regulate the machinery of neuronal migration. The PC of migrating neurons was unexpectedly discovered to display a rhythmic extracellular emergence during each cycle of migration, with this transient exposure to the external environment associated with periodic transduction of cyclic adenosine monophosphate (cAMP) signaling at the centrosome. The PC in migrating neurons thus uniquely appears as a beat maker, regulating the tempo of cyclic saltatory migration.
Collapse
Affiliation(s)
- Julie Stoufflet
- Laboratory of Molecular Regulation of Neurogenesis, GIGA-Stem Cells and GIGA-Neurosciences, Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R), University of Liège, CHU Sart Tilman, 4000 Liège, Belgium
| | - Isabelle Caillé
- Inserm U1130, Institut de Biologie Paris Seine (IBPS), Neuroscience Paris Seine (NPS), Sorbonne University, CNRS UMR8246, 75005 Paris, France
- University of Paris Cité, 75020 Paris, France
| |
Collapse
|
32
|
Effects of Exercise or Mechanical Stimulation on Bone Development and Bone Repair. Stem Cells Int 2022; 2022:5372229. [PMID: 36213684 PMCID: PMC9534715 DOI: 10.1155/2022/5372229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/07/2022] [Indexed: 11/25/2022] Open
Abstract
The development and regeneration of the bone are tightly regulated by mechanical cues. Multiple cell types, including osteoblasts, osteocytes, osteoclasts, mesenchymal stem cells (MSCs), and recently found skeletal stem cells (SSCs), are responsible for efficient bone development and injury repair. The immune cells in the environment interact with bone cells to maintain homeostasis and facilitate bone regeneration. Investigation of the mechanism by which these cells sense and respond to mechanical signals in bone is fundamental for optimal clinical intervention in bone injury healing. We discuss the effects of exercise programs on fracture healing in animal models and human patients, which encouragingly suggest that carefully designed exercise prescriptions can improve the result of fracture healing during the remodeling phase. However, additional clinical tracing and date accumulation are still required for the pervasive application of exercise prescriptions to improve fracture healing.
Collapse
|
33
|
Song S, Zhang H, Wang X, Chen W, Cao W, Zhang Z, Shi C. The role of mechanosensitive Piezo1 channel in diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 172:39-49. [PMID: 35436566 DOI: 10.1016/j.pbiomolbio.2022.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 06/14/2023]
Abstract
Mechanotransduction is associated with organ development and homoeostasis. Piezo1 and Piezo2 are novel mechanosensitive ion channels (MSCs) in mammals. MSCs are membrane proteins that are critical for the mechanotransduction of living cells. Current studies have demonstrated that the Piezo protein family not only functions in volume regulation, cellular migration, proliferation, and apoptosis but is also important for human diseases of various systems. The complete loss of Piezo1 and Piezo2 function is fatal in the embryonic period. This review summarizes the role of Piezo1 in diseases of different systems and perspectives potential treatments related to Piezo1 for these diseases.
Collapse
Affiliation(s)
- Siqi Song
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Hong Zhang
- Department of Cardiac Surgery, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Xiaoya Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Wei Chen
- Department of Urology, The Affiliated Xinqiao Hospital, The Third Military Medical University, Chongqing, 400038, China
| | - Wenxuan Cao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China
| | - Zhe Zhang
- School of Basic Medicine, College of Medicine, Qingdao University, Qingdao 266071, Shandong Province, China.
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao, 266071, Shandong Province, China.
