1
|
Chen Z, Chen P, Li J, Landao-Bassonga E, Papadimitriou J, Gao J, Liu D, Tai A, Ma J, Lloyd D, Kennedy BF, Zheng MH. External strain on the plasma membrane is relayed to the endoplasmic reticulum by membrane contact sites and alters cellular energetics. SCIENCE ADVANCES 2025; 11:eads6132. [PMID: 40561024 DOI: 10.1126/sciadv.ads6132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 05/20/2025] [Indexed: 06/28/2025]
Abstract
Mechanotransduction is essential for living cells to adapt to their extracellular environment. However, it is unclear how the biophysical adaptation of intracellular organelles responds to mechanical stress or how these adaptive changes affect cellular homeostasis. Here, using the tendon cell as a mechanosensitive cell type within a bioreactor, we show that the tension of the plasma membrane (PM) and the endoplasmic reticulum (ER) adaptively increases in response to repetitive external stimuli. Depletion of stromal interaction molecule 1 (STIM1), the highest expressed PM-ER tether protein, interfered with mechanotransduction from the PM to the ER, and affected the ER tension. We found that an optimized mechanical strain increased ER tension in a homeostatic manner, but excessive strain resulted in ER expansion, as well as activating ER stress. Last, we showed that changes in ER tension were linked with ER-mitochondria interactions and associated with cellular energetics and function. Together, these findings identify a PM-ER mechanotransduction mechanism that dose-dependently regulates cellular metabolism.
Collapse
Affiliation(s)
- Ziming Chen
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Peilin Chen
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Jiayue Li
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Nedlands, WA 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
| | - Euphemie Landao-Bassonga
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - John Papadimitriou
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Junjie Gao
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
- Department of Orthopaedics, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, PR China
| | - Delin Liu
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Andrew Tai
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| | - Jinjin Ma
- Institute of Future Health, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, PR China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, PR China
| | - David Lloyd
- Centre of Biomedical and Rehabilitation Engineering, Griffith University, Gold Coast, QLD, Australia
| | - Brendan F Kennedy
- BRITElab, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, and Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Electrical, Electronic and Computer Engineering, School of Engineering, The University of Western Australia, Nedlands, WA 6009, Australia
- Australian Research Council Centre for Personalised Therapeutics Technologies, Melbourne, VIC, Australia
- Institute of Physics, Faculty of Physics, Astronomy and Informatics, Nicolaus Copernicus University in Toruń, Grudziadzka 5, 87-100 Torun, Poland
| | - Ming Hao Zheng
- Centre for Orthopaedic Research, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA 6009, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, WA 6009, Australia
| |
Collapse
|
2
|
Rudzitis CN, Lakk M, Singh A, Redmon SN, Kirdajová D, Tseng YT, De Ieso ML, Stamer WD, Herberg S, Krizaj D. TRPV4 activation by TGFβ2 enhances cellular contractility and drives ocular hypertension. eLife 2025; 14:RP104894. [PMID: 40552711 PMCID: PMC12187138 DOI: 10.7554/elife.104894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2025] Open
Abstract
The risk for developing primary open-angle glaucoma (POAG) correlates with the magnitude of ocular hypertension (OHT) and the concentration of transforming growth factor-β2 (TGFβ2) in the aqueous humor. Effective treatment of POAG requires a detailed understanding of the interaction between pressure sensing mechanisms in the trabecular meshwork (TM) and biochemical risk factors. Here, we employed molecular, optical, electrophysiological, and tonometric strategies to establish the role of TGFβ2 in transcription and functional expression of mechanosensitive channel isoforms alongside studies of TM contractility in biomimetic hydrogels and intraocular pressure (IOP) regulation in a mouse model of TGFβ2-induced OHT. TGFβ2 upregulated expression of Trpv4 and Piezo1 transcripts and time-dependently augmented functional TRPV4 activation. TRPV4 agonists induced contractility of TM-seeded hydrogels, whereas pharmacological inhibition suppressed TGFβ2-induced hypercontractility and abrogated OHT in eyes overexpressing TGFβ2. Trpv4-deficient mice resisted TGFβ2-driven increases in IOP, but nocturnal OHT was not additive to TGFβ-evoked OHT. Our study establishes the fundamental role of TGFβ as a modulator of mechanosensing in nonexcitable cells, identifies the TRPV4 channel as the final common mechanism for TM contractility and circadian and pathological OHT, and offers insights for future treatments that can lower IOP in the sizeable cohort of hypertensive glaucoma patients that resist current treatments.
Collapse
Affiliation(s)
- Christopher Nass Rudzitis
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
| | - Monika Lakk
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
| | - Ayushi Singh
- Department of Ophthalmology and Visual SciencesSyracuseUnited States
- Department of Cell and Developmental Biology, SUNY Upstate Medical UniversitySyracuseUnited States
| | - Sarah N Redmon
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
| | - Denisa Kirdajová
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
| | - Yun-Ting Tseng
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
| | - Michael L De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke UniversityDurhamUnited States
| | - W Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke UniversityDurhamUnited States
| | - Samuel Herberg
- Department of Ophthalmology and Visual SciencesSyracuseUnited States
- Department of Cell and Developmental Biology, SUNY Upstate Medical UniversitySyracuseUnited States
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical UniversitySyracuseUnited States
| | - David Krizaj
- Department of Ophthalmology and Visual SciencesSalt Lake CityUnited States
- Department of Neurobiology, University of UtahSalt Lake CityUnited States
- Department of Bioengineering, University of UtahSalt Lake CityUnited States
| |
Collapse
|
3
|
Wunderer MS, Sparenberg V, Biehl C, Liefeith K, Lips KS. Poly-(D,L)-Lactide-ε-Caprolactone-Methacrylate Is a Suitable Scaffold Material for In Vitro Cartilage Regeneration. Int J Mol Sci 2025; 26:5837. [PMID: 40565317 DOI: 10.3390/ijms26125837] [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: 04/08/2025] [Revised: 05/22/2025] [Accepted: 06/13/2025] [Indexed: 06/28/2025] Open
Abstract
Due to the limited regeneration of cartilage, new implant materials are needed. Biodegradable polymers poly-(D,L)-lactide-ε-caprolactone-methacrylate (LCM) and polyamid-ε-caprolactone-methacrylate (ACM) were recently established and coated with heparin, making them able to prevent blood coagulation and cartilage mineralization. The aim of this study was to analyze the suitability of LCM and ACM alone or coated with heparin (the latter are abbreviated as LCMH and ACMH, respectively) as implant material for cartilage repair. Therefore, mesenchymal stem cells were chondrogenically differentiated in 2D cultures with polymer discs. Differentiation was induced by the supplementation of cell medium with dimethyloxalylglycine, TGF-β, and BMP2. After 5 days, no increase in proinflammatory factors was observed. Cell viability declined on ACM and ACMH discs. During early chondrogenesis, SOX9 expression increased on LCM and LCMH discs, while TRPV4 expression decreased on ACMH discs. At day 20, the level of collagen type II increased on LCM, LCMH, and ACM discs, demonstrating the ability of chondrogenic development on these implants. In summary, coating with heparin showed no advantages compared to pure LCM and ACM. For cartilage repair, LCM is more suitable than ACM in this 2D in vitro model, which needs to be verified by long-term 3D models and in vivo studies.
Collapse
Affiliation(s)
- Michelle Sophie Wunderer
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Veronika Sparenberg
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Christoph Biehl
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital of Giessen-Marburg GmbH, Campus Giessen, Rudolf Buchheim Strasse 7, 35392 Giessen, Germany
| | - Klaus Liefeith
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, 37308 Heilbad Heiligenstadt, Germany
| | - Katrin Susanne Lips
- Experimental Trauma Surgery, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| |
Collapse
|
4
|
Otoo BS, Moo EK, Komeili A, Hart DA, Herzog W. From fluctuations to stability: In-Situ chondrocyte response to cyclic compressive loading. J Biomech 2025; 186:112734. [PMID: 40319787 DOI: 10.1016/j.jbiomech.2025.112734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/26/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Chondrocytes, the sole cellular components in articular cartilage, are mechanosensitive and undergo significant morphological and volumetric changes in response to mechanical loading. These changes activate ion channels, initiating cellular mechanotransduction processes crucial for maintaining cartilage health. Dynamic loading has been shown to elicit anabolic responses that preserve cartilage integrity, while prolonged mechanical unloading leads to atrophy. However, the intricacies of how chondrocytes respond to dynamic loading remain poorly understood, largely due to technical limitations in capturing real-time cellular responses during loading cycles. This study aimed to advance our understanding of chondrocyte behavior during dynamic cyclic compression loading through high-speed imaging techniques. We developed a protocol to capture changes in chondrocyte volume, shape, and surface area at critical moments of maximal and minimal tissue stress during cyclic loading. Our findings revealed that chondrocyte volume fluctuated cyclically during the first 20 loading cycles, increasing by up to 4 % during load application and decreasing by as much as 8 % during unloading. These volume fluctuations stabilized over time, returning to baseline levels after approximately 100 cycles. Volume changes over time translate to shape change, causing similar oscillatory pattern in cell width and depth strains but not height strain, which remained relatively constant throughout the loading protocol. Changes in surface area mirrored the volume changes but were less pronounced (< 2 % increase), suggesting a protective mechanism against cell membrane rupture. This research offers valuable insights into the dynamic behavior of chondrocytes during cyclic loading, highlighting the importance of considering dynamic environments in cellular biomechanics studies.
Collapse
Affiliation(s)
- Baaba S Otoo
- Human Performance Laboratory, University of Calgary, Calgary, AB, Canada; Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.
| | - Eng Kuan Moo
- Human Performance Laboratory, University of Calgary, Calgary, AB, Canada; Department of Mechanical and Aerospace Engineering, Carleton University, Ottawa, ON, Canada; Department of Technical Physics, University of Eastern Finland, Kuopio, Finland.
| | - Amin Komeili
- Human Performance Laboratory, University of Calgary, Calgary, AB, Canada; Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada; Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB, Canada.
| | - David A Hart
- Human Performance Laboratory, University of Calgary, Calgary, AB, Canada; McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada; Department of Surgery, University of Calgary, Calgary, AB, Canada.
| | - Walter Herzog
- Human Performance Laboratory, University of Calgary, Calgary, AB, Canada; Department of Biomedical Engineering, University of Calgary, Calgary, AB, Canada; Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
5
|
Tong X, Ayushman M, Lee HP, Yang F. Tuning local matrix compliance accelerates mesenchymal stem cell chondrogenesis in 3D sliding hydrogels. Biomaterials 2025; 317:123112. [PMID: 39827509 DOI: 10.1016/j.biomaterials.2025.123112] [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: 10/18/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
The mechanical properties of the extracellular matrix critically regulate stem cell differentiation in 3D. Alginate hydrogels with tunable bulk stiffness and viscoelasticity can modulate differentiation in 3D through mechanotransduction. Such enhanced differentiation is correlated with changes in the local matrix compliance- the extent of matrix deformation under applied load. However, the causal effect of local matrix compliance changes without altering bulk hydrogel mechanics on stem cell differentiation remains unclear. To address this, we report sliding hydrogel (SG) designs with tunable local matrix compliance obtained by varying the molecular mobility of the hydrogel network without changing bulk mechanics. Atomic force microscopy showed increasing SG mobility allowed cells to increasingly deform local niches with lesser forces, indicating higher local matrix compliance. Increasing SG mobility accelerates MSC chondrogenesis in a mobility-dependent manner and is independent of exogenous adhesive ligands or cell volume expansion. The enhanced chondrogenesis in SG is accompanied by enhanced cytoskeletal organization and TRPV4 expression, and blocking these elements abolished the effect. In conclusion, this study establishes a causal link between local matrix compliance and stem cell differentiation and establishes it as a crucial hydrogel design parameter. Furthermore, it offers novel SG designs to probe the role of local matrix compliance in various biological processes.
Collapse
Affiliation(s)
- Xinming Tong
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Manish Ayushman
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Hung-Pang Lee
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Fan Yang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, 94305, USA; Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
6
|
Wang X, Stefanello ST, Shahin V, Qian Y. From Mechanoelectric Conversion to Tissue Regeneration: Translational Progress in Piezoelectric Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2417564. [PMID: 40434211 DOI: 10.1002/adma.202417564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 05/03/2025] [Indexed: 05/29/2025]
Abstract
Piezoelectric materials, capable of converting mechanical stimuli into electrical signals, have emerged as promising tools in regenerative medicine due to their potential to stimulate tissue repair. Despite a surge in research on piezoelectric biomaterials, systematic insights to direct their translational optimization remain limited. This review addresses the current landscape by bridging fundamental principles with clinical potential. The biomimetic basis of piezoelectricity, key molecular pathways involved in the synergy between mechanical and electrical stimulation for enhanced tissue regeneration, and critical considerations for material optimization, structural design, and biosafety is discussed. More importantly, the current status and translational quagmire of mechanisms and applications in recent years are explored. A mechanism-driven strategy is proposed for the therapeutic application of piezoelectric biomaterials for tissue repair and identify future directions for accelerated clinical applications.
