Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (75)

Search Parameters:
Keywords = mechanosensitive tissue

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 5936 KB  
Article
Piezo1 and Piezo2 Ion Channels in Neuronal and Astrocytic Responses to MEA Implants in the Rat Somatosensory Cortex
by Pegah Haghighi, Thomas J. Smith, Ghazaal Tahmasebi, Sophia Vargas, Madison S. Jiang, Ajaree C. Massaquoi, Johnathan Huff, Jeffrey R. Capadona and Joseph J. Pancrazio
Int. J. Mol. Sci. 2025, 26(18), 9001; https://doi.org/10.3390/ijms26189001 - 16 Sep 2025
Viewed by 553
Abstract
Intracortical microelectrode arrays (MEAs) are tools for recording and stimulating neural activity, with potential applications in prosthetic control and treatment of neurological disorders. However, when chronically implanted, the long-term functionality of MEAs is hindered by the foreign body response (FBR), characterized by gliosis, [...] Read more.
Intracortical microelectrode arrays (MEAs) are tools for recording and stimulating neural activity, with potential applications in prosthetic control and treatment of neurological disorders. However, when chronically implanted, the long-term functionality of MEAs is hindered by the foreign body response (FBR), characterized by gliosis, neuronal loss, and the formation of a glial scar encapsulating layer. This response begins immediately after implantation and is exacerbated by factors such as brain micromotion and the mechanical mismatch between stiff electrodes and soft brain tissue, leading to signal degradation. Despite progress in mitigating these issues, the underlying mechanisms of the brain’s response to MEA implantation remain unclear, particularly regarding how cells sense and respond to the associated mechanical forces. Mechanosensitive ion channels, such as the Piezo family, are key mediators of cellular responses to mechanical stimuli. In this study, silicon-based NeuroNexus MEAs consisting of four shanks were implanted in the rat somatosensory cortex for sixteen weeks. Weekly neural recordings were conducted to assess signal quality over time, revealing a decline in active electrode yield and signal amplitude. Immunohistochemical analysis showed an increase in GFAP intensity and decreased neuronal density near the implant site. Furthermore, Piezo1—but not Piezo2—was strongly expressed in GFAP-positive astrocytes within 25 µm of the implant. Piezo2 expression appeared relatively uniform within each brain slice, both in and around the MEA implantation site across cortical layers. Our study builds on previous work by demonstrating a potential role of Piezo1 in the chronic FBR induced by MEA implantation over a 16-week period. Our findings highlight Piezo1 as the primary mechanosensitive channel driving chronic FBR, suggesting it may be a target for improving MEA design and long-term functionality. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

31 pages, 7761 KB  
Article
Proteome Differences in Smooth Muscle Cells from Diabetic and Non-Diabetic Abdominal Aortic Aneurysm Patients Reveal Metformin-Induced Mechanisms
by Tara A. R. van Merrienboer, Karlijn B. Rombouts, Albert C. W. A. van Wijk, Jaco C. Knol, Thang V. Pham, Sander R. Piersma, Connie R. Jimenez, Ron Balm, Kak K. Yeung and Vivian de Waard
Med. Sci. 2025, 13(3), 184; https://doi.org/10.3390/medsci13030184 - 10 Sep 2025
Viewed by 541
Abstract
Aims: Surgery remains the only definitive treatment option for abdominal aortic aneurysms (AAA), as no conclusive evidence supports drug effectiveness in preventing AAA growth. Although type 2 diabetes (T2D) is an important cardiovascular risk factor, patients with T2D show reduced AAA presence [...] Read more.
Aims: Surgery remains the only definitive treatment option for abdominal aortic aneurysms (AAA), as no conclusive evidence supports drug effectiveness in preventing AAA growth. Although type 2 diabetes (T2D) is an important cardiovascular risk factor, patients with T2D show reduced AAA presence and growth, associated with metformin use. We aimed to investigate the potential benefits of metformin on AAA using proteomics and in vitro experiments. Methods: Proteomics analysis using tandem mass spectrometry was performed on aortic smooth muscle cells (SMCs) from non-pathological controls (C-SMC, n = 8), non-diabetic (ND, n = 19) and diabetic (D, n = 5) AAA patients. Key findings were subsequently validated in aortic tissue using mass spectrometry-based proteomics. SMCs were cultured with/without metformin and analyzed. Results: Comparison of the proteome of SMCs from ND-AAA patients with controls revealed a reduction in proteins associated with metabolic processes and mitochondrial function. Cytoskeletal and extracellular matrix (ECM) proteins were elevated in ND-AAA-SMCs versus C-SMCs, with a similar cluster of mechanosensitive proteins being increased in ND-AAA-SMCs versus D-AAA-SMCs. D-AAA-SMCs showed an improved metabolic and antioxidant profile, enriched in pentose phosphate pathway proteins responsible for NAD(P)H generation (G6PD, PGD) and NAD(P)H-dependent antioxidants (NQO1, CBR1, AKR1C1, AKR1B1, GSTM1), all regulated by NRF2, an antioxidant transcription factor. Over half of the proteins identified in the protein–protein interaction network, constructed from proteins with higher expression in D-AAA SMCs versus ND-AAA SMCs, were verified in D-AAA aortic tissue. In vitro, metformin causes a shift from aerobic to anaerobic metabolism, increased AMPK activation and elevated mitochondrial biogenesis, indicated by increased PGC-1α expression. Metformin increased the gene expression of PGD, CBR1 and the protein expression of NQO1, with enhanced translocation of pNRF2 to the nucleus, due to reduced KEAP1 as negative regulator of NRF2. Consequently, metformin enhanced the gene expression of well-known antioxidant regulators SOD2 and CAT. Conclusions: This study identified significant differences in the proteome of SMCs derived from controls, ND-AAA and D-AAA patients. It highlights distinct pathways in relation to mechanosensing, metabolism and redox balance as therapeutic targets of metformin that may underlie its inhibition of AAA progression. Full article
Show Figures