| |
Collapse
|
34
|
Jeon HH, Kang J, Li J(M, Kim D, Yuan G, Almer N, Liu M, Yang S. The Effect of IFT80 Deficiency in Osteocytes on Orthodontic Loading-Induced and Physiologic Bone Remodeling: In Vivo Study. Life (Basel) 2022; 12:1147. [PMID: 36013326 PMCID: PMC9410307 DOI: 10.3390/life12081147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Osteocytes are the main mechanosensory cells during orthodontic and physiologic bone remodeling. However, the question of how osteocytes transmit mechanical stimuli to biological responses remains largely unanswered. Intraflagellar transport (IFT) proteins are important for the formation and function of cilia, which are proposed to be mechanical sensors in osteocytes. In particular, IFT80 is highly expressed in mouse skulls and essential for ciliogenesis. This study aims to investigate the short- and long-term effects of IFT80 deletion in osteocytes on orthodontic bone remodeling and physiological bone remodeling in response to masticatory force. We examined 10-week-old experimental DMP1 CRE+.IFT80f/f and littermate control DMP1 CRE-.IFT80f/f mice. After 5 and 12 days of orthodontic force loading, the orthodontic tooth movement distance and bone parameters were evaluated using microCT. Osteoclast formation was assessed using TRAP-stained paraffin sections. The expression of sclerostin and RANKL was examined using immunofluorescence stain. We found that the deletion of IFT80 in osteocytes did not significantly impact either orthodontic or physiologic bone remodeling, as demonstrated by similar OTM distances, osteoclast numbers, bone volume fractions (bone volume/total volume), bone mineral densities, and the expressions of sclerostin and RANKL. Our findings suggest that there are other possible mechanosensory systems in osteocytes and anatomic limitations to cilia deflection in osteocytes in vivo.
Collapse
Affiliation(s)
- Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (J.L.); (D.K.); (N.A.)
| | - Jessica Kang
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (J.L.); (D.K.); (N.A.)
| | - Jiahui (Madelaine) Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (J.L.); (D.K.); (N.A.)
| | - Douglas Kim
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (J.L.); (D.K.); (N.A.)
| | - Gongsheng Yuan
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Nicolette Almer
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (J.K.); (J.L.); (D.K.); (N.A.)
| | - Min Liu
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Shuying Yang
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- The Penn Center for Musculoskeletal Disorders, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Innovation & Precision Dentistry, School of Dental Medicine, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
35
|
Wang C, Qu K, Wang J, Qin R, Li B, Qiu J, Wang G. Biomechanical regulation of planar cell polarity in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166495. [PMID: 35850177 DOI: 10.1016/j.bbadis.2022.166495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2023]
Abstract
Cell polarity refers to the uneven distribution of certain cytoplasmic components in a cell with a spatial order. The planar cell polarity (PCP), the cell aligns perpendicular to the polar plane, in endothelial cells (ECs) has become a research hot spot. The planar polarity of ECs has a positive significance on the regulation of cardiovascular dysfunction, pathological angiogenesis, and ischemic stroke. The endothelial polarity is stimulated and regulated by biomechanical force. Mechanical stimuli promote endothelial polarization and make ECs produce PCP to maintain the normal physiological and biochemical functions. Here, we overview recent advances in understanding the interplay and mechanism between PCP and ECs function involved in mechanical forces, with a focus on PCP signaling pathways and organelles in regulating the polarity of ECs. And then showed the related diseases caused by ECs polarity dysfunction. This study provides new ideas and therapeutic targets for the treatment of endothelial PCP-related diseases.
Collapse
Affiliation(s)
- Caihong Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Rui Qin
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Bingyi Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
36
|
Paese CLB, Chang CF, Kristeková D, Brugmann SA. Pharmacological intervention of the FGF-PTH axis as a potential therapeutic for craniofacial ciliopathies. Dis Model Mech 2022; 15:275968. [PMID: 35818799 PMCID: PMC9403750 DOI: 10.1242/dmm.049611] [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: 04/21/2022] [Accepted: 07/05/2022] [Indexed: 11/20/2022] Open
Abstract
Ciliopathies represent a disease class characterized by a broad range of phenotypes including polycystic kidneys and skeletal anomalies. Ciliopathic skeletal phenotypes are among the most common and most difficult to treat due to a poor understanding of the pathological mechanisms leading to disease. Using an avian model (talpid2) for a human ciliopathy with both kidney and skeletal anomalies (Orofaciodigital syndrome 14), we identified disruptions in the FGF23-PTH axis that resulted in reduced calcium uptake in the developing mandible and subsequent micrognathia. While pharmacological intervention with the FDA-approved pan-FGFR inhibitor AZD4547 alone rescued expression of the FGF target Sprouty2, it did not significantly rescue micrognathia. In contrast, treatment with a cocktail of AZD4547 and Teriparatide acetate, a PTH agonist and FDA-approved treatment for osteoporosis, resulted in a molecular, cellular, and phenotypic rescue of ciliopathic micrognathia in talpid2 mutants. Together, these data provide novel insight into pathological molecular mechanisms associated with ciliopathic skeletal phenotypes and a potential therapeutic strategy for a pleiotropic disease class with limited to no treatment options.
