Benita Mercy Karunya B, Mary Purna Chacko, Ganesh Parasuraman, Abel Livingston, Boopalan Ramasamy, Solomon Sathishkumar, Elizabeth Vinod
{"title":"Cytogenetic profiling of human articular cartilage chondrocytes, fibronectin adhesion assay derived chondroprogenitors and migratory chondroprogenitors.","authors":"Benita Mercy Karunya B, Mary Purna Chacko, Ganesh Parasuraman, Abel Livingston, Boopalan Ramasamy, Solomon Sathishkumar, Elizabeth Vinod","doi":"10.1080/03008207.2025.2548271","DOIUrl":"https://doi.org/10.1080/03008207.2025.2548271","url":null,"abstract":"<p><strong>Purpose: </strong>Cartilage-derived chondroprogenitors, with inherent chondrogenic capacity and low hypertrophic potential, represent a promising avenue for cartilage regeneration. For clinical translation, assessment of cellular genomic stability is a quality control mandate. Since culturing cells to higher passage numbers for achieving the cell requirement is indispensable, it is necessary to evaluate the possibility of culture-driven mutations before transplantation. Being a relatively newly discovered cell subset, the information on the genetic profile of these cartilage-resident cells is notably limited.</p><p><strong>Methods: </strong>The study investigated the genomic stability of fibronectin adhesion assay-derived chondroprogenitors(FAA-CP), migratory chondroprogenitors(MCP) and chondrocytes (<i>n</i> = 3). Conventional karyotyping and microarray analysis were performed on early and late passage cells to assess their genomic integrity under standard culture conditions and any groups that showed variations were further evaluated for their tumorigenic potential using the soft-agar assay.</p><p><strong>Results: </strong>Chondrocytes exhibited a higher propensity for culture-induced genetic aberrations, including chromosomal losses, gains, inversions, and translocations. In contrast, both the chondroprogenitor groups demonstrated greater genomic stability throughout culture, with an instance of Trisomy-7 observed in early passage and a loss of gonosome in the later passage MCP group. Microarray analysis of chondroprogenitors showed a normal genomic profile, and soft agar assays indicated a non-tumorigenic profile for all cell groups that showed abnormal cytogenetic profiles.</p><p><strong>Conclusions: </strong>The study highlights the importance of distinguishing between inherent genetic abnormalities and those acquired during culture, particularly when considering cells for therapeutic applications. While the observed genetic variations did not confer tumorigenic potential, careful consideration is essential prior to therapy.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-14"},"PeriodicalIF":2.1,"publicationDate":"2025-08-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144945696","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aditya D Deshpande, Likhitha B N, Smriti Shukla, Rony S Emmanuel, Pranay K Konda, Khan Sharun, Rohit Kumar, Asok Kumar, Amarpal, G Saikumar, Vikash Chandra, G Taru Sharma
{"title":"Comparative evaluation and optimization of amniotic membrane derived bioscaffold for bone regeneration in critical sized bone defect in rabbit radius model.","authors":"Aditya D Deshpande, Likhitha B N, Smriti Shukla, Rony S Emmanuel, Pranay K Konda, Khan Sharun, Rohit Kumar, Asok Kumar, Amarpal, G Saikumar, Vikash Chandra, G Taru Sharma","doi":"10.1080/03008207.2025.2540951","DOIUrl":"10.1080/03008207.2025.2540951","url":null,"abstract":"<p><strong>Background: </strong>Bone regeneration and repair are critical research areas within regenerative medicine, aiming to address the challenges posed by critical-sized bone defects. Bioscaffolds and cell-based therapies have been explored to enhance osteogenesis and promote effective bone regeneration. This study aimed to assess the regenerative potential of rabbit amniotic membrane (rAM) derived bioscaffold and its comparative evaluation with another bioscaffold, the decellularized periosteum (DP) of the buffalo rib, which was recellularized with rAM derived mesenchymal stem cells (MSCs).</p><p><strong>Methods: </strong>Passage 3 (P3) rAM-MSCs were validated for positive and negative stemness marker expression by PCR, immunolocalization and trilineage differentiation. Fresh rAM was cryopreserved for three months. An autologous rabbit model was used to assess the osteogenic capacity of different bioscaffolds: fresh rAM (Group II), frozen-thawed rAM (Group III), DP (Group IV), and DP enriched with rAM-MSCs (Group V) and control (Group I) in critical-size defect of 10 mm in radius bone. Radiographic evaluation was performed on the 1<sup>st</sup>, 60<sup>th</sup> and 90<sup>th</sup> days, and ultramicroscopic and histomorphological evaluations were performed.