Collapse
Affiliation(s)
- Xinyu Wang
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Sílvio Terra Stefanello
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Yun Qian
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| |
Collapse
|
7
|
Ely EV, Lenz KL, Paradi SG, Ack S, Behrmann A, Dunivan S, Braxton L, Liedtke W, Chen Y, Collins KH, Guilak F. Chondrocyte-Specific Knockout of Piezo1 and Piezo2 Protects Against Post-Traumatic Osteoarthritis Structural Damage and Pain in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.22.655585. [PMID: 40501713 PMCID: PMC12154791 DOI: 10.1101/2025.05.22.655585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/22/2025]
Abstract
Background Osteoarthritis (OA) is a debilitating joint disease characterized by cartilage degeneration, synovial inflammation, and bone remodeling, with limited therapeutic options targeting the underlying pathophysiology. Mechanosensitive ion channels Piezo1 and Piezo2 play crucial roles in chondrocyte responses to mechanical stress, mediating mechanotransduction pathways that influence chondrocyte survival, matrix production, and inflammatory signaling, but their distinct contributions to OA pathogenesis remain unclear. Methods Using inducible, chondrocyte-specific Aggrecan-Cre ( Acan ) mice, we investigated Piezo1 , Piezo2 , and combined Piezo1 / 2 conditional knockouts (cKOs) using the destabilization of the medial meniscus (DMM) model of post-traumatic OA in male and female mice. Pain and behavioral assessments were conducted at four time points to evaluate OA progression, while cartilage damage, bone remodeling, and synovial inflammation were assessed at the final endpoint of 28 weeks. Statistical analyses included one-way and two-way ANOVA with Tukey's multiple comparisons test. Results Piezo1 cKO delayed pain onset but ultimately exacerbated cartilage degradation and synovitis, emphasizing its dual role in protective and pathogenic mechanotransduction. While the Piezo2 cKO reduced pain and preserved activity, it failed to protect cartilage. Notably, Piezo1/2 cKO provided the greatest protection against cartilage degeneration, synovitis, and pain. Micro-computed tomography analyses revealed that Piezo2 is critical for maintaining trabecular bone integrity, with a Piezo2 cKO leading to decreased bone volume, thickness, and density, independent of injury. Piezo2 cKO also reduced normal meniscal ossification that occurs with age in mice. In contrast, a Piezo1/2 cKO normalized most bone remodeling parameters observed in Piezo2 cKO mice but did not restore medial tibial plateau thickness, highlighting Piezo2 's essential role in bone structure. Conclusions These findings demonstrate the overlapping and compensatory roles of Piezo1 and Piezo2 in OA pathogenesis. Dual inhibition of Piezo1 and Piezo2 may offer a novel, effective therapeutic strategy targeting both structural and symptomatic aspects of the disease.
Collapse
Affiliation(s)
- Erica V. Ely
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Kristin L. Lenz
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Sophie G. Paradi
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
| | - Seth Ack
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
| | - Abraham Behrmann
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Sarah Dunivan
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
| | - Lauryn Braxton
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, North Carolina
| | - Yong Chen
- Department of Neurology, Duke University, Durham, North Carolina
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| |
Collapse
|
8
|
van Mourik M, Abinzano F, Ito K. The Regulation of Pericellular Matrix Synthesis During Articular Cartilage Tissue Engineering. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40402857 DOI: 10.1089/ten.teb.2024.0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Articular cartilage, vital to the health and functioning of joints, remains challenging to regenerate. The pericellular matrix (PCM) is critical for transducing biophysical stimuli to the articular chondrocytes (ACs) that it envelops. Given the mechanobiological sensitivity of ACs, it is pivotal in maintaining the chondrogenic phenotype and the production of extracellular matrix (ECM) during articular cartilage tissue engineering. While the maintenance of the native PCM significantly improves the quality of neocartilage, current isolation methods are limited. A solution to this challenge is facilitating ACs to regenerate their PCM. However, the regulation of PCM synthesis remains poorly understood, hindering the development of effective tissue engineering strategies. This narrative review aims to provide a comprehensive analysis of the complex interplay between extracellular cues and intracellular pathways regulating PCM synthesis during articular cartilage tissue engineering. Our analysis reveals that mechanical cues, such as material stiffness and mechanical stimulation, are the primary regulators of PCM synthesis. Additionally, the use of scaffold-free techniques potentially affects the structuring of newly created PCM. Tuning these stimuli can significantly impact the quality of the formed PCM, ultimately influencing neocartilage quality. Furthermore, we highlight intracellular mechanisms involved in the transduction of these extracellular cues, including actin polymerization, yes-associated protein and transcriptional coactivator with PDZ-binding motif localization, and transforming growth factor beta-induced Smad signaling. Although the current literature suggests the involvement of these signaling pathways in regulating the synthesis of PCM components, we found that studies investigating these processes in ACs are lacking. Elucidating the relationships between extracellular stimuli, intracellular signaling, and the expression of PCM components could provide a framework for designing new cartilage tissue engineering approaches that facilitate proper PCM synthesis. Ultimately, this can improve ECM quality and advance articular cartilage regeneration.
Collapse
Affiliation(s)
- Marloes van Mourik
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Florencia Abinzano
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
9
|
Palmer DR, Nims R, Zhang B, Guilak F. Activation of the mechanosensitive ion channels TRPV4 and PIEZO1 downregulates key regulatory systems in the chondrocyte mechanome. Connect Tissue Res 2025:1-24. [PMID: 40395084 DOI: 10.1080/03008207.2025.2498512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/25/2025] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Chondrocytes, the only native cell type in cartilage, use mechanosensitive ion channels such as Transient Receptor Potential Vanilloid 4 (TRPV4) and PIEZO1 to transduce mechanical forces into transcriptomic changes that regulate cell behavior under both physiologic and pathologic conditions. Recent work has identified and characterized the differentially expressed genes (DEGs) that are upregulated following TRPV4 or PIEZO1 activation, but the transcriptomic systems downregulated by these ion channels also represent an important aspect of the chondrocyte regulatory process that remains poorly studied. METHODS Here, we utilized previously established bulk RNAsequencing libraries to analyze the transcriptomes downregulated by activation of TRPV4 and PIEZO1 through differential gene expression analysis (using DESeq2), Gene Ontology, RT-qPCR, and Weighted Gene Correlation Network Analysis (WGCNA). RESULTS TRPV4 and PIEZO1 activations downregulated largely unique sets of DEGs, though the set of DEGs downregulated by TRPV4 exhibited a notable overlap with genes downregulated by treatment with inflammatory mediator Interleukin-1 (IL-1). The DEG set downregulated by PIEZO1 activation included genes associated with the G2/M cell cycle checkpoint, a system that checks cells for DNA damage prior to entry into mitosis, and this result was confirmed with RT-qPCR. WGCNA revealed modules of gene regulation negatively correlated with TRPV4, PIEZO1, and IL-1, outlining how these downregulated DEGs may interact to form gene regulatory networks (GRNs). CONCLUSION This study complements previous work in describing the full mechanosensitive transcriptome (or "mechanome") of differential gene expression in response to activation of mechanosensitive ion channels TRPV4 and PIEZO1 Q2 and suggests potential avenues for future therapeutic treatment design.
Collapse
Affiliation(s)
- Daniel R Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Robert Nims
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, MO, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
10
|
Dietmar HF, Weidmann PA, Alberton P, Teichwart T, Gerstner M, Renkawitz T, Vortkamp A, Aszodi A, Richter W, Diederichs S. Load activated FGFR and beta1 integrins target distinct chondrocyte mechano-response genes. Matrix Biol 2025:S0945-053X(25)00047-2. [PMID: 40379111 DOI: 10.1016/j.matbio.2025.05.002] [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: 03/18/2025] [Revised: 05/02/2025] [Accepted: 05/12/2025] [Indexed: 05/19/2025]
Abstract
In response to mechanical stimuli, chondrocytes adapt their transcriptional activity, thereby shaping the cellular mechano-response; however, it remains unclear whether the activation of cell surface receptors during mechanical loading converge in the activation of the same mechano-response genes, or whether pathway-specific genes can be defined. We aimed to determine whether load-activated FGF/FGFR signalling and β1 integrins activate ERK and control the same or distinct subsets of mechano-regulated genes. To this end, tissue-engineered neocartilage was generated from murine costal chondrocytes or human articular chondrocytes and subjected to dynamic unconfined compression with or without FGFR inhibition. To assess the role of β1 integrins, neocartilage was generated from embryonic β1 integrin-deficient or wild type costal chondrocytes. Load-activated FGFR signalling drove ERK activation in murine chondrocytes, and partially also in human chondrocytes, and mechano-response genes could be classified according to their regulation: Fosl1, Itga5, Ngf and Timp1 were regulated by load-activated FGFR depending on the developmental stage, whereas β1 integrins controlled Inhba expression. In human chondrocytes, load-activated FGFR signalling controlled expression of BMP2, PTGS2 and DUSP5, but not FOSB. We show here that the chondrocyte loading response is coordinated by concurrent activation of multiple receptors, and identified for the first time distinct target genes of activated receptors. These insights open up the opportunity to pharmacologically shape the mechano-response of chondrocytes in future studies with promising implications for the management of osteoarthritis and the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Helen F Dietmar
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Pia A Weidmann
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Paolo Alberton
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), Ludwig-Maximilians-University (LMU), Munich, Germany; Division of Hand, Plastic, and Aesthetic Surgery, LMU University Hospital, LMU Munich, Germany
| | - Terrilyn Teichwart
- Department of Developmental Biology, Center for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Matthias Gerstner
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Tobias Renkawitz
- Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Vortkamp
- Department of Developmental Biology, Center for Medical Biotechnology, University Duisburg-Essen, Essen, Germany
| | - Attila Aszodi
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Centre Munich (MUM), Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Wiltrud Richter
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Solvig Diederichs
- Experimental Orthopaedics, Research Centre for Molecular and Regenerative Orthopaedics, Department of Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
11
|
Huang Y, Prastyaningrum LL, Wang X, Xu F, Wang Z, Wang Z, Tan X, Dai G, Chen G, Gong X, Yang L. MICU1 is the nexus for Ca V3.3 regulation of mitochondrial calcium, redox balance and chondrocyte viability. Int J Biol Macromol 2025; 312:144127. [PMID: 40354861 DOI: 10.1016/j.ijbiomac.2025.144127] [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: 03/08/2025] [Revised: 04/17/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Voltage-gated calcium channels are emerging regulators of cellular homeostasis, but their molecular interplay with mitochondrial bioenergetics in chondrocytes remains poorly characterized. This study elucidates how the T-type calcium channel CaV3.3 governs mitochondrial calcium-redox coupling through structural interactions with MICU1, the regulatory subunit of the mitochondrial calcium uniporter (MCU) complex. The absence of the CaV3.3 precipitated mitochondrial ultrastructural disorganization characterized, coupled with MICU1 downregulation and consequent loss of MCU gating fidelity. Through integrated transcriptomic-proteomic profiling and live-cell imaging, we demonstrate that CaV3.3 deficiency induces pathological mitochondrial calcium influx, triggering Reactive oxygen species (ROS) overproduction and bioenergetic collapse, these metabolic derangements activated intrinsic apoptosis. Notably, lentiviral overexpression of MICU1 in CaV3.3 knockout cells restored the mitochondrial calcium set point and inhibited ROS burst, while rescued cell proliferation and inhibited apoptosis execution. Our findings establish CaV3.3 as a redox rheostat coordinating MICU1-mediated mitochondrial calcium buffering, with direct implications for cartilage matrix maintenance and osteoarthritis therapy targeting calcium-handling macromolecules.
Collapse
Affiliation(s)
- Yumengfei Huang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China; Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Lucky Laras Prastyaningrum
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xin Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Fa Xu
- Knorigene Technologies, Chongqing 400084, China
| | - Zonghan Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Zhi Wang
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Xin Tan
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Gang Dai
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China
| | - Guangxing Chen
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| | - Xiaoyuan Gong
- Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| | - Liu Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing 400044, China; Center for Joint Surgery, Intelligent Manufacturing and Rehabilitation Engineering Center, The First Affiliated Hospital of Army Medical University, Chongqing 400038, China; Chongqing Municipal Science and Technology Bureau Key Laboratory of Precision Medicine in Joint Surgery, Chongqing 400038, China; Chongqing Municipal Education Commission Key Laboratory of Joint Biology, Chongqing 400038, China.
| |
Collapse
|
12
|
Crump KB, Kanelis E, Segarra-Queralt M, Pascuet-Fontanet A, Bermudez-Lekerika P, Alminnawi A, Geris L, Alexopoulos LG, Noailly J, Gantenbein B. TNF induces catabolism in human cartilaginous endplate cells in 3D agarose culture under dynamic compression. Sci Rep 2025; 15:15849. [PMID: 40328789 PMCID: PMC12056083 DOI: 10.1038/s41598-025-00538-w] [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: 02/18/2025] [Accepted: 04/29/2025] [Indexed: 05/08/2025] Open
Abstract
Intervertebral disc (IVD) degeneration is the leading cause of low back pain in young adults, and the cartilaginous endplate (CEP) is likely to play a key role in early IVD degeneration. To elucidate the effects of pro-inflammatory cytokines on the mechanobiology of the CEP, human CEP cells were seeded into 2% agarose, dynamically compressed up to 7%, and stimulated with tumor necrosis factor (TNF). It was hypothesized that dynamic compression would be sufficient to induce anabolism, while stimulation with TNF would induce catabolism. TNF was sufficient to induce a catabolic, time-dependent response in human CEP cells through downregulation of anabolic gene expression and increased secretion of pro-inflammatory proteins associated with herniated discs, bacteria inhibition, and pain. However, 7% strain or scaffold material, agarose, may not lead to full activation of integrins and downregulation of pro-inflammatory pathways, demonstrated in part through the unchanged gene expression of integrin subunits α5 and β1.