Figure 1

27 pages, 3474 KB  
Article
A ‘Spicy’ Mechanotransduction Switch: Capsaicin-Activated TRPV1 Receptor Modulates Osteosarcoma Cell Behavior and Drug Sensitivity
by Arianna Buglione, David Becerril Rodriguez, Simone Dogali, Giulia Alloisio, Chiara Ciaccio, Marco Luce, Stefano Marini, Luisa Campagnolo, Antonio Cricenti and Magda Gioia
Int. J. Mol. Sci. 2025, 26(18), 8816; https://doi.org/10.3390/ijms26188816 - 10 Sep 2025
Viewed by 507
Abstract
Osteosarcoma (OS), the most common primary malignant bone tumor, arises in highly mechanosensitive tissue and exhibits marked heterogeneity and resistance to conventional therapies. While molecular drivers have been extensively characterized, the role of mechanical stimuli in OS progression remains underexplored. Here, we identify [...] Read more.
Osteosarcoma (OS), the most common primary malignant bone tumor, arises in highly mechanosensitive tissue and exhibits marked heterogeneity and resistance to conventional therapies. While molecular drivers have been extensively characterized, the role of mechanical stimuli in OS progression remains underexplored. Here, we identify the transient receptor potential vanilloid 1 (TRPV1) channel as a key regulator of mechanotransduction and drug responsiveness in OS cells. Using uniaxial cyclic stretch, we show that aggressive U-2 OS cells undergo TRPV1-dependent perpendicular reorientation, unlike the inert SAOS-2 cells. Confocal microscopy, immunohistochemistry, and atomic force microscopy reveal that nanomolar concentrations of capsaicin—a well-characterized TRPV1 agonist—chemically mimic this mechanical phenotype, altering metastatic traits including adhesion, edge architecture, migration, nuclear-to-cytoplasmic ratio, and sensitivity to doxorubicin and cisplatin. TRPV1 activation, whether mechanical or chemical, induces subtype-specific effects absent in healthy hFOB osteoblasts. Notably, it differentially regulates nuclear localization of the proto-oncogene Src in U-2 OS versus SAOS-2 cells. Corresponding changes in Src and acetylated histone H3 (acH3) levels support a role for TRPV1 in modulating the Src–acH3 mechanosignaling axis. These effects are tumor-specific, positioning TRPV1 as a mechanosensitive signaling hub that integrates mechanical and chemical cues to drive epigenetic remodeling and phenotypic plasticity in OS, with potential as a therapeutic target in aggressive, drug-resistant subtypes Full article
Show Figures