</p><p><strong>Results: </strong>Groups II and V presented the highest levels of remodeling and osteogenesis, a reduction in the defect size and total radiological scores. Groups II and V had the highest levels of osteogenesis, bone marrow development, cortical bone production, and medullary bone formation and the highest total histology score.</p><p><strong>Conclusion: </strong>These findings revealed that fresh rAM, a rich source of MSCs, and DP enhanced with rAM-MSCs are the preferred bioscaffolds for critical-sized bone defect repair.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-14"},"PeriodicalIF":2.1,"publicationDate":"2025-08-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144882364","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sedat Dogru, Nicholas Simone, Halide Z Haciguzeller, Gabriela M Alba, Matthew D Layne, Michael B Albro
{"title":"Latent TGF-β content of articular cartilage with development and age.","authors":"Sedat Dogru, Nicholas Simone, Halide Z Haciguzeller, Gabriela M Alba, Matthew D Layne, Michael B Albro","doi":"10.1080/03008207.2025.2543383","DOIUrl":"https://doi.org/10.1080/03008207.2025.2543383","url":null,"abstract":"<p><strong>Background: </strong>An important regulatory feature of transforming growth factor-β (TGF-β) in cartilage stems from its extracellular matrix (ECM) sequestration as an inactive latent complex (LTGF-β), a configuration that enables need-based activation in response to physiologic stimuli. Recent work has elucidated the dysregulation of TGF-β signaling with development, age, and pathology. However, characterizations of LTGF-β content levels in cartilage are limited. Here, we characterize the variation of LTGF-β in articular cartilage with tissue development and age.</p><p><strong>Methods: </strong>LTGF-β evolution with development is characterized via measures of bovine cartilage explants from fetal, skeletally immature (2-4 months), and skeletally mature (60 months) animals. LTGF-β evolution with cartilage aging is characterized via measures of human cartilage explants procured via cadavers (n=19 donors) ranging in age from 13 to 80 years.</p><p><strong>Results: </strong>For bovine cartilage, while LTGF-β per unit tissue volume decreases with development (50.5 ± 22.0 to 16.4 ± 7.4 ng/mL for LTGF-β1; p<0.01; 29.8 ± 10.7 to 12.6 ± 4.1 ng/mL for LTGF- β2; p<0.01), LTGF-β1 and LTGF-β2 contents per cell number in the tissue does not change with development (p>0.41), indicating that the amount of LTGF-β available for each chondrocyte remains conserved. LTGF-β1 content in human tissues exhibits a dynamic range of an order of magnitude (6.4 ± 3.6 to 70.6 ± 21.8 ng/mL), but donor age is not predictive of LTGF-β1 content in cartilage (p=0.87).</p><p><strong>Conclusions: </strong>These results suggest that development and aging are not limiting factors for LTGF-β availability in cartilage.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-11"},"PeriodicalIF":2.1,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144816024","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yan Liu, Xuan Wang, Jianjun Yuan, Quan Zhang, Guang Li, Rong Tian
{"title":"METTL3-mediated m6A modification of ZIC2 promotes osteosarcoma development.","authors":"Yan Liu, Xuan Wang, Jianjun Yuan, Quan Zhang, Guang Li, Rong Tian","doi":"10.1080/03008207.2025.2540041","DOIUrl":"https://doi.org/10.1080/03008207.2025.2540041","url":null,"abstract":"<p><strong>Background: </strong>Zic family member 2 (ZIC2) is closely associated with cancer development, however, its role in the progression of osteosarcoma (OS) remains unknown. This study aims to reveal the function of ZIC2 in OS cell tumor progression and the underlying mechanism.</p><p><strong>Methods: </strong>This work performed quantitative real-time polymerase chain reaction to detect mRNAlevels of ZIC2, retinoic acid receptor alpha (RARA) and methyltransferase 3,N6-adenosine-methyltransferase complex catalytic subunit (METTL3),whereas protein level was detected by western blotting assay and immunohistochemistry assay. CCK-8 together with 5-Ethynyl-2'-deoxyuridine (EdU) assay were performed to analyze cell growth. Cell apoptosis was assessed by flow cytometry. Transwell assay and wound-healing assay were used for measuring cell invasion and migration. A xenograft mouse model assay was conducted to reveal the effect of ZIC2 on tumor formation <i>invivo</i>. The association of ZIC2 and METTL3 was identified by m6A RNA immunoprecipitation assay, dual-luciferase reporter gene assay, Actinomycin D assay and co-immunoprecipitation assay.