Collapse
Affiliation(s)
- Katherine B Crump
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Exarchos Kanelis
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | | | | | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012, Bern, Switzerland
| | - Ahmad Alminnawi
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Liesbet Geris
- GIGA In Silico Medicine, University of Liège, Liège, 4000, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000, Leuven, Belgium
| | - Leonidas G Alexopoulos
- School of Mechanical Engineering, National Technical University of Athens, 15772, Zografou, Greece
- Protavio Ltd, 15341, Agia Paraskevi, Greece
| | - Jérôme Noailly
- BCN Medtech, Universitat Pompeu Fabra, 08018, Barcelona, Spain
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Faculty of Medicine, University of Bern, 3008, Bern, Switzerland.
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Faculty of Medicine, University of Bern, 3010, Bern, Switzerland.
| |
Collapse
|
13
|
Cai C, Wang M, Wang L, Guo J, Wang L, Zhang Y, Wu G, Hua B, Stuart MAC, Guo X, Cao L, Yan Z. Zwitterionic Brush-Grafted Interfacial Bio-Lubricant Evades Complement C3-Mediated Macrophage Phagocytosis for Osteoarthritis Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501137. [PMID: 40304130 DOI: 10.1002/adma.202501137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/10/2025] [Indexed: 05/02/2025]
Abstract
Administering a bio-lubricant is a promising therapeutic approach for the treatment of osteoarthritis (OA), in particular, if it can both manage symptoms and halt disease progression. However, the clearance of these bio-lubricants mediated by synovial macrophages leads to reduced therapeutic efficiency and adverse inflammatory responses. Herein, it is shown that this process is predominantly mediated by the specific binding of complement C3 (on nanoparticle) and CD11b (on macrophage). More importantly, through a systematic evaluation of various interface modifications, a macrophage-evading nanoparticle strategy is proposed, which not only minimizes friction, but also largely suppresses C3 adsorption. It involves employing a zwitterionic poly-2-methacryloyloxyethyl phosphorylcholine (PMPC) brush layer grafted from a crosslinked gelatin core. In vitro studies demonstrate that such a nanoparticle lubricant can evade macrophage phagocytosis and further prevent the pro-inflammatory M1 polarization and subsequent harmful release of cytokines. In vivo studies show that the designed PMPC brush layer effectively mitigates synovial inflammation, alleviates OA-associated pain, and protects cartilage from degeneration, thus preventing OA progression. These findings clarify the pivotal role of complement C3-mediated macrophage recognition in nanoparticles clearance and offer a promising nanoparticle design strategy to restore joint lubrication.
Collapse
Affiliation(s)
- Chuandong Cai
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Mingwei Wang
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Nijmegen, 6525 EX, The Netherlands
| | - Luman Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
- Department of Immunology, School of Basic Medical Sciences, Shanghai Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiangtao Guo
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Lipeng Wang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Yingkai Zhang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Guohao Wu
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Bingxuan Hua
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Martien A Cohen Stuart
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Physical Chemistry and Soft Matter, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Xuhong Guo
- School of Chemical Engineering, State-Key Laboratory of Chemical Engineering and Shanghai Key Laboratory of Multiphase Materials Chemical Engineering, East China University of Science and Technology, Shanghai, 200237, China
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha, 410205, China
| | - Lu Cao
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
| | - Zuoqin Yan
- Department of Orthopaedic Surgery, Zhongshan Hospital, Institute of Bone and Joint Diseases, Fudan University, Shanghai, 200032, China
- Department of Orthopaedic Surgery, Shanghai Geriatric Medical Center, 2560 Chunshen Road, Shanghai, 201104, China
| |
Collapse
|
14
|
Sun Y, Luo Z, Fu Y, Ngo T, Wang W, Wang Y, Kong Y. Primary cilia and inflammatory response: unveiling new mechanisms in osteoarthritis progression. Exp Biol Med (Maywood) 2025; 250:10490. [PMID: 40357414 PMCID: PMC12066368 DOI: 10.3389/ebm.2025.10490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Osteoarthritis (OA) is a common degenerative joint disease that can lead to chronic pain and disability. The pathogenesis of OA involves chronic low-grade inflammation, characterized by the degradation of chondrocytes, inflammation of the synovium, and systemic low-grade inflammation. This inflammatory response accelerates the progression of OA and contributes to pain and functional impairment. Primary cilia play a crucial role in cellular signal transduction and the maintenance of cartilage matrix homeostasis, and their dysfunction is closely linked to inflammatory responses. Given these roles, primary cilia may significantly contribute to the pathogenesis of OA. This review explores inflammation-associated signaling pathways in OA, including NF-κB, MAPK, JAK/STAT, and PI3K/AKT/mTOR signaling. In addition, we place particular emphasis on cilia-mediated inflammatory modulation in OA. Primary cilia mediate chondrocyte responses to mechanical loading and inflammatory cytokines via pathways including NF-κB, MAPK, TRPV4, and Hedgehog signaling. Notably, alterations in the length and incidence of primary cilia in chondrocytes during OA further underscore their potential role in disease pathogenesis. The identification of biomarkers and therapeutic targets related to primary cilia and inflammatory pathways offers new potential for the treatment and management of OA.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ying Kong
- Department of Rehabilitation, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
June RK. Articular chondrocytes transduce mechanical signals through the Wnt pathway. Osteoarthritis Cartilage 2025:S1063-4584(25)00971-9. [PMID: 40216219 DOI: 10.1016/j.joca.2025.04.002] [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: 03/25/2025] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/16/2025]
Affiliation(s)
- Ronald K June
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, MT, USA; Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, USA.
| |
Collapse
|
16
|
Harissa Z, Kim Y, Dicks AR, Steward N, Guilak F. Skeletal dysplasia-causing mutations in TRPV4 alter the chondrocyte transcriptomic response to mechanical loading. Am J Physiol Cell Physiol 2025; 328:C1135-C1149. [PMID: 40019039 DOI: 10.1152/ajpcell.01066.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 02/21/2025] [Indexed: 03/01/2025]
Abstract
Transient receptor potential vanilloid 4 (TRPV4) is a mechanosensitive ion channel highly expressed in chondrocytes that supports cartilage development and homeostasis. Mutations in the channel can cause skeletal dysplasias, including the gain-of-function mutations V620I and T89I, which lead to brachyolmia and metatropic dysplasia, respectively. These mutations suppress hypertrophic differentiation, but the mechanisms by which they alter chondrocyte response to mechanical load remain to be elucidated. To determine the effect of these mutations on chondrocyte mechanotransduction, tissue-engineered cartilage was derived from differentiated clustered regularly interspaced short palindromic repeats (CRISPR)-edited human-induced pluripotent stem cells (hiPSCs) harboring the moderate V620I or severe T89I TRPV4 mutations. Wild-type and mutant tissue-engineered hiPSC-derived cartilage contructs were subjected to compressive mechanical loading at physiological levels, and transcriptomic signatures were assessed by RNA-sequencing. Our results demonstrate that the V620I and T89I mutations diminish the mechanoresponsiveness of chondrocytes, as evidenced by reduced gene expression downstream of TRPV4 activation, including those involved in endochondral ossification. Changes in the expression of genes involved in extracellular matrix production and organization were found to contribute toward the phenotype in V620I mutant chondrocytes, whereas dysregulated retinoic acid signaling was linked to T89I, and disrupted proliferation was common to both. Our findings suggest that dysfunctional mechanotransduction due to V620I and T89I mutations in TRPV4 contribute to the developmental phenotypes, supporting TRPV4 modulation as a potential pharmacologic target.NEW & NOTEWORTHY Gain-of-function mutations in TRPV4, a mechano- and osmosensitive ion channel, are linked to skeletal dysplasias, but their effects on chondrocyte mechanotransduction remain unknown. Using human iPSCs harboring skeletal dysplasia-causing mutations, we developed and mechanically loaded tissue-engineered cartilage. Our findings show that V620I and T89I mutations reduce chondrocyte mechanoresponsiveness, evidenced by decreased gene expression downstream of TRPV4 activation, providing insight into TRPV4-related skeletal disorders and potential pharmacological targets.
Collapse
Affiliation(s)
- Zainab Harissa
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Yuseon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Amanda R Dicks
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Nancy Steward
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, United States
- Shriners Hospitals for Children-St. Louis, St. Louis, Missouri, United States
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, United States
- Center of Regenerative Medicine, Washington University, St. Louis, Missouri, United States
| |
Collapse
|
17
|
Wang Y, Lee B, Yang Z, Ho T, Ci H, Jackson B, Pushon T, Wang B, Levy J, Ho S. Chewing-Activated TRPV4/PIEZO1- HIF-1α-Zn Axes in a Rat Periodontal Complex. J Dent Res 2025; 104:398-407. [PMID: 39876056 PMCID: PMC11909774 DOI: 10.1177/00220345241294001] [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: 01/30/2025] Open
Abstract
The upstream mechanobiological pathways that regulate the downstream mineralization rates in periodontal tissues are limitedly understood. Herein, we spatially colocalized and correlated compression and tension strain profiles with the expressions of mechanosensory ion channels (MS-ion) TRPV4 and PIEZO1, biometal zinc, mitochondrial function marker (MFN2), cell senescence indicator (p16), and oxygen status marker hypoxia-inducible factor-1α (HIF-1α) in rats fed hard and soft foods. The observed zinc and related cellular homeostasis in vivo were ascertained by TRPV4 and PIEZO1 agonists and antagonists on human periodontal ligament fibroblasts ex vivo. Four-week-old male Sprague-Dawley rats were fed hard (n = 3) or soft (n = 3) foods for 4 wk (in vivo). Significant changes in alveolar socket and root shapes with decreased periodontal ligament space and increased cementum volume fraction were observed in maxillae on reduced loads (soft food). Reduced loads impaired distally localized compression-stimulated PIEZO1 and mesially localized tension-stimulated TRPV4, decreased mitochondrial function (MFN2), and increased cell senescence in mesial and distal periodontal regions. The switch in HIF-1α from hard food-distal to soft food-mesial indicated a plausible effect of shear-regulated blood and oxygen flows in the periodontal complex. Blunting or activating TRPV4 or PIEZO1 MS-ion channels by channel-specific antagonists or agonists in human periodontal ligament fibroblast cultures (in vitro) indicated a positive correlation between zinc levels and zinc transporters but not with MS-ion channel expressions. The effects of reduced chewing loads in vivo were analogous to TRPV4 and PIEZO1 antagonists in vitro. Study results collectively illustrated that tension-induced TRPV4 and compression-induced PIEZO1 activations are necessary for cell metabolism. An increased hypoxic state with reduced functional loads can be a conducive environment for cementum growth. From a practical standpoint, dose rate-controlled loads can modulate tension and compression-specific MS-ion channel activation, cellular zinc, and HIF-1α transcription. These mechanobiochemical events indicate the plausible catalytic role of biometal zinc in mineralization, periodontal maintenance, and dentoalveolar joint function.