Figure 1

15 pages, 1898 KB  
Review
Mechano-Signal Transduction Pathways of the Diaphragmatic Muscle and Role of Cytoskeleton
by Junaith S. Mohamed, Patricia S. Pardo and Aladin M. Boriek
Genes 2025, 16(8), 968; https://doi.org/10.3390/genes16080968 - 18 Aug 2025
Viewed by 819
Abstract
Mechanotransduction, also referred to as mechano-signal transduction, is a biophysical process wherein cells perceive and respond to mechanical stimuli by converting them into biochemical signals that initiate specific cellular responses. This mechanism is fundamental to the development and growth, and proper functioning of [...] Read more.
Mechanotransduction, also referred to as mechano-signal transduction, is a biophysical process wherein cells perceive and respond to mechanical stimuli by converting them into biochemical signals that initiate specific cellular responses. This mechanism is fundamental to the development and growth, and proper functioning of mechanically active tissues, such as the diaphragm—a respiratory muscle vital for breathing in mammals. In vivo, the diaphragm is subjected to transdiaphragmatic pressure, and therefore, its muscle fibers are subjected to mechanical forces not only in the direction of the muscle fibers but also in the direction transverse to the fibers. Previous research conducted in our laboratory uncovered that stretching the diaphragm in either the longitudinal or transverse direction activates distinct mechanotransduction pathways. This indicates that signaling pathways in the diaphragm muscle are regulated in an anisotropic manner. In this review paper, we discussed the underlying mechanisms that regulate the anisotropic signaling pathways in the diaphragmatic muscle, emphasizing the mechanical role of cytoskeletal proteins in this context. Furthermore, we explored the regulatory mechanisms governing mechanosensitive gene transcription, including microRNAs (mechanomiRs), within the diaphragm muscle. Finally, we examined potential links between anisotropic signaling in the diaphragm muscle and various skeletal muscle disorders. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

13 pages, 1509 KB  
Review
PIEZO Channels in Mechano-Inflammation: Gatekeepers of Neuroimmune Crosstalk
by Carmelo Pirri
Diseases 2025, 13(8), 263; https://doi.org/10.3390/diseases13080263 - 15 Aug 2025
Cited by 1 | Viewed by 1240
Abstract
Mechanical forces shape immune responses in both health and disease. PIEZO1 and PIEZO2, two mechanosensitive ion channels, have emerged as critical transducers of these forces, influencing inflammation, pain, fibrosis, and neuroimmune regulation. This review aims to synthesize the current evidence on the role [...] Read more.
Mechanical forces shape immune responses in both health and disease. PIEZO1 and PIEZO2, two mechanosensitive ion channels, have emerged as critical transducers of these forces, influencing inflammation, pain, fibrosis, and neuroimmune regulation. This review aims to synthesize the current evidence on the role of PIEZO channels in mechano-inflammation, with a specific focus on their regulatory function in neuroimmune crosstalk. A comprehensive narrative synthesis was performed using the literature from PubMed, Scopus, and Web of Science up to June 2025. Experimental, translational, and mechanistic studies involving PIEZO channels in inflammatory, fibrotic, and neuroimmune processes were included. PIEZO1 is broadly expressed in immune cells, fibroblasts, and endothelial cells, where it regulates calcium-dependent activation of pro-inflammatory pathways, such as NF-kB and STAT1. PIEZO2, enriched in sensory neurons, contributes to mechanosensory amplification of inflammatory pain. Both channels are mechanistically involved in neuroinflammation, glial activation, blood–brain barrier dysfunction, connective tissue fibrosis, and visceral hypersensitivity. PIEZO channels act as integrators of biomechanical and immunological signaling. Their roles as context-dependent gatekeepers of neuroimmune crosstalk make them attractive targets for novel therapies. Full article
Show Figures

Figure 1

17 pages, 1664 KB  
Article
Mechanosensitivity and Adipose Thickness as Determinants of Pressure to Reach Deep Fasciae in Cervical and Thoracolumbar Regions
by Sílvia Pires, Marta Freitas, Francisco Pinho and Sofia Brandão
Sensors 2025, 25(16), 5073; https://doi.org/10.3390/s25165073 - 15 Aug 2025
Viewed by 527
Abstract
Objective: We aimed to quantify the mechanical pressure required to reach the deep cervical and thoracolumbar fasciae, to examine its association with pressure pain threshold (PPT) and adipose tissue thickness, and to determine whether PPT and adipose tissue thickness can predict the mechanical [...] Read more.
Objective: We aimed to quantify the mechanical pressure required to reach the deep cervical and thoracolumbar fasciae, to examine its association with pressure pain threshold (PPT) and adipose tissue thickness, and to determine whether PPT and adipose tissue thickness can predict the mechanical pressure needed to reach the fascia. Methods: Forty-three subjects’ PPT, mechanical pressure and skinfold in the trapezius and lumbar regions were evaluated using an algometer, an ultrasound scanner, and an adipometer. The Wilcoxon test, Student’s t-test, and Pearson and Spearman’s correlation tests were used (p < 0.05). Results: The values of mechanical pressure in the cervical and lumbar regions were 6.06 ± 0.186 N/cm2 and 5.85 ± 5.280, 7.287 N/cm2, respectively. PPT values were 18.88 ± 0.917 N/cm2, and 46.46 ± 2.408 N/cm2, respectively (p < 0.001), and the adipose tissue thickness values in the cervical and lumbar regions were 0.88 ± 0.675 cm, 1.08 and 1.48 ± 0.069 cm (p < 0.001). There was no correlation between the variables in either region under study. Conclusions: The mechanical pressure required to reach the deep cervical and thoracolumbar fasciae was similar with an average value of approximately 6 N/cm2, suggesting a consistent mechanical response across these anatomical regions. Mechanosensitivity and subcutaneous adipose tissue thickness did not influence the mechanical pressure needed to access the deep fascial tissue. Full article
(This article belongs to the Special Issue Sensors and Data Analysis for Biomechanics and Physical Activity)
Show Figures