</p><p><strong>Results: </strong>ZIC2and METTL3 mRNA expression were upregulated in OS tissues relative to paracancerous normal tissues. ZIC2 was a prognostic biomarker for OS,and its expression was significantly associated with TNM stage, lymph node metastasis, and tumor size of OS patients. Additionally, ZIC2depletion inhibited proliferation, invasion and migration and induced apoptosis of OS cells, but ZIC2 overexpression had the opposite effects. Moreover, ZIC2 knockdown delayed tumor formation <i>invivo</i>. METTL3stabilized ZIC2 mRNA through m6A methylation modification. Further,METTL3 deficiency repressed OS cell malignancy by reducing ZIC2expression.</p><p><strong>Conclusion: </strong>METTL3-mediated m6Amodification of ZIC2 contributed to OS development, providing therapeutic targets for OS.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-12"},"PeriodicalIF":2.1,"publicationDate":"2025-08-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144793679","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Impact of VX-765 and VX-740 on chondrogenesis and inflammatory cytokine release in murine micromass cultures.","authors":"Barbora Vesela, Katerina Dadakova, Katerina Holomkova, Corina Blecha, Natalia Obratilova, Susanne Grässel, Eva Matalova","doi":"10.1080/03008207.2025.2539414","DOIUrl":"https://doi.org/10.1080/03008207.2025.2539414","url":null,"abstract":"<p><strong>Aim: </strong>Caspase-1 inhibition is a promising option for degenerative joint diseases such as osteoarthritis; however, there is still a long way to go toward clinical use. One of the open challenges is associated with the non-inflammatory role of this caspase in the inflammatory environment as well as under physiological conditions. This study therefore focuses on two already pre-clinically tested caspase-1 inhibitors, VX-765 and VX-740, to specify their effects on chondrogenic cells.</p><p><strong>Material and methods: </strong>The analysis was performed on mouse micromass cultures where chondrocyte differentiation, inflammatory cytokine release, and gene expression were examined.</p><p><strong>Results: </strong>Our data indicate that the inhibitor VX-740 increases chondrogenesis, suggesting osteocalcin as a target molecule. In the inflammatory environment induced by IL-1β, there was an increase in chondrogenic nodules and partial compensation of differentiation for both investigated inhibitors. Morphological changes were not primarily due to changes in chondrogenic/osteogenic gene expression, but different levels of inflammatory molecules were found in the culture supernatant. While an increase in anti-inflammatory cytokine levels was observed with VX-765, a decrease in pro-inflammatory cytokines was recorded in the case of VX-740 treatment.</p><p><strong>Conclusion: </strong>The results demonstrate the differential effects of the caspase-1 inhibitors VX-765 and VX-740 on chondrogenic cell cultures and point to molecules that may be potential targets for use in the local treatment of osteoarthritis.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-11"},"PeriodicalIF":2.1,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Microengineering the synovial membrane microenvironment for osteoarthritis research.","authors":"Hyon-U Pak, Daqing Wang, Jianhua Qin, Hongjing Li","doi":"10.1080/03008207.2025.2534723","DOIUrl":"https://doi.org/10.1080/03008207.2025.2534723","url":null,"abstract":"<p><p>Osteoarthritis (OA) is a multifactorial joint disease characterized by cartilage degradation, subchondral bone remodeling, synovitis, and cartilage matrix degradation. The synovial membrane plays a pivotal role in the progression of OA through low-grade inflammation and secretion of catabolic enzymes under altered mechanical homeostasis. While widely used to study OA pathogenesis and therapies, in vitro models (e.g. 2D synoviocyte co-cultures) frequently lack critical aspects of the in vivo synovial microenvironment, such as cellular heterogeneity, physiologically relevant mechanical stress, and dynamic cell-matrix crosstalk. These shortcomings reduce their translational value. This translational gap indicates the need for advanced 3D microengineered platforms that integrate patient-specific cells, biomechanical elements, and real-time biosensing to bridge <i>in vitro</i> findings to clinical outcomes. Recent advances in microengineering offer innovative in vitro systems such as OA synovium-on-a-chip, 3D-printed constructs, and hydrogel-based organoids that recapitulate key features of the synovial microenvironment. These tools enable precise control over mechanical stimuli, matrix composition, and cell-cell signaling. This review summarizes the microenvironment of the OA synovium, critiques existing model systems, and highlights emerging microengineering strategies aimed at better mimicking OA pathophysiology and advancing translational research.