Next Article in Journal
The Role of IL-23 in the Development of Inflammatory Diseases
Previous Article in Journal
Alpine Adaptive Mechanism on Rhizosphere Microbes Recruitment of Crepis napifera (Franch.) Babc. by Multi-Omics Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanotransduction in Development: A Focus on Angiogenesis

1
Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
2
Department of Biomolecular Strategies, Genetics, Cutting-Edge Therapies, Istituto Euro-Mediterraneo di Scienza e Tecnologia (I.E.ME.S.T.), Street Michele Miraglia 20, 90139 Palermo, Italy
3
Neuroradiology Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Street Consolare Valeria 1, 98125 Messina, Italy
4
IRCCS Istituto Delle Scienze Neurologiche di Bologna, Street Altura 3, 40123 Bologna, Italy
5
Department of Biomedical and NeuroMotor Sciences (DiBiNeM), Alma Mater Studiorum—University of Bologna, 40127 Bologna, Italy
*
Author to whom correspondence should be addressed.
Biology 2025, 14(4), 346; https://doi.org/10.3390/biology14040346
Submission received: 8 March 2025 / Revised: 22 March 2025 / Accepted: 25 March 2025 / Published: 27 March 2025
(This article belongs to the Section Cell Biology)

Simple Summary

This review article aims to describe the main features of a specific type of proteins mainly exposed on the cell surface. These proteins share the mechanism by which they respond to external mechanical stimuli in a process called mechanotransduction. By this process, mechanosensitive proteins act as receptors, converting these mechanical cues into biological signals that are elaborated within the cell. Several biochemical functions are controlled by mechanical stimuli including vascular development. Therefore, the role of mechanosensitive proteins in development and blood vessel formation is extensively discussed. The development of brain vessels is also addressed.

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 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.

1. Introduction

Mechanotransduction refers to the cell ability to respond to mechanical stimuli. Both exogenous and endogenous cues continuously act on plasma membrane, inducing the cell to generate specific biological responses. Generated biochemical signals are not uniform; rather they depend on several force properties such as direction, magnitude, duration. Extrinsic stimuli including shear stress, blood flow, hydrostatic pressure, tension and matrix stiffness usually originate in the extracellular environment. On the other hand, intracellular tension contributes to reciprocal cell–cell mechanical regulation. This tension arises from forces related to actin polymerization and myosin contraction, as membrane curvature, cytoskeletal protrusive and traction cues, and nuclear envelope deformation. As biological significance, all molecular events involved in this signal transduction result in the modulation of gene expression, post-translational modifications and subcellular protein redistribution and they are known as mechanotransduction [1,2,3].
This process is allowed by specific proteins that enable both structural and biochemical communication between the extracellular matrix (ECM) and the subcellular structures, like the membrane, cytoskeleton and nucleus. Proteins responsive to mechanical cues are known as mechanosensors and comprise ion channels, integrins and transcription factors. All these proteins are capable of changing their conformation or subcellular localization, when a force is applied [4]. However, in polarized cells also the primary cilium responds to mechanical stimuli [5]. The primary cilium controls cell shape and motility during development and differentiation, since embryo stage to adulthood. In addition, stiffness, adhesion and tension are dynamic cues that make mechanotransduction a spatio-temporal specific response. Therefore, several physiological and pathological cell processes, as tissue remodelling, axon guidance, adhesion, tissue repair, autophagy and cancer are controlled by mechanotransduction [6,7,8,9,10]. For this reason, mechanotransduction is often accompanied by changes in cell metabolism [11,12,13]. In the last years, knowledge about the role of mechanosensation in regulating cell metabolism has rapidly increasing. This review summarizes the main findings on the different mechanoreceptors and is conceived to discuss how mechanotransduction contributes to development. In particular, blood vessel morphogenesis is considered, with a focus on blood–brain barrier (BBB) and its property maintaining.

1.1. Mechanosensors and Their Classification

Mechanosensation is not limited to a single family of proteins. Several membrane receptors, cytoskeletal proteins and transcription factors exhibit mechanosensitive properties. However, main concerns are on ion channels and the way by these can be considered as mechanotransducers. For this reason, criteria to classify an ion channel a mechanosensor were proposed by Arnadóttir et al. [14] and include: (i) presence of a pore-forming subunit for rapid ion conductance; (ii) ability to gate upon tension application; (iii) altered conductance upon structural defects of the channel domains; (iv) mechanosensation in a non-mechanosensitive cell upon forced expression of the channel; (v) both gene and protein mandatory expression in the mechanosensitive cell; (vi) abolition of cell mechanosensation due to mechanochannel gene loss of function. As above mentioned, several membrane proteins have been surprisingly found to be responsive to mechanical cues. However, all mechanosensors act by changing their conformation when cues are applied. This conformational change results in a biochemical response within the cell [15]. In general, signal transduction following a mechanical cue can occur by two different mechanisms. The first one strictly depends on membrane lipid bilayer deformation, following force application. This modification is perceived by integral membrane proteins, including enzymes and ion channels. As consequence, they become active triggering biochemical responses [16]. The second mechanism is based on both physical and functional continuity between ECM and cytoskeleton. In this context, forces directly act on membrane proteins, further responsive to stimuli coming from adjacent cells. Then, cues induce conformational changes such as to modify protein interaction pattern [17]. In this way, mechanosensing is even perceived to the nucleus [18].