Collapse
Affiliation(s)
- Y. Wang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - B.H. Lee
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - Z. Yang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - T.J. Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - H. Ci
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - B. Jackson
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - T. Pushon
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - B. Wang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - J. Levy
- Department of Pathology and Computational Biomedicine, Cedars Sinai, Los Angeles, CA, USA
| | - S.P. Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
- Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
18
|
Munsch AE, Evans-Pickett A, Davis-Wilson H, Pietrosimone B, Rauch CM, Roth JD, Franz JR. Limb Underloading in Walking Transmits Less Dynamic Knee Joint Contact Forces after Anterior Cruciate Ligament Reconstruction. Med Sci Sports Exerc 2025; 57:849-856. [PMID: 39809224 PMCID: PMC11908909 DOI: 10.1249/mss.0000000000003607] [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: 01/16/2025]
Abstract
INTRODUCTION Individuals with anterior cruciate ligament reconstruction (ACLR) often walk with a less dynamic vertical ground reaction force (vGRF), exemplified by a reduced first peak vGRF and elevated midstance vGRF compared with uninjured controls. However, the mechanism by which altered limb loading affects actual tibial plateau contact forces during walking remains unclear. METHODS Our purpose was to use musculoskeletal simulation to evaluate the effects of first peak vGRF biofeedback on bilateral tibiofemoral contact forces relevant to the development of post-traumatic osteoarthritis in 20 individuals with ACLR. We hypothesized that reduced first peak vGRF would produce less dynamic tibial plateau contact forces during walking in individuals with ACLR. RESULTS As the pivotal outcome from this study, and in support of our hypothesis, we found that less dynamic vGRF profiles in individuals with ACLR-observations that have associated in prior studies with more cartilage breakdown serum biomarkers and reduced proteoglycan density-are accompanied by less dynamic tibiofemoral joint contact forces during walking. CONCLUSION We conclude that more sustained limb-level loading, a phenotype that associates with worse knee joint health outcomes after ACLR and was prescribed herein using biofeedback, alters the loading profile and magnitude of force applied to tibiofemoral cartilage.
Collapse
Affiliation(s)
- Amanda E. Munsch
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC
| | | | | | - Brian Pietrosimone
- Department of Exercise and Sport Science, UNC Chapel Hill, Chapel Hill, NC
| | - Carly M. Rauch
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC
| | - Joshua D. Roth
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI
| | - Jason R. Franz
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC
| |
Collapse
|
19
|
Guan M, Han X, Liao B, Han W, Chen L, Zhang B, Peng X, Tian Y, Xiao G, Li X, Kuang L, Zhu Y, Bai D. LIPUS Promotes Calcium Oscillation and Enhances Calcium Dependent Autophagy of Chondrocytes to Alleviate Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413930. [PMID: 40013941 PMCID: PMC12021083 DOI: 10.1002/advs.202413930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/02/2025] [Indexed: 02/28/2025]
Abstract
Osteoarthritis (OA) is a degenerative disease which places an enormous burden on society, effective treatments are still limited. As a non-invasive and safe physical therapy, low-intensity pulsed ultrasound (LIPUS) can alleviate OA progression, but the underlying mechanism is not fully understood, especially the mechanical transduction between LIPUS and the organism. In this pioneering study, the biomechanical effects of LIPUS on living mice chondrocytes and living body zebrafish are investigate by using fluorescence imaging technology, to dynamically "visualize" its invisible mechanical stimuli in the form of calcium oscillations. It is also confirmed that LIPUS maintains cartilage homeostasis by promoting chondrocyte autophagy in a calcium-dependent manner. In addition, chondrocyte ion channels are screened by scRNA-seq and confirm that the mechanosensitive ion channel transient receptor potential vanilloid 4 (TRPV4) mediated the biological effects of LIPUS on chondrocytes. Finally, it is found that a combination of pharmacologically induced and LIPUS-induced Ca2+ influx in chondrocytes enhances the cartilage-protective effect of LIPUS, which may provide new insights for optimizing LIPUS in the treatment of OA.
Collapse
Affiliation(s)
- Mengtong Guan
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaoyu Han
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| | - Bo Liao
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Wang Han
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Lin Chen
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Bin Zhang
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Xiuqin Peng
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Yu Tian
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Gongyi Xiao
- Department of OrthopedicsChonggang General HospitalChongqing400000China
| | - Xinhe Li
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| | - Liang Kuang
- Center of Bone Metabolism and repair laboratory for Prevention and rehabilitation of Training injuries State Key laboratory of Trauma Burns and combined injury Trauma centerResearch Institute of Surgery Daping Hospital Army Medical University (Third Military Medical University)Chongqing400000China
| | - Ying Zhu
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Dingqun Bai
- Department of Rehabilitation MedicineKey Laboratory of Physical Medicine and Precision Rehabilitation of Chongqing Municipal Health CommissionThe First Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqing400016China
| |
Collapse
|
20
|
Faure LM, Venturini V, Roca-Cusachs P. Cell compression - relevance, mechanotransduction mechanisms and tools. J Cell Sci 2025; 138:jcs263704. [PMID: 40145202 DOI: 10.1242/jcs.263704] [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: 03/28/2025] Open
Abstract
From border cell migration during Drosophila embryogenesis to solid stresses inside tumors, cells are often compressed during physiological and pathological processes, triggering major cell responses. Cell compression can be observed in vivo but also controlled in vitro through tools such as micro-channels or planar confinement assays. Such tools have recently become commercially available, allowing a broad research community to tackle the role of cell compression in a variety of contexts. This has led to the discovery of conserved compression-triggered migration modes, cell fate determinants and mechanosensitive pathways, among others. In this Review, we will first address the different ways in which cells can be compressed and their biological contexts. Then, we will discuss the distinct mechanosensing and mechanotransducing pathways that cells activate in response to compression. Finally, we will describe the different in vitro systems that have been engineered to compress cells.
Collapse
Affiliation(s)
- Laura M Faure
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Valeria Venturini
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain
- University of Barcelona (UB), 08036 Barcelona, Spain
| |
Collapse
|
21
|
Nieuwstraten J, Riester R, Hofmann UK, Guilak F, Danalache M. Matrix metalloproteinases accelerate pericellular matrix breakdown and disrupt mechanotransduction in osteoarthritis. Acta Biomater 2025; 195:73-82. [PMID: 39956307 DOI: 10.1016/j.actbio.2025.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 01/25/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
The pericellular matrix (PCM) is a specialized, narrow matrix surrounding each chondrocyte in articular cartilage, together constituting the chondron - the fundamental metabolic and functional unit of cartilage. The PCM plays a vital role in mediating biomechanical and biochemical signals essential for chondrocyte function. In osteoarthritis (OA), a chronic joint disorder characterized by progressive cartilage degradation, the PCM is one of the earliest sites of catabolic degradation, primarily driven by matrix metalloproteinases (MMPs). This study aims to investigate the functional relationship between PCM degradation and chondrocyte mechanosignaling, with an emphasis on MMP-driven changes in mechanotransduction in osteoarthritic cartilage. Human chondrons (N = 64) were incubated with MMP-2, MMP-3, and MMP-7, and structural changes were assessed histologically by evaluating perlecan and collagen type VI. Cellular elasticity was measured using atomic force microscopy (AFM), and mechanically evoked intracellular Ca2+ transients were assessed via AFM single-cell indentations (500 nN). All three MMPs induced pronounced catabolic effects on the PCM structure, showing distinct impacts on collagen type VI and perlecan, as well as on the biomechanical properties (p < 0.001). MMP-driven alterations in PCM integrity significantly reduced the Ca2+ transients of chondrons in response to mechanical stimuli (p < 0.001). While TRPV4 activation was elevated in intact chondrons, PIEZO channels were involved in mechanotransduction in both healthy and MMP-treated chondrons. In osteoarthritic stages, the mechanotransduction dynamics shifted significantly towards PIEZO channels. This study elucidates the interplay between MMP-mediated PCM degradation, structural-functional dynamics, and chondrocyte mechanotransduction, underscoring the critical role of the PCM in maintaining normal chondrocyte functionality and mechanosensing. STATEMENT OF SIGNIFICANCE: Osteoarthritis (OA) is a prevalent degenerative joint disease affecting millions worldwide. Central to its pathology is the degradation of the pericellular matrix (PCM) by matrix metalloproteinases (MMPs), which disrupts chondrocyte mechanotransduction, altering cellular responses to mechanical stimuli. This study explores the impact of MMP-2, MMP-3, and MMP-7 on PCM structure and chondrocyte mechanosensing. Our results reveal that MMP-induced degradation significantly compromises PCM structural integrity, leading to altered mechanotransduction dynamics in chondrocytes. Degradation specifically redirects the primary function of ion channels from TRPV4 to PIEZO channels in cells impacted by MMPs. This highlights the interplay between MMP-mediated PCM degradation, chondrocyte mechanotransduction and as thus structural-functional dynamics, underscoring the critical role of the PCM in maintaining normal chondrocyte functionality and mechanosensing.
Collapse
Affiliation(s)
- Jule Nieuwstraten
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany
| | - Rosa Riester
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany
| | - Ulf Krister Hofmann
- Department of Orthopedic, Trauma, and Reconstructive Surgery, Division of Arthroplasty, RWTH Aachen University Hospital, Pauwelsstraße 30, D-52074 Aachen, Germany
| | - Farshid Guilak
- Department of Orthopedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children, St. Louis, MO 63110, USA
| | - Marina Danalache
- Laboratory of Cell Biology, Department of Orthopedic Surgery, University Hospital of Tübingen, Waldhörnlestraße 22, D-7207 Tübingen, Germany.
| |
Collapse
|
22
|
Liu X, Zhang M, Wang P, Zheng K, Wang X, Xie W, Pan X, Shen R, Liu R, Ding J, Wei Q. Nanoscale distribution of bioactive ligands on biomaterials regulates cell mechanosensing through translocation of actin into the nucleus. Proc Natl Acad Sci U S A 2025; 122:e2501264122. [PMID: 40042901 PMCID: PMC11912452 DOI: 10.1073/pnas.2501264122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cells respond to adhesive ligands such as arginine-glycine-aspartate (RGD) through integrins, which regulates cellular activities via influencing cytoskeleton assembly. Herein, we report that the nanoscale distribution of active ligands on biomaterials regulates cells through not only cytoplasmic tension but also nuclear tension. This is particularly related to translocation of actin into nucleus and highlighted in our interpretation of an "abnormal" phenomenon that large RGD nanospacing (>70 nm) disassembles integrin clusters, inhibits cell adhesion, but promotes osteogenic differentiation of mesenchymal stem cells. Our studies reveal that the unstable adhesion at the 150 nm RGD distance increases actin dynamics, resulting in the nuclear translocation of globular (G) actin. The compartment polymerization of more G-actins to filamentous actins in nucleus increases nuclear tension, facilitating transcription activity and releasing calcium ions from the endoplasmic reticulum. This noncanonical mechanotransduction process sheds insight into mechanotransduction pertinent to cell-material interactions.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan250012, China
| | - Man Zhang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Biomedical Engineering, Sichuan University, Chengdu610065, China
| | - Peng Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
- Sauvage Laboratory for Smart Materials, School of Integrated Circuits, Harbin Institute of Technology, Shenzhen518055, China
| | - Kaikai Zheng
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Xinlei Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Wenyan Xie
- National Key Laboratory of Biotherapy, Sichuan University, Chengdu610041, China
| | - Xiaokai Pan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| | - Runjia Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
| | - Qiang Wei
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai200438, China
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu610065, China
| |
Collapse
|
23
|
Matsuo T, Takeoka Y, Yurube T, Tsujimoto T, Kanda Y, Miyazaki K, Ohnishi H, Ryu M, Kumagai N, Kuroshima K, Hiranaka Y, Kuroda R, Kakutani K. Transient Receptor Potential Vanilloid 4 Knockdown Decreases Extracellular Matrix Synthesis via Autophagy Suppression in the Rat Intervertebral Disc. JOR Spine 2025; 8:e70046. [PMID: 39963549 PMCID: PMC11832302 DOI: 10.1002/jsp2.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/02/2024] [Accepted: 12/31/2024] [Indexed: 02/20/2025] Open
Abstract
Background Transient receptor potential vanilloid 4 (TRPV4) has been identified as a Ca2+-permeable channel and is activated under physiological mechanical stimulation in disc nucleus pulposus (NP) cells. Meanwhile, the Ca2+-dependent AMP-activated protein kinase (AMPK)/mTOR pathway activates autophagy in notochordal cells. We hypothesized that TRPV4 is involved in the maintenance of intradiscal homeostasis via autophagy. Our objective was to elucidate the role of TRPV4 in extracellular matrix (ECM) metabolism and autophagy in the rat intervertebral disc through a loss-of-function study with the RNA interference (RNAi) technique. Methods In vitro study: Small interfering RNA (siRNA) was applied to knockdown TRPV4 by the reverse transfection method in rat disc NP cells. Expression of TRPV4, AMPK/mTOR pathway-related markers, and autophagy markers were measured by Western blotting (WB). Next, ECM metabolism was assessed under serum starvation and/or proinflammatory interleukin-1 beta (IL-1β) stimulation. In vivo study: TRPV4 and control siRNAs were injected into rat discs. To confirm in vivo transfection, WB for TRPV4 was conducted in rat disc NP-tissue protein extracts 2, 28, and 56 days after injection. Furthermore, 24-h temporary static compression-induced disruption of TRPV4 siRNA-injected discs was observed by radiography, histomorphology, and immunofluorescence. Results In vitro study: In disc cells, three different TRPV4 siRNAs consistently suppressed autophagy with TRPV4 protein knockdown (mean 33.2% [95% CI: -50.8, -15.5], 44.1% [-61.7, -26.4], 58.3% [-76.0, -40.7]). ECM metabolism was significantly suppressed by TRPV4 RNAi under proinflammatory IL-1β stimulation. In vivo study: The WB displayed sustained decreases in TRPV4 protein expression 2, 28, and 56 days after injection. Under the loaded condition, TRPV4 siRNA-injected discs presented radiographic height loss ([-31.7, -7.75]), histomorphological damage ([0.300, 4.70]), and immunofluorescent suppression of autophagy ([1.61, 20.5]) and ECM metabolism ([-25.2, -6.41]) compared to control siRNA-injected discs at 56 days. Conclusions The TRPV4 could be a therapeutic target for intervertebral disc diseases via modulating autophagy.