Collapse
Affiliation(s)
- Christian Louis Bonatto Paese
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Ching-Fang Chang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daniela Kristeková
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, v.v.i., Czech Academy of Sciences, Brno 602 00, Czech Republic.,Department of Experimental Biology, Faculty of Science, Masaryk University, Brno 625 00, Czech Republic
| | - Samantha A Brugmann
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
37
|
TRPP2 ion channels: The roles in various subcellular locations. Biochimie 2022; 201:116-127. [PMID: 35760123 DOI: 10.1016/j.biochi.2022.06.010] [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: 11/14/2021] [Revised: 06/14/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022]
Abstract
TRPP2 (PC2, PKD2 or Polycytin-2), encoded by PKD2 gene, belongs to the nonselective cation channel TRP family. Recently, the three-dimensional structure of TRPP2 was constructed. TRPP2 mainly functions in three subcellular compartments: endoplasmic reticulum, plasma membrane and primary cilia. TRPP2 can act as a calcium-activated intracellular calcium release channel on the endoplasmic reticulum. TRPP2 also interacts with other Ca2+ release channels to regulate calcium release, like IP3R and RyR2. TRPP2 acts as an ion channel regulated by epidermal growth factor through activation of downstream factors in the plasma membrane. TRPP2 binding to TRPC1 in the plasma membrane or endoplasmic reticulum is associated with mechanosensitivity. In cilium, TRPP2 was found to combine with PKD1 and TRPV4 to form a complex related to mechanosensitivity. Because TRPP2 is involved in regulating intracellular ion concentration, TRPP2 mutations often lead to autosomal dominant polycystic kidney disease, which may also be associated with cardiovascular disease. In this paper, we review the molecular structure of TRPP2, the subcellular localization of TRPP2, the related functions and mechanisms of TRPP2 at different sites, and the diseases related to TRPP2.
Collapse
|
38
|
Primary cilia in satellite cells are the mechanical sensors for muscle hypertrophy. Proc Natl Acad Sci U S A 2022; 119:e2103615119. [PMID: 35671424 PMCID: PMC9214504 DOI: 10.1073/pnas.2103615119] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Skeletal muscle atrophy is commonly associated with aging, immobilization, muscle unloading, and congenital myopathies. Generation of mature muscle cells from skeletal muscle satellite cells (SCs) is pivotal in repairing muscle tissue. Exercise therapy promotes muscle hypertrophy and strength. Primary cilium is implicated as the mechanical sensor in some mammalian cells, but its role in skeletal muscle cells remains vague. To determine mechanical sensors for exercise-induced muscle hypertrophy, we established three SC-specific cilium dysfunctional mouse models-Myogenic factor 5 (Myf5)-Arf-like Protein 3 (Arl3)-/-, Paired box protein Pax-7 (Pax7)-Intraflagellar transport protein 88 homolog (Ift88)-/-, and Pax7-Arl3-/--by specifically deleting a ciliary protein ARL3 in MYF5-expressing SCs, or IFT88 in PAX7-expressing SCs, or ARL3 in PAX7-expressing SCs, respectively. We show that the Myf5-Arl3-/- mice develop grossly the same as WT mice. Intriguingly, mechanical stimulation-induced muscle hypertrophy or myoblast differentiation is abrogated in Myf5-Arl3-/- and Pax7-Arl3-/- mice or primary isolated Myf5-Arl3-/- and Pax7-Ift88-/- myoblasts, likely due to defective cilia-mediated Hedgehog (Hh) signaling. Collectively, we demonstrate SC cilia serve as mechanical sensors and promote exercise-induced muscle hypertrophy via Hh signaling pathway.