Graphical abstract

26 pages, 2661 KB  
Article
Simulated Microgravity Attenuates Stretch Sensitivity of Mechanically Gated Channels in Rat Ventricular Myocytes
by Andrey S. Bilichenko, Alexandra D. Zolotareva, Olga V. Kamkina, Valentin I. Zolotarev, Anastasia S. Rodina, Viktor E. Kazansky, Vadim M. Mitrokhin, Mitko I. Mladenov and Andre G. Kamkin
Int. J. Mol. Sci. 2025, 26(14), 6653; https://doi.org/10.3390/ijms26146653 - 11 Jul 2025
Cited by 1 | Viewed by 478
Abstract
Cardiomyocytes, similarly to cells in various tissues, are responsive to mechanical stress of all types, which is reflected in the significant alterations to their electrophysiological characteristics. This phenomenon, known as mechanoelectric feedback, is based on the work of mechanically gated channels (MGCs) and [...] Read more.
Cardiomyocytes, similarly to cells in various tissues, are responsive to mechanical stress of all types, which is reflected in the significant alterations to their electrophysiological characteristics. This phenomenon, known as mechanoelectric feedback, is based on the work of mechanically gated channels (MGCs) and mechano-sensitive channels (MSCs). Since microgravity (MG) in space, as well as simulated microgravity (SMG), changes the morphological and physiological properties of the heart, it was assumed that this result would be associated with a change in the expression of genes encoding MGCs and MSCs, leading to a change in the synthesis of channel proteins and, ultimately, a change in channel currents during cell stretching. In isolated ventricular cardiomyocytes of rats exposed to SMG for 14 days, the amount of MGCs and MSCs gene transcripts was studied using the RNA sequencing method by normalizing the amount of “raw” reads using the Transcripts Per Kilobase Million (TPM) method. Changes in the level of channel protein, using the example of the MGCs TRPM7, were assessed by the Western blot method, and changes in membrane ion currents in the control and during cardiomyocyte stretching were assessed by the patch-clamp method in the whole-cell configuration. The data obtained demonstrate that SMG results in a multidirectional change in the expression of genes encoding various MGCs and MSCs. At the same time, a decrease in the TPM of the MGCs TRPM7 gene leads to a decrease in the amount of TRPM7 protein. The resulting redistribution in the synthesis of most channel proteins leads to a marked decrease in the sensitivity of the current through MGCs to cell stretching and, ultimately, to a change in the functioning of the heart. Full article
(This article belongs to the Special Issue New Insights into Cardiac Ion Channel Regulation 3.0)
Show Figures