Collapse
Affiliation(s)
- Tomoya Matsuo
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yoshiki Takeoka
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Takashi Yurube
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Takeru Tsujimoto
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yutaro Kanda
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Kunihiko Miyazaki
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Hiroki Ohnishi
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Masao Ryu
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Naotoshi Kumagai
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Kohei Kuroshima
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Yoshiaki Hiranaka
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Ryosuke Kuroda
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| | - Kenichiro Kakutani
- Department of Orthopaedic SurgeryKobe University Graduate School of MedicineKobeJapan
| |
Collapse
|
24
|
Schöffl VR, Lutter C, Lang HC, Perl M, Moser O, Simon M. Efficacy of a new treatment algorithm for capsulitis of the fingers in rock climbers. Front Sports Act Living 2025; 7:1497110. [PMID: 39902135 PMCID: PMC11788292 DOI: 10.3389/fspor.2025.1497110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/02/2025] [Indexed: 02/05/2025] Open
Abstract
Background Although finger joint capsulitis has been described among the most frequent injuries in climbers, no clinical studies on treatment strategies and outcomes are available. Study design Prospective case series study. Methods Between 2015 and 2018 we prospectively treated 50 patients (38 male, 12 female) with a total number of 69 independent finger joint capsulitis according to a clinic specific treatment regimen and evaluated the outcome retrospectively. Therapy consisted of either conservative management, steroid injections, radiosynoviorthesis or a combination depending on the treatment regimen, prior therapy and timeline of symptoms. Outcomes were assessed using visual analogue scale (VAS), Buck-Gramcko score and a climbing specific outcome score with secondary patient recall. Results The proximal interphalangeal joint of the middle finger was the most commonly affected joint, and there was no correlation with osteoarthritis. All climbers returned to sport within 12 months. The majority were able to maintain their level of performance after injury and the difference in climbing level before and after injury was not statistically significant (p = 0.22). The total time spent climbing was significantly less after the injury than before the injury (p < 0.001). The Buck-Gramcko score showed excellent results. The overall functional outcome was good to very good with a mean score of 1.6 ± 0.7, as was the climbing specific score of 1.7 ± 0.9. Pain was significantly less after treatment than before (p < 0.001). Conclusion Good to very good functional and sport-specific outcomes were seen with the stage-specific treatment regimen presented, allowing all patients studied to resume climbing. A better understanding of the underlying pathogenesis is essential in order to better assess long-term progress.
Collapse
Affiliation(s)
- Volker Rainer Schöffl
- Department of Traumaand Orthopedic Surgery, Klinikum Bamberg, Bamberg, Germany
- Department of Trauma and Orthopedic Surgery, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
- Section of Wilderness Medicine, Department of Emergency Medicine, University of Colorado School of Med., Denver, CO, United States
- School of Health, Leeds Becket University, Leeds, United Kingdom
- Division of Exercise Physiology and Metabolism, Department of Sport Science, University of Bayreuth, Bavaria, Germany
| | - Christoph Lutter
- School of Health, Leeds Becket University, Leeds, United Kingdom
- Department of Orthopedics, University Medical Center, Rostock, Germany
| | - Hans-Christoph Lang
- Department of Traumaand Orthopedic Surgery, Klinikum Bamberg, Bamberg, Germany
| | - Mario Perl
- Department of Trauma and Orthopedic Surgery, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| | - Othmar Moser
- Division of Exercise Physiology and Metabolism, Department of Sport Science, University of Bayreuth, Bavaria, Germany
| | - Michael Simon
- Department of Trauma and Orthopedic Surgery, Friedrich-Alexander University Erlangen-Nuremberg, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
25
|
Marushack GK, Savadipour A, Tang R, Garcia-Castorena JM, Rashidi N, Nims RJ, Harasymowicz NS, Kim YS, Guilak F. Polyunsaturated fatty acids suppress PIEZO ion channel mechanotransduction in articular chondrocytes. FASEB J 2025; 39:e70290. [PMID: 39786170 DOI: 10.1096/fj.202400544rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Osteoarthritis (OA) is characterized by articular cartilage degeneration, leading to pain and loss of joint function. Recent studies have demonstrated that omega-3 (ω3) polyunsaturated fatty acid (PUFA) supplementation can decrease injury-induced OA progression in mice fed a high-fat diet. Furthermore, PUFAs have been shown to influence the mechanical properties of chondrocyte membranes, suggesting that alterations in mechanosensitive ion channel signaling could contribute to the mechanism by which ω3 PUFAs decreased OA pathogenesis. Here, we hypothesized that PUFAs may alter mechanical signaling through PIEZO1 (activated by changes in membrane tension) and TRPV4 (activated by physiologic mechano-osmotic signals), as these mechanosensitive cation channels have been shown to influence OA progression. Our results demonstrated that PUFAs reduced chondrocyte sensitivity to single-cell mechanical compression and to pharmacologic agonists of PIEZO1 and TRPV4, with ω3 PUFAs having the most significant effects overall. We also found that supplementation with ω6 PUFA linoleic acid (LA) altered the biophysical properties of chondrocytes, as evidenced by increased intracellular lipid droplet formation and more rapid membrane rupture in response to hypo-osmotic shock, suggesting that LA increases chondrocyte membrane susceptibility to damage. Our findings underscore the differential impacts of specific PUFAs on chondrocyte signaling and membrane properties and provide important considerations in the development of nutritional interventions to prevent or treat OA.
Collapse
Affiliation(s)
- Gabrielle K Marushack
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alireza Savadipour
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Ruhang Tang
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jaquelin M Garcia-Castorena
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Neda Rashidi
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| | - Robert J Nims
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Natalia S Harasymowicz
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Yu Seon Kim
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Mechanical Engineering, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
26
|
Suito H, Minamizono W, Yashima N, Matsunaga H, Fujikawa K, Ohsako M. Effect of load reduction on the calcification of rat tibial tuberosity: Focus on calcification factors and chondrocyte mechanosensors. J Anat 2025; 246:148-159. [PMID: 39234669 PMCID: PMC11684375 DOI: 10.1111/joa.14128] [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: 09/06/2023] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
The tibial tuberosity has a superficial patellar tendon-embedded portion and a deep uncalcified cartilage portion. Suppressed calcification of the tibial tuberosity leads to Osgood-Schlatter disease. The tibial tuberosity calcifies with age; load reduction degrades the cartilage matrix and promotes calcification, suggesting that reduced mechanical stimulation of the tibial tuberosity promotes calcification. However, this is yet to be clarified. Therefore, in this study, we aimed to investigate the effects of mechanical stimulation reduction on the tibial tuberosity tissue structure and calcification mechanism. Specifically, we examined the effect of load reduction on tibial tuberosity calcification in 20 male 7-week-old Wistar rats classified into two groups: hind-limb suspension (HS, n = 10) and control (CO, n = 10). We observed superficial and deep tibial tuberosities in both groups. The tibial tuberosity in the HS group had narrower areas of deep portions than did those in the CO group (p = 0.000539), and immature bone tissue and cartilage tissue were observed in the HS group. Enpp1 expression did not significantly differ between the groups (p = 0.804). In contrast, Alpl (p = 0.001) and Mmp3 (p = 0.006) expression increased whereas Timp3 expression decreased (p = 0.002) in the HS group. Thus, these results showed a maturing of bone ossification, and this gene expression trend was similar to that observed in a murine join instability model of osteoarthritis with articular cartilage calcification and ossification. The HS tibial tuberosity also showed immature bone tissue. In conclusion, reduced mechanical stimulation caused tibial tuberosity calcification and pathological changes. These findings highlight the importance of optimal exercise to avoid premature pathological structural changes in bones and joints.
Collapse
Affiliation(s)
- Hirai Suito
- Life Innovation Institute, Toyo University, Tokyo, Japan
- Department of Anatomy, Teikyo University of School of Medicine, Tokyo, Japan
| | | | - Nao Yashima
- Graduate School of Health Sports Science, Toyo University, Tokyo, Japan
| | - Hiroya Matsunaga
- Graduate School of Health Sports Science, Toyo University, Tokyo, Japan
| | - Kaoru Fujikawa
- Department of Oral Anatomy and Developmental Biology, Showa University School of Dentistry, Tokyo, Japan
| | - Masafumi Ohsako
- Graduate School of Health Sports Science, Toyo University, Tokyo, Japan
| |
Collapse
|
27
|
Wang Y, He J, Li M, Xu J, Yang H, Zhang Y. Abamectin at environmentally relevant concentrations impairs bone development in zebrafish larvae. Comp Biochem Physiol C Toxicol Pharmacol 2025; 287:110039. [PMID: 39265967 DOI: 10.1016/j.cbpc.2024.110039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/30/2024] [Accepted: 09/08/2024] [Indexed: 09/14/2024]
Abstract
Abamectin (ABM) is a widely used pesticide in agriculture and veterinary medicine, which primarily acts by disrupting the neurological physiology of pests, leading to their paralysis and death. Its extensive application has resulted in contamination of many natural water bodies. While the adverse effects of ABM on the growth and development of non-target organisms are well documented, its impact on bone development remains inadequately studied. The present study aimed to investigate the effects of environmentally relevant concentrations of ABM (1, 5, 25 μg/L) on early bone development in zebrafish. Our results indicated that ABM significantly affected both cartilage and bone development of zebrafish larvae, accompanied by dose-dependent increase in deformity and mortality rates, as well as exacerbated apoptosis. ABM exposure led to deformities in the ceratobranchial (cb) and hyosymplectic (hs), accompanied by significant increases in the length of the palatoquadrate (pq). Furthermore, significant decreases in the CH-CH angle, Meckel's-Meckel's angle, and Meckel's-PQ angle were noted. Even at the safe concentration of 5 μg/L (1/10 of the 96 h LC50), ABM delayed the process of bone mineralization in zebrafish larvae. Real-time fluorescent quantitative PCR results demonstrated that ABM induced differential gene expression associated with cartilage and bone development in zebrafish. Thus, this study provides preliminary insights into the effects and molecular mechanisms underlying ABM's impact on the bone development of zebrafish larvae and offers new evidence for a better understanding of its toxicity.
Collapse
Affiliation(s)
- Yuting Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiawen He
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou 225009, China
| | - Min Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Jiawen Xu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Hui Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
| | - Yingying Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
28
|
Ma Z, Chawla S, Lan X, Zhou E, Mulet-Sierra A, Kunze M, Sommerfeldt M, Adesida AB. Functional heterogeneity of meniscal fibrochondrocytes and microtissue models is dependent on modality of fibrochondrocyte isolation. Cell Prolif 2025; 58:e13735. [PMID: 39377189 DOI: 10.1111/cpr.13735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/13/2024] [Accepted: 08/05/2024] [Indexed: 10/09/2024] Open
Abstract
Collagenase digestion (d) and cellular outgrowth (og) are the current modalities of meniscus fibrochondrocytes (MFC) isolation for bioengineering and mechanobiology-related studies. However, the impact of these modalities on study outcomes is unknown. Here, we show that og- and d-isolated MFC have distinct proliferative capacities, transcriptomic profiles via RNA sequencing (RNAseq), extracellular matrix (ECM)-forming, and migratory capacities. Our data indicate that microtissue pellet models developed from og-isolated MFC display a contractile phenotype with higher expressions of alpha-smooth muscle actin (ACTA2) and transgelin (TAGLN) and are mechanically stiffer than their counterparts from d-MFC. Moreover, we introduce a novel method of MFC isolation designated digestion-after-outgrowth (dog). The transcriptomic profile of dog-MFC is distinct from d- and og-MFC, including a higher expression of mechanosensing caveolae-associated caveolin-1 (CAV1). Additionally, dog-MFC were superior chondrogenically and generated larger-size microtissue pellet models containing a higher frequency of smaller collagen fibre diameters. Thus, we demonstrate that the modalities of MFC isolation influence the downstream outcomes of bioengineering and mechanobiology-related studies.