Collapse
|
39
|
Serra R. New and Unexpected Roles for Primary Cilia in Coordinating Response to Mechanical Load in Articular and Growth Plate Cartilages. J Bone Miner Res 2022; 37:1079-1080. [PMID: 35451172 DOI: 10.1002/jbmr.4557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/08/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Rosa Serra
- Cell Developmental and Integrative Biology, University of Alabama, Birmingham, AL, USA
| |
Collapse
|
40
|
Rux D, Helbig K, Han B, Cortese C, Koyama E, Han L, Pacifici M. Primary Cilia Direct Murine Articular Cartilage Tidemark Patterning Through Hedgehog Signaling and Ambulatory Load. J Bone Miner Res 2022; 37:1097-1116. [PMID: 35060644 PMCID: PMC9177786 DOI: 10.1002/jbmr.4506] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/01/2022] [Accepted: 01/08/2022] [Indexed: 11/06/2022]
Abstract
Articular cartilage (AC) is essential for body movement but is highly susceptible to degenerative diseases and has poor self-repair capacity. To improve current subpar regenerative treatments, developmental mechanisms of AC should be clarified and, specifically, how its postnatal multizone organization is acquired. Primary cilia are cell surface organelles crucial for mammalian tissue morphogenesis. Although their importance for chondrocyte function is appreciated, their specific roles in postnatal AC morphogenesis remain unclear. To explore these mechanisms, we used a murine conditional loss-of-function approach (Ift88-flox) targeting joint-lineage progenitors (Gdf5Cre) and monitored postnatal knee AC development. Joint formation and growth up to juvenile stages were largely unaffected. However, mature AC (aged 2 months) exhibited disorganized extracellular matrix, decreased aggrecan and collagen II due to reduced gene expression (not increased catabolism), and marked reduction of AC modulus by 30%-50%. In addition, and unexpectedly, we discovered that tidemark patterning was severely disrupted, as was hedgehog signaling, and exhibited specificity based on regional load-bearing functions of AC. Interestingly, Prg4 expression was markedly increased in highly loaded sites in mutants. Together, our data provide evidence that primary cilia orchestrate postnatal AC morphogenesis including tidemark topography, zonal matrix composition, and ambulation load responses. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Danielle Rux
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kimberly Helbig
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Courtney Cortese
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA
| | - Maurizio Pacifici
- Translational Research Program in Pediatric Orthopaedics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
41
|
Kleene SJ. Regenerative Calcium Currents in Renal Primary Cilia. Front Physiol 2022; 13:894518. [PMID: 35620606 PMCID: PMC9127361 DOI: 10.3389/fphys.2022.894518] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/18/2022] [Indexed: 11/13/2022] Open
Abstract
Polycystic kidney disease (PKD) is a leading cause of end-stage renal disease. PKD arises from mutations in proteins, one a Ca2+-conducting channel, expressed in the primary cilia of renal epithelial cells. A common hypothesis is that Ca2+ entering through ciliary ion channels may reduce cystogenesis. The cilia have at least two Ca2+-conducting channels: polycystin-2 (PC2) and TRPV4 (transient receptor potential (TRP) cation channel, subfamily V, member 4), but how substantially they can increase intraciliary Ca2+ is unknown. By recording channel activities in isolated cilia, conditions are identified under which the channels can increase free Ca2+ within the cilium by at least 500-fold through regenerative (positive-feedback) signaling. Ca2+ that has entered through a channel can activate the channel internally, which increases the Ca2+ influx, and so on. Regenerative signaling is favored when the concentration of the Ca2+ buffer is reduced or when a slower buffer is used. Under such conditions, the Ca2+ that enters the cilium through a single PC2 channel is sufficient to almost fully activate that same channel. Regenerative signaling is not detectable with reduced external Ca2+. Reduced buffering also allows regenerative signaling through TRPV4 channels, but not through TRPM4 (TRP subfamily M, member 4) channels, which are activated by Ca2+ but do not conduct it. On a larger scale, Ca2+ that enters through TRPV4 channels can cause secondary activation of PC2 channels. I discuss the likelihood of regenerative ciliary Ca2+ signaling in vivo, a possible mechanism for its activation, and how it might relate to cystogenesis.