Figure 1

20 pages, 8199 KB  
Article
Piezo-Type Mechanosensitive Ion Channel Component 1 (PIEZO1) as a Potential Prognostic Marker in Renal Clear Cell Carcinoma
by Paulina Antosik, Martyna Szachniewicz, Michał Baran, Klaudia Bonowicz, Dominika Jerka, Ewelina Motylewska, Maciej Kwiatkowski, Maciej Gagat and Dariusz Grzanka
Int. J. Mol. Sci. 2025, 26(14), 6598; https://doi.org/10.3390/ijms26146598 - 9 Jul 2025
Viewed by 1202
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of kidney cancer and is often diagnosed at advanced stages. PIEZO1, a mechanosensitive ion channel, has been implicated in cancer progression, but its prognostic relevance in ccRCC remains unclear. This study [...] Read more.
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of kidney cancer and is often diagnosed at advanced stages. PIEZO1, a mechanosensitive ion channel, has been implicated in cancer progression, but its prognostic relevance in ccRCC remains unclear. This study aimed to evaluate the expression pattern of PIEZO1 in ccRCC and its association with clinicopathological characteristics and patient survival. Immunohistochemical analysis was performed on formalin-fixed, paraffin-embedded tumor tissues from 111 patients with ccRCC, along with 23 matched peritumoral non-cancerous tissues. Protein expression was quantified using the H-score system. Associations with tumor grade, staging, and overall survival (OS) were analyzed. mRNA expression data were retrieved from The Cancer Genome Atlas (TCGA) to validate the protein-level findings. Functional enrichment and pathway analyses were conducted to explore the biological context of PIEZO1-related gene expression. PIEZO1 showed predominantly cytoplasmic localization, with significantly lower expression in tumor tissues compared to adjacent non-malignant tissue (p < 0.0001). High PIEZO1 expression was correlated with higher tumor grade (p = 0.0147) and shorter OS (p = 0.0047). These findings were confirmed at the mRNA level in the TCGA cohort. Multivariate Cox regression analysis identified PIEZO1 as an independent prognostic factor for OS. In conclusion, PIEZO1 may serve as a clinically relevant biomarker in ccRCC. Its overexpression is associated with more aggressive tumor characteristics and poor prognosis, underscoring the need for further investigation into its functional role and potential as a therapeutic target. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

19 pages, 2479 KB  
Article
Yoda1 Inhibits TGFβ-Induced Cardiac Fibroblast Activation via a BRD4-Dependent Pathway
by Perwez Alam, Sara M. Stiens, Hunter J. Bowles, Hieu Bui and Douglas K. Bowles
Cells 2025, 14(13), 1028; https://doi.org/10.3390/cells14131028 - 4 Jul 2025
Viewed by 1132
Abstract
Fibrosis represents a pivotal pathological process in numerous diseases, characterized by excessive deposition of extracellular matrix (ECM) that disrupts normal tissue architecture and function. In the heart, cardiac fibrosis significantly impairs both structural integrity and functional capacity, contributing to the progression of heart [...] Read more.
Fibrosis represents a pivotal pathological process in numerous diseases, characterized by excessive deposition of extracellular matrix (ECM) that disrupts normal tissue architecture and function. In the heart, cardiac fibrosis significantly impairs both structural integrity and functional capacity, contributing to the progression of heart failure. Central to this process are cardiac fibroblasts (CFs), which, upon activation, differentiate into contractile myofibroblasts, driving pathological ECM accumulation. Transforming growth factor-beta (TGFβ) is a well-established regulator of fibroblast activation; however, the precise molecular mechanisms, particularly the involvement of ion channels, remain poorly understood. Emerging evidence highlights the regulatory role of ion channels, including calcium-activated potassium (KCa) channels, in fibroblast activation. This study elucidates the role of ion channels and investigates the mechanism by which Yoda1, an agonist of the mechanosensitive ion channel Piezo1, modulates TGFβ-induced fibroblast activation. Using NIH/3T3 fibroblasts, we demonstrated that TGFβ-induced activation is regulated by tetraethylammonium (TEA)-sensitive potassium channels, but not by specific K⁺ channel subtypes such as BK, SK, or IK channels. Intriguingly, Yoda1 was found to inhibit TGFβ-induced fibroblast activation through a Piezo1-independent mechanism. Transcriptomic analysis revealed that Yoda1 modulates fibroblast activation by altering gene expression pathways associated with fibrotic processes. Bromodomain-containing protein 4 (BRD4) was identified as a critical mediator of Yoda1’s effects, as pharmacological inhibition of BRD4 with JQ1 or ZL0454 suppressed TGFβ-induced expression of the fibroblast activation marker Periostin (Postn). Conversely, BRD4 overexpression attenuated the inhibitory effects of Yoda1 in both mouse and rat CFs. These results provide novel insights into the pharmacological modulation of TGFβ-induced cardiac fibroblast activation and highlight promising therapeutic targets for the treatment of fibrosis-related cardiac pathologies. Full article
(This article belongs to the Section Cells of the Cardiovascular System)
Show Figures