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-9"},"PeriodicalIF":2.1,"publicationDate":"2025-07-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144741380","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yunfei Wang, Ling Wu, Sujing Zong, Pengcui Li, Lixun Chai, Li Guo, Xinping Chen, Shiping Yu, Jian Sun
{"title":"Histone deacetylase 4 in limb mesenchyme is essential for chondrocyte proliferation, growth plate maintenance and proper bone formation.","authors":"Yunfei Wang, Ling Wu, Sujing Zong, Pengcui Li, Lixun Chai, Li Guo, Xinping Chen, Shiping Yu, Jian Sun","doi":"10.1080/03008207.2025.2530045","DOIUrl":"https://doi.org/10.1080/03008207.2025.2530045","url":null,"abstract":"<p><strong>Background: </strong>Dysregulation of well-ordered chondrocyte proliferation and differentiation leads to distorted architecture of the growth plate, resulting in skeletal dysplasia with impaired longitudinal bone growth. Histone deacetylase 4 (HDAC4) is essential for chondrocyte hypertrophy and endochondral bone formation, but its role in postnatal bone development remains unexplored due to early lethality in Hdac4-ablated mice. Furthermore, a direct in vivo effect of Hdac4 on mesenchymal cell specification and bone development has not been investigated.</p><p><strong>Methods: </strong>We generated <i>Prx1-Cre;Hdac4<sup>fl/fl</sup>, Sp7-Cre;Hdac4<sup>fl/fl</sup>, Acan-CreERT2;Hdac4<sup>fl/fl</sup></i>, and <i>Hdac4<sup>fl/fl</sup></i> transgenic mice, respectively. The genotyping of transgenic mice was performed via conventional PCR. Whole-body radiographs and x-ray analyses of limbs were conducted. Trabecular and cortical bone microstructures of tibias from 21-day-old mice were evaluated using micro-computed tomography. EdU label-retention assay investigated cell proliferation, while histological analyses included H&E, TRAP, and Von Kossa staining. RT-qPCR and Immunohistochemistry to detect the pro-osteogenic function of HDAC4.</p><p><strong>Results: </strong>Hdac4 inactivation in limb mesenchyme cells resulted in limb shortening, premature growth plate closure, abnormal bone morphologies, and loss of the rounded articular surface. HDAC4 was crucial for regulating chondrocyte proliferation and secondary ossification center formation. Micro-computed tomography showed increased trabecular and cortical bone <i>Prx1-Cre;Hdac4<sup>fl/fl</sup></i> mice at 3 weeks, with altered microarchitecture. .</p><p><strong>Conclusion: </strong>Hdac4 in limb mesenchymal cells plays an indispensable role in chondrocyte proliferation, maintenance of the growth plate and formation of secondary ossification centers, its pro-osteogenic role was accomplished through premature differentiation of chondrocytes, along with accelerated cartilage-to-bone conversion.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"1-18"},"PeriodicalIF":2.8,"publicationDate":"2025-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144689132","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Sofia Pettenuzzo, Alice Berardo, Elisa Belluzzi, Assunta Pozzuoli, Pietro Ruggieri, Emanuele Luigi Carniel, Chiara Giulia Fontanella
{"title":"Mechanical insights into fat pads: a comparative study of infrapatellar and suprapatellar fat pads in osteoarthritis.","authors":"Sofia Pettenuzzo, Alice Berardo, Elisa Belluzzi, Assunta Pozzuoli, Pietro Ruggieri, Emanuele Luigi Carniel, Chiara Giulia Fontanella","doi":"10.1080/03008207.2025.2502591","DOIUrl":"10.1080/03008207.2025.2502591","url":null,"abstract":"<p><strong>Objective: </strong>Osteoarthritis (OA) is the most common musculoskeletal disorder, primarily affecting knee joints and causing pain and disability. The infrapatellar (IFP) and the suprapatellar (SFP) fat pad are knee adipose tissues that play essential mechanical roles during articular activity but are also sources of adipokines and cytokines, contributing to OA progression. For this reason, this work aims to provide new insights into IFP and SFP implications in knee OA.