Collapse
Affiliation(s)
- Steven J. Kleene
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
42
|
Spasic M, Duffy MP, Jacobs CR. Fenoldopam Sensitizes Primary Cilia-Mediated Mechanosensing to Promote Osteogenic Intercellular Signaling and Whole Bone Adaptation. J Bone Miner Res 2022; 37:972-982. [PMID: 35230705 PMCID: PMC9098671 DOI: 10.1002/jbmr.4536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 02/01/2022] [Accepted: 02/12/2022] [Indexed: 11/05/2022]
Abstract
Bone cells actively respond to mechanical stimuli to direct bone formation, yet there is no current treatment strategy for conditions of low bone mass and osteoporosis designed to target the inherent mechanosensitivity of bone. Our group has previously identified the primary cilium as a critical mechanosensor within bone, and that pharmacologically targeting the primary cilium with fenoldopam can enhance osteocyte mechanosensitivity. Here, we demonstrate that potentiating osteocyte mechanosensing with fenoldopam in vitro promotes pro-osteogenic paracrine signaling to osteoblasts. Conversely, impairing primary cilia formation and the function of key ciliary mechanotransduction proteins attenuates this intercellular signaling cascade. We then utilize an in vivo model of load-induced bone formation to demonstrate that fenoldopam treatment sensitizes bones of both healthy and osteoporotic mice to mechanical stimulation. Furthermore, we show minimal adverse effects of this treatment and demonstrate that prolonged treatment biases trabecular bone adaptation. This work is the first to examine the efficacy of targeting primary cilia-mediated mechanosensing to enhance bone formation in osteoporotic animals. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Milos Spasic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Michael P Duffy
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | |
Collapse
|
43
|
Mechanical regulation of bone remodeling. Bone Res 2022; 10:16. [PMID: 35181672 PMCID: PMC8857305 DOI: 10.1038/s41413-022-00190-4] [Citation(s) in RCA: 178] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/04/2021] [Accepted: 12/13/2021] [Indexed: 12/17/2022] Open
Abstract
Bone remodeling is a lifelong process that gives rise to a mature, dynamic bone structure via a balance between bone formation by osteoblasts and resorption by osteoclasts. These opposite processes allow the accommodation of bones to dynamic mechanical forces, altering bone mass in response to changing conditions. Mechanical forces are indispensable for bone homeostasis; skeletal formation, resorption, and adaptation are dependent on mechanical signals, and loss of mechanical stimulation can therefore significantly weaken the bone structure, causing disuse osteoporosis and increasing the risk of fracture. The exact mechanisms by which the body senses and transduces mechanical forces to regulate bone remodeling have long been an active area of study among researchers and clinicians. Such research will lead to a deeper understanding of bone disorders and identify new strategies for skeletal rejuvenation. Here, we will discuss the mechanical properties, mechanosensitive cell populations, and mechanotransducive signaling pathways of the skeletal system.
Collapse
|
44
|
Aurora A and AKT Kinase Signaling Associated with Primary Cilia. Cells 2021; 10:cells10123602. [PMID: 34944109 PMCID: PMC8699881 DOI: 10.3390/cells10123602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 02/07/2023] Open
Abstract
Dysregulation of kinase signaling is associated with various pathological conditions, including cancer, inflammation, and autoimmunity; consequently, the kinases involved have become major therapeutic targets. While kinase signaling pathways play crucial roles in multiple cellular processes, the precise manner in which their dysregulation contributes to disease is dependent on the context; for example, the cell/tissue type or subcellular localization of the kinase or substrate. Thus, context-selective targeting of dysregulated kinases may serve to increase the therapeutic specificity while reducing off-target adverse effects. Primary cilia are antenna-like structures that extend from the plasma membrane and function by detecting extracellular cues and transducing signals into the cell. Cilia formation and signaling are dynamically regulated through context-dependent mechanisms; as such, dysregulation of primary cilia contributes to disease in a variety of ways. Here, we review the involvement of primary cilia-associated signaling through aurora A and AKT kinases with respect to cancer, obesity, and other ciliopathies.