Figure 1

12 pages, 3452 KB  
Article
Unveiling the Role of Hydrogel Stiffness Threshold in Schwann Cell Context: Regulating Adhesion Through TRIP6 Gene Expression
by Fang Liu, Mengjie Xu, Yi Cao, Weiyan Wu, Chunzhen Jiang, Feng Li, Yifan Li, Yumin Yang and Jianghong He
Coatings 2025, 15(7), 753; https://doi.org/10.3390/coatings15070753 - 25 Jun 2025
Viewed by 2816
Abstract
Adhesion between Schwann cells (SCs, a type of glial cell in the peripheral nervous system) and their underlying substrates is a fundamental process that holds critical importance for the proper functioning of the peripheral nervous system. Conducting further in-depth research into the adhesion [...] Read more.
Adhesion between Schwann cells (SCs, a type of glial cell in the peripheral nervous system) and their underlying substrates is a fundamental process that holds critical importance for the proper functioning of the peripheral nervous system. Conducting further in-depth research into the adhesion mechanisms of nerve cells is of paramount significance, as it can pave the way for the development of highly effective biomaterials and facilitate the repair of nerve injuries. Thyroid Receptor Interaction Protein 6 (TRIP6), a member of the ZYXIN family of LIM domain-containing proteins, serves as a key component of focal adhesions. It plays a pivotal role in regulating a diverse array of cellular responses, including the reorganization of the actin cytoskeleton and cell adhesion. Accumulated data indicate that RSC96 cells (rat Schwann cells), which are rat Schwann cells, exhibit integrin-based mechanosensitivity during the initial phase of adhesion, specifically within the first 24 h. This enables the cells to sense and respond to alterations in matrix stiffness. The results of immunofluorescence staining experiments revealed intriguing findings. An increase in matrix stiffness not only led to significant changes in the morphological parameters of RSC96 ells, such as circularity, aspect ratio, and cell spreading area, but also enhanced the expression levels of TRIP6, focal adhesion kinase (FAK), and vinculin within these cells. These changes collectively promoted the adhesion of RSC96 cells to the matrix. Furthermore, when TRIP6 expression was silenced in RSC96 cells cultured on hydrogels, a notable decrease in the expression of both FAK and vinculin was observed. This, in turn, had a detrimental impact on cell adhesion. In summary, the present study strongly suggests that TRIP6 may play a crucial role in promoting the adhesion of RSC96 cells to polyacrylamide hydrogels with varying stiffness. This research not only offers a fresh perspective on the study of the integrin-mediated force regulation of cell adhesion but also lays a solid foundation for potential applications in tissue engineering, regenerative medicine, and other related fields. Full article
Show Figures

Figure 1

37 pages, 3382 KB  
Review
Mechanical Modulation, Physiological Roles, and Imaging Innovations of Intercellular Calcium Waves in Living Systems
by Cole Mackey, Yuning Feng, Chenyu Liang, Angela Liang, He Tian, Om Prakash Narayan, Jiawei Dong, Yongchen Tai, Jingzhou Hu, Yu Mu, Quang Vo, Lizi Wu, Dietmar Siemann, Jing Pan, Xianrui Yang, Kejun Huang, Thomas George, Juan Guan and Xin Tang
Cancers 2025, 17(11), 1851; https://doi.org/10.3390/cancers17111851 - 31 May 2025
Cited by 1 | Viewed by 2614
Abstract
Long-range intercellular communication is essential for multicellular biological systems to regulate multiscale cell–cell interactions and maintain life. Growing evidence suggests that intercellular calcium waves (ICWs) act as a class of long-range signals that influence a broad spectrum of cellular functions and behaviors. Importantly, [...] Read more.
Long-range intercellular communication is essential for multicellular biological systems to regulate multiscale cell–cell interactions and maintain life. Growing evidence suggests that intercellular calcium waves (ICWs) act as a class of long-range signals that influence a broad spectrum of cellular functions and behaviors. Importantly, mechanical signals, ranging from single-molecule-scale to tissue-scale in vivo, can initiate and modulate ICWs in addition to relatively well-appreciated biochemical and bioelectrical signals. Despite these recent conceptual and experimental advances, the full nature of underpinning mechanotransduction mechanisms by which cells convert mechanical signals into ICW dynamics remains poorly understood. This review provides a systematic analysis of quantitative ICW dynamics around three main stages: initiation, propagation, and regeneration/relay. We highlight the landscape of upstream molecules and organelles that sense and respond to mechanical stimuli, including mechanosensitive membrane proteins and cytoskeletal machinery. We clarify the roles of downstream molecular networks that mediate signal release, spread, and amplification, including adenosine triphosphate (ATP) release, purinergic receptor activation, and gap junction (GJ) communication. Furthermore, we discuss the broad pathophysiological implications of ICWs, covering pathophysiological processes such as cancer metastasis, tissue repair, and developmental patterning. Finally, we summarize recent advances in optical imaging and artificial intelligence (AI)/machine learning (ML) technologies that reveal the precise spatial-temporal-functional dynamics of ICWs and ATP waves. By synthesizing these insights, we offer a comprehensive framework of ICW mechanobiology and propose new directions for mechano-therapeutic strategies in disease diagnosis, cancer immunotherapies, and drug discovery. Full article
(This article belongs to the Special Issue Cancer Mechanosensing)
Show Figures