Collapse
Affiliation(s)
- Zhiyao Ma
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shikha Chawla
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyi Lan
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Eva Zhou
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Aillette Mulet-Sierra
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Melanie Kunze
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Mark Sommerfeldt
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Adetola B Adesida
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
29
|
Sambale M, Lively S, Espin-Garcia O, Potla P, Pastrello C, Bödecker S, Wessendorf L, Kleimann S, Paruzel P, Asgarian R, Tosun A, Intemann J, Bertrand J, Dell’Accio F, Kapoor M, Pap T, Sherwood J. TRPC1 links calcium signaling to cellular senescence in the protection against posttraumatic osteoarthritis. JCI Insight 2024; 10:e182103. [PMID: 39718827 PMCID: PMC11948585 DOI: 10.1172/jci.insight.182103] [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: 04/15/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
Transient receptor potential channel 1 (TRPC1) is a widely expressed mechanosensitive ion channel located within the endoplasmic reticulum membrane, crucial for refilling depleted internal calcium stores during activation of calcium-dependent signaling pathways. Here, we have demonstrated that TRPC1 activity is protective within cartilage homeostasis in the prevention of cellular senescence-associated cartilage breakdown during mechanical and inflammatory challenge. We revealed that TRPC1 loss is associated with early stages of osteoarthritis (OA) and plays a nonredundant role in calcium signaling in chondrocytes. Trpc1-/- mice subjected to destabilization of the medial meniscus-induced OA developed a more severe OA phenotype than WT controls. During early OA development, Trpc1-/- mice displayed an increased chondrocyte survival rate; however, remaining cells displayed features of senescence including p16INK4a expression and decreased Sox9. RNA-Seq identified differentially expressed genes related to cell number, apoptosis, and extracellular matrix organization. Trpc1-/- chondrocytes exhibited accelerated dedifferentiation, while demonstrating an increased susceptibility to cellular senescence. Targeting the mechanism of TRPC1 activation may be a promising therapeutic strategy in OA prevention.
Collapse
Affiliation(s)
- Meike Sambale
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Starlee Lively
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, and
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Osvaldo Espin-Garcia
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, and
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
- Department of Epidemiology and Biostatistics, University of Western Ontario, London, Ontario, Canada
- Dalla Lana School of Public Health and Department of Statistical Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biostatistics, University Health Network, Toronto, Ontario, Canada
| | - Pratibha Potla
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, and
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Chiara Pastrello
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, and
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Sarah Bödecker
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Linda Wessendorf
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Simon Kleimann
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Peter Paruzel
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Rojiar Asgarian
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Alexandra Tosun
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Johanna Intemann
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Jessica Bertrand
- Department of Orthopaedic Surgery, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Francesco Dell’Accio
- Experimental Medicine and Rheumatology, Queen Mary University of London, London, United Kingdom
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, and
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Thomas Pap
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Joanna Sherwood
- Institute of Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| |
Collapse
|
30
|
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
|
31
|
Sánchez-Hernández R, Benítez-Angeles M, Hernández-Vega AM, Rosenbaum T. Recent advances on the structure and the function relationships of the TRPV4 ion channel. Channels (Austin) 2024; 18:2313323. [PMID: 38354101 PMCID: PMC10868539 DOI: 10.1080/19336950.2024.2313323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/18/2024] [Indexed: 02/16/2024] Open
Abstract
The members of the superfamily of Transient Receptor Potential (TRP) ion channels are physiologically important molecules that have been studied for many years and are still being intensively researched. Among the vanilloid TRP subfamily, the TRPV4 ion channel is an interesting protein due to its involvement in several essential physiological processes and in the development of various diseases. As in other proteins, changes in its function that lead to the development of pathological states, have been closely associated with modification of its regulation by different molecules, but also by the appearance of mutations which affect the structure and gating of the channel. In the last few years, some structures for the TRPV4 channel have been solved. Due to the importance of this protein in physiology, here we discuss the recent progress in determining the structure of the TRPV4 channel, which has been achieved in three species of animals (Xenopus tropicalis, Mus musculus, and Homo sapiens), highlighting conserved features as well as key differences among them and emphasizing the binding sites for some ligands that play crucial roles in its regulation.
Collapse
Affiliation(s)
- Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Miguel Benítez-Angeles
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Ana M. Hernández-Vega
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, División Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico, Mexico
| |
Collapse
|
32
|
Rudzitis CN, Lakk M, Singh A, Redmon SN, Kirdajova D, Tseng YT, De Ieso ML, Stamer WD, Herberg S, Križaj D. TRPV4 overactivation enhances cellular contractility and drives ocular hypertension in TGFβ2 overexpressing eyes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.05.622187. [PMID: 39574569 PMCID: PMC11580928 DOI: 10.1101/2024.11.05.622187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
The risk for developing primary open-angle glaucoma (POAG) correlates with the magnitude of ocular hypertension (OHT) and the concentration of transforming growth factor-β2 (TGFβ2) in the aqueous humor. Effective treatment of POAG requires detailed understanding of interaction between pressure sensing mechanisms in the trabecular meshwork (TM) and biochemical risk factors. Here, we employed molecular, optical, electrophysiological and tonometric strategies to establish the role of TGFβ2 in transcription and functional expression of mechanosensitive channel isoforms alongside studies of TM contractility in biomimetic hydrogels, and intraocular pressure (IOP) regulation in a mouse model of TGFβ2 -induced OHT. TGFβ2 upregulated expression of TRPV4 and PIEZO1 transcripts and time-dependently augmented functional TRPV4 activation. TRPV4 activation induced TM contractility whereas pharmacological inhibition suppressed TGFβ2-induced hypercontractility and abrogated OHT in eyes overexpressing TGFβ2. Trpv4-deficient mice resisted TGFβ2-driven increases in IOP. Nocturnal OHT was not additive to TGFβ-evoked OHT. Our study establishes the fundamental role of TGFβ as a modulator of mechanosensing in nonexcitable cells, identifies TRPV4 channel as the final common mechanism for TM contractility and circadian and pathological OHT and offers insights future treatments that can lower IOP in the sizeable cohort of hypertensive glaucoma patients that resist current treatments.
Collapse
Affiliation(s)
- Christopher N. Rudzitis
- Department of Ophthalmology and Visual Sciences
- Department of Neurobiology, University of Utah, Salt Lake City, UT
| | - Monika Lakk
- Department of Ophthalmology and Visual Sciences
| | - Ayushi Singh
- Department of Ophthalmology and Visual Sciences
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY
| | | | | | | | - Michael L. De Ieso
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC
| | - W. Daniel Stamer
- Department of Ophthalmology, Duke Eye Center, Duke University, Durham, NC
| | - Samuel Herberg
- Department of Ophthalmology and Visual Sciences
- Department of Cell and Developmental Biology, SUNY Upstate Medical University, Syracuse, NY
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY
| | - David Križaj
- Department of Ophthalmology and Visual Sciences
- Department of Neurobiology, University of Utah, Salt Lake City, UT
- Department of Bioengineering, University of Utah, Salt Lake City, UT
| |
Collapse
|
33
|
Tsadaris SA, Komatsu DE, Grubisic V, Ramos RL, Hadjiargyrou M. A GCaMP reporter mouse with chondrocyte specific expression of a green fluorescent calcium indicator. Bone 2024; 188:117234. [PMID: 39147354 PMCID: PMC11392458 DOI: 10.1016/j.bone.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 08/17/2024]
Abstract
One of the major processes occurring during the healing of a fractured long bone is chondrogenesis, leading to the formation of the soft callus, which subsequently undergoes endochondral ossification and ultimately bridges the fracture site. Thus, understanding the molecular mechanisms of chondrogenesis can enhance our knowledge of the fracture repair process. One such molecular process is calciun (Ca++) signaling, which is known to play a critical role in the development and regeneration of multiple tissues, including bone, in response to external stimuli. Despite the existence of various mouse models for studying Ca++ signaling, none of them were designed to specifically examine the skeletal system or the various musculoskeletal cell types. As such, we generated a genetically engineered mouse model that is specific to cartilage (crossed with Col2a1 Cre mice) to study chondrocytes. Herein, we report on the characterization of this transgenic mouse line using conditional expression of GCaMP6f, a Ca++-indicator protein. Specifically, this mouse line exhibits increased GCaMP6f fluorescence following Ca++ binding in chondrocytes. Using this model, we show real-time Ca++ signaling in embryos, newborn and adult mice, as well as in fracture calluses. Further, robust expression of GCaMP6f in chondrocytes can be easily detected in embryos, neonates, adults, and fracture callus tissue sections. Finally, we also report on Ca++ signaling pathway gene expression, as well as real-time Ca++ transient measurements in fracture callus chondrocytes. Taken together, these mice provide a new experimental tool to study chondrocyte-specific Ca++ signaling during skeletal development and regeneration, as well as various in vitro perturbations.
Collapse
Affiliation(s)
- Sotirios A Tsadaris
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA
| | - David E Komatsu
- Department of Orthopaedics and Rehabilitation, Stony Brook University, Stony Brook, NY, USA
| | - Vladimir Grubisic
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA; Center for Biomedical Innovation, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Raddy L Ramos
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, USA
| | - Michael Hadjiargyrou
- Department of Biological & Chemical Sciences, New York Institute of Technology, Old Westbury, NY, USA.
| |
Collapse
|
34
|
Mora KE, Mlawer SJ, Loiselle AE, Buckley MR. The Micromechanical Environment of the Impinged Achilles Tendon. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401015. [PMID: 38966889 DOI: 10.1002/smll.202401015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/13/2024] [Indexed: 07/06/2024]
Abstract
Although tendon predominantly experiences longitudinal tensile forces, transverse forces due to impingement from bone are implicated in both physiological and pathophysiological processes. However, prior studies have not characterized the micromechanical strain environment in the context of tendon impingement. To address this knowledge gap, mouse hindlimb explants are imaged on a multiphoton microscope, and image stacks of the same population of tendon cells are obtained in the Achilles tendon before and after dorsiflexion-induced impingement by the heel bone. Based on the acquired images, multiaxial strains are measured at the extracellular matrix (ECM), pericellular matrix (PCM), and cell scales. Impingement generated substantial transverse compression at the matrix-scale, which led to longitudinal stretching of cells, increased cell aspect ratio, and enormous volumetric compression of the PCM. These experimental results are corroborated by a finite element model, which further demonstrated that impingement produces high cell surface stresses and strains that greatly exceed those brought about by longitudinal tension. Moreover, in both experiments and simulations, impingement-generated microscale stresses and strains are highly dependent on initial cell-cell gap spacing. Identifying factors that influence the microscale strain environment generated by impingement could contribute to a more mechanistic understanding of impingement-induced tendinopathies.
Collapse
Affiliation(s)
- Keshia E Mora
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, 14642, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Samuel J Mlawer
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
| | - Alayna E Loiselle
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Mark R Buckley
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
- Center for Musculoskeletal Research, Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
35
|
Bian X, Liu X, Zhou M, Tang H, Wang R, Ma L, He G, Xu S, Wang Y, Tan J, Tang K, Guo L. Mechanical stimulation promotes fibrochondrocyte proliferation by activating the TRPV4 signaling pathway during tendon-bone insertion healing: CCN2 plays an important regulatory role. BURNS & TRAUMA 2024; 12:tkae028. [PMID: 39429645 PMCID: PMC11491146 DOI: 10.1093/burnst/tkae028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 10/22/2024]
Abstract
Background We previously confirmed that mechanical stimulation is an important factor in the repair of tendon-bone insertion (TBI) injuries and that mechanoreceptors such as transient receptor potential ion-channel subfamily V member 4 (TRPV4; also known as transient receptor potential vanilloid 4) are key to transforming mechanical stimulation into intracellular biochemical signals. This study aims to elucidate the mechanism of mechanical stimulation regulating TRPV4. Methods Immunohistochemical staining and western blotting were used to evaluate cartilage repair at the TBI after injury. The RNA expression and protein expression of mechanoreceptors and key pathway molecules regulating cartilage proliferation were analyzed. TBI samples were collected for transcriptome sequencing to detect gene expression. Calcium-ion imaging and flow cytometry were used to evaluate the function of TPRV4 and cellular communication network factor 2 (CCN2) after the administration of siRNA, recombinant adenovirus and agonists. Results We found that treadmill training improved the quality of TBI healing and enhanced fibrochondrocyte proliferation. The transcriptome sequencing results suggested that the elevated expression of the mechanistically stimulated regulator CCN2 and the exogenous administration of recombinant human CCN2 significantly promoted TRPV4 protein expression and fibrochondrocyte proliferation. In vitro, under mechanical stimulation conditions, small interfering RNA (siRNA)-CCN2 not only inhibited the proliferation of primary fibrochondrocytes but also suppressed TRPV4 protein expression and activity. Subsequently, primary fibrochondrocytes were treated with the TRPV4 agonist GSK1016790A and the recombinant adenovirus TRPV4 (Ad-TRPV4), and GSK1016790A partially reversed the inhibitory effect of siRNA-CCN2. The phosphoinositide 3-kinase/ protein kinase B (PI3K/AKT) signaling pathway participated in the above process. Conclusions Mechanical stimulation promoted fibrochondrocyte proliferation and TBI healing by activating TRPV4 channels and the PI3K/AKT signaling pathway, and CCN2 may be a key regulatory protein in maintaining TRPV4 activation.