Collapse
|
45
|
Robinson ST, Shyu PT, Guo XE. Mechanical loading and parathyroid hormone effects and synergism in bone vary by site and modeling/remodeling regime. Bone 2021; 153:116171. [PMID: 34492358 PMCID: PMC8499476 DOI: 10.1016/j.bone.2021.116171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/01/2021] [Accepted: 08/31/2021] [Indexed: 01/22/2023]
Abstract
Intermittent injections of parathyroid hormone (PTH) and mechanical loading are both known to effect a net increase in bone mass. Fundamentally, bone metabolism can be divided into modeling (uncoupled formation or resorption) and remodeling (subsequent formation biologically coupled to resorption in space and time). Methods to delineate the bone response between these regimes are scant but have garnered recent attention and acceptance, and will be critical tools to properly assess short- and long-term efficacy of osteoporosis treatments. To this end, we employ a time-lapse micro-computed tomography strategy to quantify and localize modeling and remodeling volumes over 4 weeks of concurrent PTH treatment and mechanical loading. Modeled and remodeled volumes are probed for differences with respect to treatment, loading, and interactions thereof in trabecular and cortical bone compartments, which were further separated by plate/rod microarchitecture and periosteal/endosteal surfaces, respectively. Loading effects are further considered independently with regard to localized strain environments. Our findings indicate that in trabecular bone, PTH and loading stimulate anabolic modeling additively, and remodeling synergistically. PTH tends to lead to bone accumulation indiscriminate of trabecular microarchitecture, whereas loading tends to more strongly affect plates than rods. The cortical surfaces responded uniquely to PTH and loading, with synergistic effects on the periosteal surface for anabolic modeling, and on the endosteal surface for catabolic modeling. The increase in catabolic modeling due to loading, which is enhanced by PTH, is concentrated to areas of the endosteal surface under low strain and to our knowledge has not previously been reported. Taken together, the effects of PTH, loading, and their interactions, are shown to be dependent on the specific bone compartment and metabolic regime; this may explain some discrepancies in previously-reported findings.
Collapse
Affiliation(s)
- Samuel T Robinson
- Bone Bioengineering Laboratory, 351 Engineering Terrace, Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| | - Peter T Shyu
- Bone Bioengineering Laboratory, 351 Engineering Terrace, Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| | - X Edward Guo
- Bone Bioengineering Laboratory, 351 Engineering Terrace, Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY 10027, USA.
| |
Collapse
|
46
|
Sudarikova A, Vasileva V, Sultanova R, Ilatovskaya D. Recent advances in understanding ion transport mechanisms in polycystic kidney disease. Clin Sci (Lond) 2021; 135:2521-2540. [PMID: 34751394 PMCID: PMC8589009 DOI: 10.1042/cs20210370] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/15/2021] [Accepted: 10/26/2021] [Indexed: 12/18/2022]
Abstract
This review focuses on the most recent advances in the understanding of the electrolyte transport-related mechanisms important for the development of severe inherited renal disorders, autosomal dominant (AD) and recessive (AR) forms of polycystic kidney disease (PKD). We provide here a basic overview of the origins and clinical aspects of ARPKD and ADPKD and discuss the implications of electrolyte transport in cystogenesis. Special attention is devoted to intracellular calcium handling by the cystic cells, with a focus on polycystins and fibrocystin, as well as other calcium level regulators, such as transient receptor potential vanilloid type 4 (TRPV4) channels, ciliary machinery, and purinergic receptor remodeling. Sodium transport is reviewed with a focus on the epithelial sodium channel (ENaC), and the role of chloride-dependent fluid secretion in cystic fluid accumulation is discussed. In addition, we highlight the emerging promising concepts in the field, such as potassium transport, and suggest some new avenues for research related to electrolyte handling.
Collapse
Affiliation(s)
| | | | - Regina F. Sultanova
- Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia
| | | |
Collapse
|
47
|
The mechanosensory and mechanotransductive processes mediated by ion channels and the impact on bone metabolism: A systematic review. Arch Biochem Biophys 2021; 711:109020. [PMID: 34461086 DOI: 10.1016/j.abb.2021.109020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Mechanical environments were associated with alterations in bone metabolism. Ion channels present on bone cells are indispensable for bone metabolism and can be directly or indirectly activated by mechanical stimulation. This review aimed to discuss the literature reporting the mechanical regulatory effects of ion channels on bone cells and bone tissue. An electronic search was conducted in PubMed, Embase and Web of Science. Studies about mechanically induced alteration of bone cells and bone tissue by ion channels were included. Ion channels including TRP family channels, Ca2+ release-activated Ca2+ channels (CRACs), Piezo1/2 channels, purinergic receptors, NMDA receptors, voltage-sensitive calcium channels (VSCCs), TREK2 potassium channels, calcium- and voltage-dependent big conductance potassium (BKCa) channels, small conductance, calcium-activated potassium (SKCa) channels and epithelial sodium channels (ENaCs) present on bone cells and bone tissue participate in the mechanical regulation of bone development in addition to contributing to direct or indirect mechanotransduction such as altered membrane potential and ionic flux. Physiological (beneficial) mechanical stimulation could induce the anabolism of bone cells and bone tissue through ion channels, but abnormal (harmful) mechanical stimulation could also induce the catabolism of bone cells and bone tissue through ion channels. Functional expression of ion channels is vital for the mechanotransduction of bone cells. Mechanical activation (opening) of ion channels triggers ion influx and induces the activation of intracellular modulators that can influence bone metabolism. Therefore, mechanosensitive ion channels provide new insights into therapeutic targets for the treatment of bone-related diseases such as osteopenia and aseptic implant loosening.