Figure 1

26 pages, 8704 KB  
Article
Genome-Wide Identification, Phylogeny, and Abiotic Stress Response Analysis of OSCA Family Genes in the Alpine Medicinal Herb Notopterygium franchetii
by Qi-Yue Zhang, Xiao-Jing He, Yan-Ze Xie, Li-Ping Zhou, Xin Meng, Jia Kang, Cai-Yun Luo, Yi-Nuo Wang, Zhong-Hu Li and Tian-Xia Guan
Int. J. Mol. Sci. 2025, 26(11), 5043; https://doi.org/10.3390/ijms26115043 - 23 May 2025
Viewed by 710
Abstract
Hyperosmolality-gated calcium-permeable cation channel protein denoted as OSCA, which are mechanosensitive pore-forming ion channels, play a pivotal role in plants’ responses to abiotic stressors. Notopterygium franchetii, an endemic perennial plant species distributed in the Qinghai–Tibetan Plateau and its adjacent high-altitude regions, is [...] Read more.
Hyperosmolality-gated calcium-permeable cation channel protein denoted as OSCA, which are mechanosensitive pore-forming ion channels, play a pivotal role in plants’ responses to abiotic stressors. Notopterygium franchetii, an endemic perennial plant species distributed in the Qinghai–Tibetan Plateau and its adjacent high-altitude regions, is likely to have undergone adaptive evolution in response to extreme abiotic stress conditions. The current study was conducted to characterize the genome-wide characteristics and phylogenetic evolution of the OSCA gene family in N. franchetii and identify its response patterns to drought and high-temperature stresses. We examined the gene family’s structural features, phylogenetic relationships, and response to abiotic stresses. The N. franchetii genome had 29 OSCA gene family members on 11 chromosomes. Subcellular localization showed they were mainly in the cell membrane, and a promoter cis-acting element study found that the OSCA gene family contained methyl jasmonate, abscisic acid, and various adversity and hormone response components. Under drought stress, most of the NofOSCAs genes showed a tendency to increase over time in the roots of N. franchetii, while in the aboveground parts, most of the NofOSCAs genes showed a tendency to increase and then decrease. The expression of different NofOSCAs genes in N. franchetii also showed alternating changes under high-temperature stress. Nine members of NofOSCAs were found to be linked to the PPI network, and these members were involved in membrane structure, transmembrane transport, and ion channel function. Our analysis of differential expression revealed that the expression of OSCA genes differed among the different N. franchetii tissues, with the roots exhibiting the highest average expression level, and many genes displayed tissue-specific high expression patterns. These results provided novel insights into the phylogenetic evolution and abiotic stress response mechanisms in the high-altitude medicinal herb N. franchetii. Full article
(This article belongs to the Special Issue Plant Genome Evolution and Environmental Adaptation)
Show Figures

Figure 1

32 pages, 2810 KB  
Review
Mechanosignaling in Osteoporosis: When Cells Feel the Force
by Nuo Chen, Marina Danalache, Chen Liang, Dorothea Alexander and Felix Umrath
Int. J. Mol. Sci. 2025, 26(9), 4007; https://doi.org/10.3390/ijms26094007 - 24 Apr 2025
Cited by 3 | Viewed by 2675
Abstract
Bone is a highly mechanosensitive tissue, where mechanical signaling plays a central role in maintaining skeletal homeostasis. Mechanotransduction regulates the balance between bone formation and resorption through coordinated interactions among bone cells. Key mechanosensing structures—including the extracellular/pericellular matrix (ECM/PCM), integrins, ion channels, connexins, [...] Read more.
Bone is a highly mechanosensitive tissue, where mechanical signaling plays a central role in maintaining skeletal homeostasis. Mechanotransduction regulates the balance between bone formation and resorption through coordinated interactions among bone cells. Key mechanosensing structures—including the extracellular/pericellular matrix (ECM/PCM), integrins, ion channels, connexins, and primary cilia, translate mechanical cues into biochemical signals that drive bone adaptation. Disruptions in mechanotransduction are increasingly recognized as an important factor in osteoporosis. Under pathological conditions, impaired mechanical signaling reduces bone formation and accelerates bone resorption, leading to skeletal fragility. Defects in mechanotransduction disrupt key pathways involved in bone metabolism, further exacerbating bone loss. Therefore, targeting mechanotransduction presents a promising pharmacological strategy for osteoporosis treatment. Recent advances have focused on developing drugs that enhance bone mechanosensitivity by modulating key mechanotransduction pathways, including integrins, ion channels, connexins, and Wnt signaling. A deeper understanding of mechanosignaling mechanisms may pave the way for novel therapeutic approaches aimed at restoring bone mass, mechanical integrity, and mechanosensitive bone adaptation. Full article
(This article belongs to the Special Issue Molecular Biology of Osteoporosis)
Show Figures