Collapse
Affiliation(s)
- Xuting Bian
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Xiao Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Mei Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Hong Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Rui Wang
- Chongqing Institute of Bio-Intelligent Manufacturing, No. 60, Xingguang Avenue, Yubei District, Chongqing, 400000, China
| | - Lin Ma
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Gang He
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Shibo Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Yunjiao Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Jindong Tan
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Kanglai Tang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| | - Lin Guo
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Orthopedics/Sports Medicine Center, First Affiliated Hospital of Army Medical University, No. 30 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
| |
Collapse
|
36
|
Zimmermann J, Farooqi AR, van Rienen U. Electrical stimulation for cartilage tissue engineering - A critical review from an engineer's perspective. Heliyon 2024; 10:e38112. [PMID: 39416819 PMCID: PMC11481755 DOI: 10.1016/j.heliyon.2024.e38112] [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: 04/05/2024] [Revised: 08/31/2024] [Accepted: 09/18/2024] [Indexed: 10/19/2024] Open
Abstract
Cartilage has a limited intrinsic healing capacity. Hence, cartilage degradation and lesions pose a huge clinical challenge, particularly in an ageing society. Osteoarthritis impacts a significant number of the population and requires the development of repair and tissue engineering methods for hyaline articular cartilage. In this context, electrical stimulation has been investigated for more than 50 years already. Yet, no well-established clinical therapy to treat osteoarthritis by means of electrical stimulation exists. We argue that one reason is the lack of replicability of electrical stimulation devices from a technical perspective together with lacking hypotheses of the biophysical mechanism. Hence, first, the electrical stimulation studies reported in the context of cartilage tissue engineering with a special focus on technical details are summarized. Then, an experimental and numerical approach is discussed to make the electrical stimulation experiments replicable. Finally, biophysical hypotheses have been reviewed on the interaction of electric fields and cells that are relevant for cartilage tissue engineering. With that, the aim is to inspire future research to enable clinical electrical stimulation therapies to fight osteoarthritis.
Collapse
Affiliation(s)
- Julius Zimmermann
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
| | - Abdul Razzaq Farooqi
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Electronic Engineering, Faculty of Engineering, The Islamia University of Bahawalpur, 63100 Bahawalpur, Pakistan
| | - Ursula van Rienen
- Institute of General Electrical Engineering, University of Rostock, 18051 Rostock, Germany
- Department of Ageing of Individuals and Society, Interdisciplinary Faculty, University of Rostock, 18051 Rostock, Germany
- Department Life, Light & Matter, University of Rostock, 18051 Rostock, Germany
| |
Collapse
|
37
|
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
|
38
|
Hernández-Vega AM, Llorente I, Sánchez-Hernández R, Segura Y, Tusié-Luna T, Morales-Buenrostro LE, García-Villegas R, Islas LD, Rosenbaum T. Identification and Properties of TRPV4 Mutant Channels Present in Polycystic Kidney Disease Patients. FUNCTION 2024; 5:zqae031. [PMID: 38984987 PMCID: PMC11384909 DOI: 10.1093/function/zqae031] [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: 04/21/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Polycystic kidney disease (PKD), a disease characterized by the enlargement of the kidney through cystic growth is the fourth leading cause of end-stage kidney disease world-wide. Transient receptor potential Vanilloid 4 (TRPV4), a calcium-permeable TRP, channel participates in kidney cell physiology and since TRPV4 forms complexes with another channel whose malfunction is associated to PKD, TRPP2 (or PKD2), we sought to determine whether patients with PKD, exhibit previously unknown mutations in TRPV4. Here, we report the presence of mutations in the TRPV4 gene in patients diagnosed with PKD and determine that they produce gain-of-function (GOF). Mutations in the sequence of the TRPV4 gene have been associated to a broad spectrum of neuropathies and skeletal dysplasias but not PKD, and their biophysical effects on channel function have not been elucidated. We identified and examined the functional behavior of a novel E6K mutant and of the previously known S94L and A217S mutant TRVP4 channels. The A217S mutation has been associated to mixed neuropathy and/or skeletal dysplasia phenotypes, however, the PKD carriers of these variants had not been diagnosed with these reported clinical manifestations. The presence of certain mutations in TRPV4 may influence the progression and severity of PKD through GOF mechanisms. PKD patients carrying TRVP4 mutations are putatively more likely to require dialysis or renal transplant as compared to those without these mutations.
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
| | - Itzel Llorente
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Raúl Sánchez-Hernández
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Yayoi Segura
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México14080, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - Luis E Morales-Buenrostro
- Departmento de Nefrología y Metabolismo Mineral, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México 14080, México
| | - 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, Mexico
| | - León D Islas
- Departamento de Fisiología, Facultad de Medicina. Universidad Nacional Autónoma de México,Ciudad de México 04510, Mexico
| | - Tamara Rosenbaum
- Departamento de Neurociencia Cognitiva, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| |
Collapse
|
39
|
Jurynec MJ, Nosyreva E, Thompson D, Munoz C, Novak KA, Matheson DJ, Kazmers NH, Syeda R. PIEZO1 variants that reduce open channel probability are associated with familial osteoarthritis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.03.24312969. [PMID: 39281748 PMCID: PMC11398433 DOI: 10.1101/2024.09.03.24312969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The synovial joints senses and responds to a multitude of physical forces to maintain joint homeostasis. Disruption of joint homeostasis results in development of osteoarthritis (OA), a disease characterized by loss of joint space, degeneration of articular cartilage, remodeling of bone and other joint tissues, low-grade inflammation, and pain. How changes in mechanosensing in the joint contribute to OA susceptibility remains elusive. PIEZO1 is a major mechanosensitive cation channel in the joint directly regulated by mechanical stimulus. To test whether altered PIEZO1 channel activity causes increased OA susceptibility, we determined whether variants affecting PIEZO1 are associated with dominant inheritance of age-associated familial OA. We identified four rare coding variants affecting PIEZO1 that are associated with familial hand OA. Single channel analyses demonstrated that all four PIEZO1 mutant channels act in a dominant-negative manner to reduce the open probability of the channel in response to pressure. Furthermore, we show that a GWAS mutation in PIEZO1 associated with reduced joint replacement results in increased channel activity when compared with WT and the mutants. Our data support the hypothesis that reduced PIEZO1 activity confers susceptibility to age-associated OA whereas increased PIEZO1 activity may be associated with reduced OA susceptibility.
Collapse
Affiliation(s)
- Michael J Jurynec
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
- Department of Human Genetics, University of Utah, Salt Lake City, UT, 84112
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112
| | - Elena Nosyreva
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - David Thompson
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - Crystal Munoz
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| | - Kendra A Novak
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Derek J Matheson
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Nikolas H Kazmers
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, 84108
| | - Ruhma Syeda
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, 75390
| |
Collapse
|
40
|
Lan M, Liu Y, Liu J, Zhang J, Haider MA, Zhang Y, Zhang Q. Matrix Viscoelasticity Tunes the Mechanobiological Behavior of Chondrocytes. Cell Biochem Funct 2024; 42:e4126. [PMID: 39324844 DOI: 10.1002/cbf.4126] [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: 04/18/2024] [Revised: 08/25/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024]
Abstract
In articular cartilage, the pericellular matrix acting as a specialized mechanical microenvironment modulates environmental signals to chondrocytes through mechanotransduction. Matrix viscoelastic alterations during cartilage development and osteoarthritis (OA) degeneration play an important role in regulating chondrocyte fate and cartilage matrix homeostasis. In recent years, scientists are gradually realizing the importance of matrix viscoelasticity in regulating chondrocyte function and phenotype. Notably, this is an emerging field, and this review summarizes the existing literatures to the best of our knowledge. This review provides an overview of the viscoelastic properties of hydrogels and the role of matrix viscoelasticity in directing chondrocyte behavior. In this review, we elaborated the mechanotransuction mechanisms by which cells sense and respond to the viscoelastic environment and also discussed the underlying signaling pathways. Moreover, emerging insights into the role of matrix viscoelasticity in regulating chondrocyte function and cartilage formation shed light into designing cell-instructive biomaterial. We also describe the potential use of viscoelastic biomaterials in cartilage tissue engineering and regenerative medicine. Future perspectives on mechanobiological comprehension of the viscoelastic behaviors involved in tissue homeostasis, cellular responses, and biomaterial design are highlighted. Finally, this review also highlights recent strategies utilizing viscoelastic hydrogels for designing cartilage-on-a-chip.
Collapse
Affiliation(s)
- Minhua Lan
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanli Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Junjiang Liu
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Jing Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Muhammad Adnan Haider
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
| | - Yanjun Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| | - Quanyou Zhang
- College of Artificial Intelligence, Taiyuan University of Technology, Taiyuan, China
- Shanxi Key Laboratory of Bone and Soft Tissue Injury Repair, Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
41
|
Kondiboyina V, Boyer TL, Mooney N, Bajpayee AG, Shefelbine SJ. Effect of dynamic loading on calcium signaling in In-Situ chondrocytes. J Biomech 2024; 174:112265. [PMID: 39137485 DOI: 10.1016/j.jbiomech.2024.112265] [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: 03/06/2024] [Revised: 07/12/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024]
Abstract
Chondrocytes respond to mechanical stimuli by increasing their intracellular calcium concentration. The response depends on the cellular environment. Previous studies have investigated chondrocytes under slow strain rates or cells embedded in hydrogels, but the response of chondrocytes in their native environment under physiologically relevant cyclic loads and dynamic hydrostatic pressure has not been studied. This study investigated the calcium signaling response of in-situ chondrocytes under physiological cyclic compressive loads and hydrostatic pressure with varying frequency and load rates. Bovine cartilage explants were stained with a fluorescent calcium indicator dye and subjected to physiologically relevant cyclic loads using a custom-built loading device secured on a confocal/multiphoton microscope. Calcium fluorescence intensities of the cells were tracked and analyzed. Loading groups were compared using one-way ANOVA followed by a post-hoc test with Tukey correction (α = 0.05). The percentage of cells signaling increased in all compressive loading conditions compared to the no-load baseline. The percentage of cells responding under 1 Hz load was significantly greater than the slow ramp and 0.1 Hz group (p < 0.05). The number of compression cycles had no effect on the calcium signaling response (p > 0.05). The width and time between consecutive peaks were not different between different loading conditions (p > 0.05). Calcium signaling of in-situ chondrocytes did not increase under dynamic hydrostatic pressure of magnitudes up to 0.2 MPa at frequencies of 0.5 Hz and 0.05 Hz (p > 0.05). In conclusion, in-situ chondrocytes respond to physiological compressive loads in a strain rate-dependent manner with an increased number of responsive cells and unaltered temporal characteristics.
Collapse
Affiliation(s)
| | - Timothy L Boyer
- Dept. of Bioengineering, Northeastern University, Boston, MA, USA.
| | - Noah Mooney
- Dept. of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA.
| | | | - Sandra J Shefelbine
- Dept. of Bioengineering, Northeastern University, Boston, MA, USA; Dept. of Mechanical and Industrial Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
42
|
Zhou R, Fu W, Vasylyev D, Waxman SG, Liu CJ. Ion channels in osteoarthritis: emerging roles and potential targets. Nat Rev Rheumatol 2024; 20:545-564. [PMID: 39122910 DOI: 10.1038/s41584-024-01146-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
Osteoarthritis (OA) is a highly prevalent joint disease that causes substantial disability, yet effective approaches to disease prevention or to the delay of OA progression are lacking. Emerging evidence has pinpointed ion channels as pivotal mediators in OA pathogenesis and as promising targets for disease-modifying treatments. Preclinical studies have assessed the potential of a variety of ion channel modulators to modify disease pathways involved in cartilage degeneration, synovial inflammation, bone hyperplasia and pain, and to provide symptomatic relief in models of OA. Some of these modulators are currently being evaluated in clinical trials. This review explores the structures and functions of ion channels, including transient receptor potential channels, Piezo channels, voltage-gated sodium channels, voltage-dependent calcium channels, potassium channels, acid-sensing ion channels, chloride channels and the ATP-dependent P2XR channels in the osteoarthritic joint. The discussion spans channel-targeting drug discovery and potential clinical applications, emphasizing opportunities for further research, and underscoring the growing clinical impact of ion channel biology in OA.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Wenyu Fu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA
| | - Dmytro Vasylyev
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephen G Waxman
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| | - Chuan-Ju Liu
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
43
|
Jing X, Kotelsky A, Zhang Y, Dirksen R, Mannava S, Buckley M, Lee W. Mechano-adaptation: Exercise-driven Piezo1 & Piezo2 augmentation and chondroprotection in articular cartilage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.02.606183. [PMID: 39211060 PMCID: PMC11361185 DOI: 10.1101/2024.08.02.606183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Chondrocytes in adult joints are mechanosensitive post-mitotic quiescent cells with robustly expressed both Piezo1 and Piezo2 ion channels. Here, we examined the mechano-adaptation and Piezo modulations in articular chondrocytes using a mouse exercise model. We first found differential expression patterns of PIEZO1 and PIEZO2 in articular chondrocytes of healthy knee joints; chondrocytes in tibial cartilage (T) exhibit significantly higher PIEZO1 and PIEZO2 than femoral chondrocytes (F). Interestingly, a few weeks of exercise caused both PIEZO1 and PIEZO2 augmentation in F and T compared to the sedentary control group. Despite the increased expression levels of these mechanosensors, chondrocytes in exercised cartilage exhibit significantly reduced mechanical susceptibility against 1mJ impact. PIEZO1 modulation was relatively more rapid than PIEZO2 channels post-exercise. We tested the exercise-induced effect using Piezo1-conditional knockout (Pz1-cKO; Agc1 CreERT2 ;Piezo1 fl/fl ). Pz1-cKO mice exhibit diminished exercise-driven chondroprotection against 1mJ impact, suggesting essential roles of Piezo1-mediated mechanotransduction for physiologic-induced cartilage matrix homeostasis. In addition, using a mouse OA model, we further found the modulated PIEZO1 in chondrocytes, consistent with reports in Ren et al., but without PIEZO2 modulations over OA progression. In summary, our data reveal the distinctly tuned Piezo1 and Piezo2 channels in chondrocytes post-exercise and post-injury, in turn modulating the mechanical susceptibility of chondrocytes. We postulate that Piezo1 is a tightly-regulated biphasic biomarker ; Piezo1 antagonism may increase cellular survival post-injury and Piezo1 (with Piezo2) agonism to promote cartilage ECM restoration.