Collapse
|
48
|
Abstract
As the world's population ages, the treatment of osteoporosis is a major problem to be addressed. The cause of osteoporosis remains unclear. Ca2+ is not only an important component of bones but also plays a key role in osteoporosis treatment. Transient receptor potential vanilloid (TRPV) channels are one of the TRP channel families that is widely distributed in various organs, playing an important role in the physiological regulation of the human body. Bone formation and bone absorption may require Ca2+ transport via TRPV channels. It has been proven that the TRPV subtypes 1, 2, 4, 5, 6 (TRPV1, TRPV2, TRPV4, TRPV5, TRPV6) may affect bone metabolism balance through selective regulation of Ca2+. They significantly regulate osteoblast/osteoclast proliferation, differentiation and function. The purpose of this review is to explore the mechanisms of TRPV channels involved in regulation of the differentiation of osteoblasts and osteoclasts, as well as to discuss the latest developments in current researches, which may provide new clues and directions for an in-depth study of osteoporosis and other related bone metabolic diseases.
Collapse
|
49
|
Vasileva VY, Sultanova RF, Sudarikova AV, Ilatovskaya DV. Insights Into the Molecular Mechanisms of Polycystic Kidney Diseases. Front Physiol 2021; 12:693130. [PMID: 34566674 PMCID: PMC8456103 DOI: 10.3389/fphys.2021.693130] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 08/10/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant (AD) and autosomal recessive (AR) polycystic kidney diseases (PKD) are severe multisystem genetic disorders characterized with formation and uncontrolled growth of fluid-filled cysts in the kidney, the spread of which eventually leads to the loss of renal function. Currently, there are no treatments for ARPKD, and tolvaptan is the only FDA-approved drug that alleviates the symptoms of ADPKD. However, tolvaptan has only a modest effect on disease progression, and its long-term use is associated with many side effects. Therefore, there is still a pressing need to better understand the fundamental mechanisms behind PKD development. This review highlights current knowledge about the fundamental aspects of PKD development (with a focus on ADPKD) including the PC1/PC2 pathways and cilia-associated mechanisms, major molecular cascades related to metabolism, mitochondrial bioenergetics, and systemic responses (hormonal status, levels of growth factors, immune system, and microbiome) that affect its progression. In addition, we discuss new information regarding non-pharmacological therapies, such as dietary restrictions, which can potentially alleviate PKD.
Collapse
Affiliation(s)
| | - Regina F Sultanova
- Saint-Petersburg State Chemical Pharmaceutical University, St. Petersburg, Russia.,Department of Physiology, Augusta University, Augusta, GA, United States
| | | | | |
Collapse
|
50
|
Primary cilia in hard tissue development and diseases. Front Med 2021; 15:657-678. [PMID: 34515939 DOI: 10.1007/s11684-021-0829-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/13/2020] [Indexed: 10/20/2022]
Abstract
Bone and teeth are hard tissues. Hard tissue diseases have a serious effect on human survival and quality of life. Primary cilia are protrusions on the surfaces of cells. As antennas, they are distributed on the membrane surfaces of almost all mammalian cell types and participate in the development of organs and the maintenance of homeostasis. Mutations in cilium-related genes result in a variety of developmental and even lethal diseases. Patients with multiple ciliary gene mutations present overt changes in the skeletal system, suggesting that primary cilia are involved in hard tissue development and reconstruction. Furthermore, primary cilia act as sensors of external stimuli and regulate bone homeostasis. Specifically, substances are trafficked through primary cilia by intraflagellar transport, which affects key signaling pathways during hard tissue development. In this review, we summarize the roles of primary cilia in long bone development and remodeling from two perspectives: primary cilia signaling and sensory mechanisms. In addition, the cilium-related diseases of hard tissue and the manifestations of mutant cilia in the skeleton and teeth are described. We believe that all the findings will help with the intervention and treatment of related hard tissue genetic diseases.
Collapse
|