</p><p><strong>Materials and methods: </strong>IFP and SFP tissue mechanical properties were studied through compression, indentation and shear mechanical tests performed on samples collected from patients who underwent total knee arthroplasty surgery due to end-stage OA. The energy loss, peak stress, and initial and final elastic moduli were calculated from the unconfined compression tests. The time-dependent response, evaluated in terms of equilibrium relative stiffness, was computed from stress-relaxation loading conditions. Considering shear tests, they provided strain-energy dissipation density, peak shear stress, and the shear moduli.</p><p><strong>Results: </strong>Experimental results showed the typical adipose tissue mechanics features: non-linear stiffening with strain and time-dependent response. Experimental results showed that OA IFP is stiffer than OA SFP, indeed IFP final compression elastic modulus was greater than the SFP (84.43 kPa vs 35.54 kPa respectively) (<i>p</i> = 0.042). Regarding the viscoelastic properties they were comparable: the equilibrium relative stiffness was reported as 0.13 for IFP and 0.11 for SFP (<i>p</i> = 0.026).</p><p><strong>Conclusions: </strong>These outcomes provide new insights into the OA influence on knee mechanics and lay the basis for developing computational tools to improve knee prosthesis design.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"272-283"},"PeriodicalIF":2.1,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143957785","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Daniel R Palmer, Robert Nims, Bo Zhang, Farshid Guilak
{"title":"Activation of the mechanosensitive ion channels TRPV4 and PIEZO1 downregulates key regulatory systems in the chondrocyte mechanome.","authors":"Daniel R Palmer, Robert Nims, Bo Zhang, Farshid Guilak","doi":"10.1080/03008207.2025.2498512","DOIUrl":"10.1080/03008207.2025.2498512","url":null,"abstract":"<p><strong>Background: </strong>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.</p><p><strong>Methods: </strong>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).</p><p><strong>Results: </strong>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).</p><p><strong>Conclusion: </strong>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.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"239-262"},"PeriodicalIF":2.1,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12237534/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Response of cartilage explants to LPS cultured in normoxic and hypoxic conditions is modulated by Spirulina: implications for exercise recovery in vivo.","authors":"Nadia Golestani, Wendy Pearson","doi":"10.1080/03008207.2025.2507239","DOIUrl":"10.1080/03008207.2025.2507239","url":null,"abstract":"<p><p>Exercise-induced inflammation and free radical production are crucial for recovery, yet excess inflammation poses risks to equine athletes, leading to conditions like arthritis. Spirulina, recognized for its antioxidant and anti-inflammatory properties, could mitigate degenerative diseases without hindering post-exercise recovery. This study investigates Spirulina's direct impact on cartilage responses to LPS-induced inflammation in normoxic and hypoxic conditions, focusing on outcomes relevant to cartilage matrix turnover and exercise-induced inflammation. Spirulina underwent simulated digestion and liver metabolism, yielding a simulated biological extract (SP<sub>sim</sub>). In the normoxic experiment, porcine cartilage explants were cultured with SP<sub>sim</sub> (0, 30, or 90 μg/mL) for 72 h after 24 h in basal media, with LPS (0 or 10 μg/mL) added for the final 48 h. The hypoxic experiment mirrored this, with explants transferred to a hypoxia chamber for the final 48 h. Media samples collected at 0, 24, and 48 h were analyzed for biomarkers related to cartilage turnover (GAG), and exercise-induced inflammation (IL-6 and NO). Cell viability, assessed by live:dead staining, remained > 97% and unaffected by oxygen tension. In normoxic conditions, SP<sub>sim</sub> (30 μg/mL) significantly reduced GAG release at 48 h. Under hypoxia, SP<sub>sim</sub> (30 and 90 μg/mL) inhibited LPS-induced GAG release. SP<sub>sim</sub> (90 μg/mL) increased IL-6 and NO production in LPS-stimulated explants in normoxia, and a similar effect was observed with the lower SP<sub>sim</sub> dose (30 μg/mL) in hypoxic conditions. These results suggest that Spirulina may enhance cartilage mediators, potentially promoting healthy cartilage turnover during exercise recovery.</p>","PeriodicalId":10661,"journal":{"name":"Connective Tissue Research","volume":" ","pages":"284-297"},"PeriodicalIF":2.1,"publicationDate":"2025-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144126861","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}