Collapse
|
44
|
Guo Y, Stampoultzis T, Karami P, Nasrollahzadeh N, Rana VK, Pioletti DP. HSP70-A key regulator in chondrocyte homeostasis under naturally coupled hydrostatic pressure-thermal stimuli. Osteoarthritis Cartilage 2024; 32:896-908. [PMID: 38679285 DOI: 10.1016/j.joca.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/01/2024]
Abstract
OBJECTIVE During physical activities, chondrocytes experience coupled stimulation of hydrostatic pressure (HP) and a transient increase in temperature (T), with the latter varying within a physiological range from 32.5 °C to 38.7 °C. Previous short-term in vitro studies have demonstrated that the combined hydrostatic pressure-thermal (HP-T) stimuli more significantly enhance chondroinduction and chondroprotection of chondrocytes than isolated applications. Interestingly, this combined benefit is associated with a corresponding increase in HSP70 levels when HP and T are combined. The current study therefore explored the indispensable role of HSP70 in mediating the combined effects of HP-T stimuli on chondrocytes. DESIGN In this mid-long-term study of in vitro engineered cartilage constructs, we assessed chondrocyte responses to HP-T stimuli using customized bioreactor in standard and HSP70-inhibited cultures. RESULTS Surprisingly, under HSP70-inhibited conditions, the usually beneficial HP-T stimuli, especially its thermal component, exerted detrimental effects on chondrocyte homeostasis, showing a distinct and unfavorable shift in gene and protein expression patterns compared to non-HSP70-inhibited settings. Such effects were corroborated through mechanical testing and confirmed using a secondary cell source. A proteomic-based mechanistic analysis revealed a disruption in the balance between biosynthesis and fundamental cellular structural components in HSP70-inhibited conditions under HP-T stimuli. CONCLUSIONS Our results highlight the critical role of sufficient HSP70 induction in mediating the beneficial effects of coupled HP-T stimulation on chondrocytes. These findings help pave the way for new therapeutic approaches to enhance physiotherapy outcomes and potentially shed light on the elusive mechanisms underlying the onset of cartilage degeneration, a long-standing enigma in orthopedics.
Collapse
Affiliation(s)
- Yanheng Guo
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | | | - Peyman Karami
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Naser Nasrollahzadeh
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Vijay K Rana
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland
| | - Dominique P Pioletti
- Laboratory of Biomechanical Orthopedics, Institute of Bioengineering, EPFL, Switzerland.
| |
Collapse
|
45
|
Nims R, Palmer DR, Kassab J, Zhang B, Guilak F. The chondrocyte "mechanome": Activation of the mechanosensitive ion channels TRPV4 and PIEZO1 drives unique transcriptional signatures. FASEB J 2024; 38:e23778. [PMID: 38959010 PMCID: PMC11327906 DOI: 10.1096/fj.202400883r] [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: 04/18/2024] [Revised: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
The mechanosensitive ion channels Transient Receptor Potential Vanilloid 4 (TRPV4) and PIEZO1 transduce physiologic and supraphysiologic magnitudes of mechanical signals in the chondrocyte, respectively. TRPV4 activation promotes chondrogenesis, while PIEZO1 activation by supraphysiologic deformations drives cell death. The mechanisms by which activation of these channels discretely drives changes in gene expression to alter cell behavior remain to be determined. To date, no studies have contrasted the transcriptomic response to activation of these channels nor has any published data attempted to correlate these transcriptomes to alterations in cellular function. This study used RNA sequencing to comprehensively investigate the transcriptomes associated with activation of TRPV4 or PIEZO1, revealing that TRPV4 and PIEZO drive distinct transcriptomes and also exhibit unique co-regulated clusters of genes. Notably, activation of PIEZO1 through supraphysiologic deformation induced a transient inflammatory profile that overlapped with the interleukin (IL)-1-responsive transcriptome and contained genes associated with cartilage degradation and osteoarthritis progression. However, both TRPV4 and PIEZO1 were also shown to elicit anabolic effects. PIEZO1 expression promoted a pro-chondrogenic transcriptome under unloaded conditions, and daily treatment with PIEZO1 agonist Yoda1 significantly increased sulfated glycosaminoglycan deposition in vitro. These findings emphasize the presence of a broad "mechanome" with distinct effects of TRPV4 and PIEZO1 activation in chondrocytes, suggesting complex roles for PIEZO1 in both the physiologic and pathologic responses of chondrocytes. The identification of transcriptomic profiles unique to or shared by PIEZO1 and TRPV4 (distinct from IL-1-induced inflammation) could inform future therapeutic designs targeting these channels for the management and treatment of osteoarthritis.
Collapse
Affiliation(s)
- Robert Nims
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Daniel R Palmer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Jordan Kassab
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Bo Zhang
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children-Saint Louis, St. Louis, Missouri, USA
- Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
46
|
Liu Y, Jia F, Li K, Liang C, Lin X, Geng W, Li Y. Critical signaling molecules in the temporomandibular joint osteoarthritis under different magnitudes of mechanical stimulation. Front Pharmacol 2024; 15:1419494. [PMID: 39055494 PMCID: PMC11269110 DOI: 10.3389/fphar.2024.1419494] [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: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
The mechanical stress environment in the temporomandibular joint (TMJ) is constantly changing due to daily mandibular movements. Therefore, TMJ tissues, such as condylar cartilage, the synovial membrane and discs, are influenced by different magnitudes of mechanical stimulation. Moderate mechanical stimulation is beneficial for maintaining homeostasis, whereas abnormal mechanical stimulation leads to degeneration and ultimately contributes to the development of temporomandibular joint osteoarthritis (TMJOA), which involves changes in critical signaling molecules. Under abnormal mechanical stimulation, compensatory molecules may prevent degenerative changes while decompensatory molecules aggravate. In this review, we summarize the critical signaling molecules that are stimulated by moderate or abnormal mechanical loading in TMJ tissues, mainly in condylar cartilage. Furthermore, we classify abnormal mechanical stimulation-induced molecules into compensatory or decompensatory molecules. Our aim is to understand the pathophysiological mechanism of TMJ dysfunction more deeply in the ever-changing mechanical environment, and then provide new ideas for discovering effective diagnostic and therapeutic targets in TMJOA.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yanxi Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Mobasheri A, Matta C, Giles W, Choi H, Ivanavicius S. The interplay between inflammatory mediators and mechanotransduction is mediated by ion channels in articular chondrocytes: Functional consequences in osteoarthritis. Phys Life Rev 2024; 49:123-126. [PMID: 38692123 DOI: 10.1016/j.plrev.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 04/21/2024] [Indexed: 05/03/2024]
Affiliation(s)
- Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Department of Joint Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, PR China; World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium.
| | - Csaba Matta
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Wayne Giles
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Heonsik Choi
- Healthcare Research Institute, Kolon Advanced Research Cluster, Magok-dong, Gangseo-gu, Seoul, South Korea
| | | |
Collapse
|
48
|
Mark Kim MK, Lawrence M, Quinonez D, Brooks C, Ramachandran R, Séguin CA. Transient receptor potential vanilloid 4 regulates extracellular matrix composition and mediates load-induced intervertebral disc degeneration in a mouse model. Osteoarthritis Cartilage 2024; 32:881-894. [PMID: 38604493 DOI: 10.1016/j.joca.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE Transient receptor potential vanilloid 4 (TRPV4) is a multi-modally activated cation channel that mediates mechanotransduction pathways by which musculoskeletal tissues respond to mechanical load and regulate tissue health. Using conditional Trpv4 knockout mice, we investigated the role of Trpv4 in regulating intervertebral disc (IVD) health and injury-induced IVD degeneration. METHODS Col2-Cre;Trpv4fl/f (Trpv4 KO) mice were used to knockout Trpv4 in all type 2 collagen-expressing cells. Effects of gene targeting alone was assessed in lumbar spines, using vertebral bone length measurement, histological, immunohistochemistry and gene expression analyses, and mechanical testing. Disc puncture was performed on caudal IVDs of wild-type (WT) and Trpv4 KO mice at 2.5- and 6.5-months-of-age. Six weeks after puncture (4- and 8-months-of-age at sacrifice), caudal spines were assessed using histological analyses. RESULTS While loss of Trpv4 did not significantly alter vertebral bone length and tissue histomorphology compared to age-matched WT mice, Trpv4 KO mice showed decreased proteoglycan and PRG4 staining in the annulus fibrosus compared to WT. At the gene level, Trpv4 KO mice showed significantly increased expression of Acan, Bgn, and Prg4 compared to WT. Functionally, loss of Trpv4 was associated with significantly increased neutral zone length in lumbar IVDs. Following puncture, both Trpv4 KO and WT mice showed similar signs of degeneration at the site of injury. Interestingly, loss of Trpv4 prevented mechanically-induced degeneration in IVDs adjacent to sites of injury. CONCLUSION These studies suggest a role for Trpv4 in regulating extracellular matrix synthesis and mediating the response of IVD tissues to mechanical stress.
Collapse
Affiliation(s)
- Min Kyu Mark Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Matthew Lawrence
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Diana Quinonez
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
49
|
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
|
50
|
Pratt SJP, Plunkett CM, Kuzu G, Trinh T, Barbara J, Choconta P, Quackenbush D, Huynh T, Smith A, Barnes SW, New J, Pierce J, Walker JR, Mainquist J, King FJ, Elliott J, Hammack S, Decker RS. A high throughput cell stretch device for investigating mechanobiology in vitro. APL Bioeng 2024; 8:026129. [PMID: 38938688 PMCID: PMC11210978 DOI: 10.1063/5.0206852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024] Open
Abstract
Mechanobiology is a rapidly advancing field, with growing evidence that mechanical signaling plays key roles in health and disease. To accelerate mechanobiology-based drug discovery, novel in vitro systems are needed that enable mechanical perturbation of cells in a format amenable to high throughput screening. Here, both a mechanical stretch device and 192-well silicone flexible linear stretch plate were designed and fabricated to meet high throughput technology needs for cell stretch-based applications. To demonstrate the utility of the stretch plate in automation and screening, cell dispensing, liquid handling, high content imaging, and high throughput sequencing platforms were employed. Using this system, an assay was developed as a biological validation and proof-of-concept readout for screening. A mechano-transcriptional stretch response was characterized using focused gene expression profiling measured by RNA-mediated oligonucleotide Annealing, Selection, and Ligation with Next-Gen sequencing. Using articular chondrocytes, a gene expression signature containing stretch responsive genes relevant to cartilage homeostasis and disease was identified. The possibility for integration of other stretch sensitive cell types (e.g., cardiovascular, airway, bladder, gut, and musculoskeletal), in combination with alternative phenotypic readouts (e.g., protein expression, proliferation, or spatial alignment), broadens the scope of high throughput stretch and allows for wider adoption by the research community. This high throughput mechanical stress device fills an unmet need in phenotypic screening technology to support drug discovery in mechanobiology-based disease areas.
Collapse
Affiliation(s)
- Stephen J. P. Pratt
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | | | - Guray Kuzu
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Ton Trinh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joshua Barbara
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Paula Choconta
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Doug Quackenbush
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Truc Huynh
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Anders Smith
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - S. Whitney Barnes
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Joel New
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Pierce
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - John R. Walker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - James Mainquist
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Frederick J. King
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Jimmy Elliott
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Scott Hammack
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| | - Rebekah S. Decker
- Novartis, Biomedical Research 10675 John Jay Hopkins Dr, San Diego, California 92121, USA
| |
Collapse
|