Figure 1

40 pages, 4060 KB  
Review
Mechanotransduction in Development: A Focus on Angiogenesis
by Simona Alibrandi, Carmela Rinaldi, Sergio Lucio Vinci, Alfredo Conti, Luigi Donato, Concetta Scimone, Antonina Sidoti and Rosalia D’Angelo
Biology 2025, 14(4), 346; https://doi.org/10.3390/biology14040346 - 27 Mar 2025
Cited by 1 | Viewed by 3413
Abstract
Cells respond to external mechanical cues and transduce these forces into biological signals. This process is known as mechanotransduction and requires a group of proteins called mechanosensors. This peculiar class of receptors include extracellular matrix proteins, plasma membrane proteins, the cytoskeleton and the [...] Read more.
Cells respond to external mechanical cues and transduce these forces into biological signals. This process is known as mechanotransduction and requires a group of proteins called mechanosensors. This peculiar class of receptors include extracellular matrix proteins, plasma membrane proteins, the cytoskeleton and the nuclear envelope. These cell components are responsive to a wide spectrum of physical cues including stiffness, tensile force, hydrostatic pressure and shear stress. Among mechanotransducers, the Transient Receptor Potential (TRP) and the PIEZO family members are mechanosensitive ion channels, coupling force transduction with intracellular cation transport. Their activity contributes to embryo development, tissue remodeling and repair, and cell homeostasis. In particular, vessel development is driven by hemodynamic cues such as flow direction and shear stress. Perturbed mechanotransduction is involved in several pathological vascular phenotypes including hereditary hemorrhagic telangiectasia. This review is conceived to summarize the most recent findings of mechanotransduction in development. We first collected main features of mechanosensitive proteins. However, we focused on the role of mechanical cues during development. Mechanosensitive ion channels and their function in vascular development are also discussed, with a focus on brain vessel morphogenesis. Full article
(This article belongs to the Section Cell Biology)
Show Figures

Figure 1

20 pages, 7299 KB  
Article
Piezo1 Regulates Odontogenesis via a FAM83G-Mediated Mechanism in Dental Papilla Cells In Vitro and In Vivo
by Xinyue Sheng, Jingzhou Li, Haozhen Ma, Hongwen He, Qin Liu, Shilin Jia, Fuping Zhang and Fang Huang
Biomolecules 2025, 15(3), 316; https://doi.org/10.3390/biom15030316 - 20 Feb 2025
Viewed by 1140
Abstract
This study explored the role of Piezo1 in the odontogenic differentiation of dental papilla cells (DPCs) and tissue, focusing on a mechanism involving family with sequence similarity 83, member G (FAM83G). Here, we found Piezo1, a mechanosensitive cation channel, was upregulated during odontogenesis [...] Read more.
This study explored the role of Piezo1 in the odontogenic differentiation of dental papilla cells (DPCs) and tissue, focusing on a mechanism involving family with sequence similarity 83, member G (FAM83G). Here, we found Piezo1, a mechanosensitive cation channel, was upregulated during odontogenesis in DPCs and dental papilla tissues. Knockdown of Piezo1 impaired odontogenic differentiation, while its activation by Yoda1 enhanced the process. Using a 3D culture model and an ectopic transplantation model, we confirmed Piezo1’s role in vivo. RNA sequencing (RNA-seq) analysis revealed that FAM83G was upregulated in Piezo1-knockdown cells, and FAM83G silencing enhanced odontogenesis in DPCs. These findings indicate that Piezo1 positively regulates odontogenesis by inhibiting FAM83G in DPCs both in vitro and in vivo, with Piezo1 representing a potential target for dental tissue regeneration. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Back to TopTop