1.2. The ECM–Membrane–Cytoskeleton–Nucleus Axis in Mechanosensing

Living cells are able to convert ECM signals into biochemical events. This requires a large number of proteins to make cells responsive. Extracellular matrix mechanical cues drive several processes as differentiation, proliferation, apoptosis, degeneration and malignancy through physical continuity among ECM, membrane proteins and cytoskeleton. Then, intracellular signals are further propagated within the nucleoplasm via the nuclear envelope [19]. Looking at this complex machinery, collagen, fibronectin, laminins are the main mechanical inducers in the ECM. Intracellular transmission is mediated by multimeric membrane receptors known as integrins. According to tissue distribution, their heterogeneous composition makes each cytotype differentially responsive to specific cues [20]. Other membrane proteins involved in mechanotransduction include receptors for extracellular soluble ligands, as transforming growth beta factor receptors (TGFβRs) or Wnt family members, and adhesion molecules [21,22,23]. Within the cell, stiffness and tension are transmitted to cytoskeleton by several integrin-associated proteins as vinculin, paxillin and talin [24]. In particular, talin was the first mechanosensitive cytoskeleton-associated protein, able to endure cues due to its structure. At the C-terminus, indeed, it contains 13 mechanosensitive rod subdomains that spread forces by switching between the unfolded/folded conformation [25]. By binding F-actin, talin transmits forces to the microfilament/myosin complexes that trigger contractility and, then, cell morphological change and protein subcellular redistribution. In this field, role of β-catenin was largely investigated. It usually localizes at adherens junctions (AJs) to stabilize cell–cell interaction. However, following mechanical stimulation, β-catenin is also able to translocate within the nucleus, where regulates expression of mechanosensitive genes [8,26]. It was shown that cytoskeletal cues are able to induce nuclear pore complex dilation, allowing nuclear shuttling of mechanosensitive transcription factors as the Krüppel-like Factor 2/4 (KLF), the Yes-associated protein (YAP) and the Transcriptional coactivator with PDZ-binding motif (TAZ) [27]. Adhesion proteins also participate in mechanotransduction. In particular, at AJs, proteins undergo conformational changes when a mechanical cue is applied; then, extracellular domains of the cadherins exposed on adjacent cells more strongly interact, tightening plasma membranes [28]. Likewise, their cytoplasmic tail binds actin. In this complex, the p120-catenin and the β-catenin contribute to cell junction stability [29]. The cytoskeleton responds to mechanical cues by further generating protrusive and traction forces. Traction requires myosin and allows intracellular signal transmission to ECM and neighbor cells [30]. Protrusive forces, instead, depend on actin polymerization and can result in filipodia formation, in certain cell types as neurons. Their dynamics mainly depends on extracellular ion concentration [31]. However, a recent study suggested that focal adhesions, actin orientation and intracellular force distribution are driven by cell morphology and, according to the adhesion curvature, they are able to pilot stem cell differentiation [32]. As mentioned, nucleus itself responds to forces by regulating envelope and nucleoskeleton remodelling, chromatin redistribution and histone modification. The Linker of Nucleoskeleton and Cytoskeleton (LINC) is the term coined to indicate this functional continuity between cytoskeleton and nucleoplasm. Structurally, LINC comprises two different protein families, nesprins and SUN proteins. By the cytoplasmic region, nesprins 1 and 2 bind actin, nesprin 3 binds intermediate filaments through the plectin, and nesprin 4 binds microtubule-associated proteins to be connected with microtubules. Nesprins span within the nuclear envelop where bind SUN proteins, that, in the nucleoplasm, are linked to the lamin A and lamin C. The SUN/lamin/chromatin complex is stabilized by emerin, acting as mechanosensor for the nucleus [33]. This mechanism is known as direct and it is such to lead heterochromatin redistribution at the inner nuclear membrane (Figure 1).
Conversely, another type of nuclear mechanotransduction called indirect depends on the monomer (G): polymer (F) actin ratio. Actin polymerization as response to certain mechanical cues, indeed, promotes nuclear shuttling of the Mixed-Lineage Kinase 1 (MLK1), a transcription factor targeting the serum response factor. In the absence of mechanical stimuli, MLK1 is retained in the cytoplasm through its binding to the G-actin [34]. Failed mechanotransduction at the nuclear envelope results in biogenesis of micronuclei, genome fragmentation and chromatin release into the cytoplasm [35]. This phenomenon usually occurs due to defects in LINC proteins. Laminopathies comprise more than 15 different phenotypes, as the Hutchinson–Gilford progeria syndrome [36] or the Greenberg dysplasia [37], arising due to mutations in lamin A (LMNA) and lamin B receptor (LBR) coding genes, respectively [38]. However, lamin dysfunctions have also been reported in cancer [39,40].

2. Mechanosensitive Ion Channels

Mechanosensitive ion channels (MCs) include heterogeneous membrane receptors activated by external mechanical cues. They show tissue-specific expression and selective responsiveness to specific stimuli. However, their common function is the intracellular ion flux following their activation. Mechanosensitive ion channels usually carry cations and they may be more or less selective [41]. The most characterized families include the epithelial sodium channel (ENaC) family, the TWIK-Related K+ (TREK) channels, the Big Potassium (BK) channels, the G-Protein Coupled Receptors (GPCRs), the Transient Receptor Potential (TRP) superfamily and the Piezo channels (Table 1). The ENaC family comprises 4 members contributing to electrolyte balance in epithelial tissues by regulating Na+ reabsorption, following hormone stimulation or proteolytic cleavage [42,43]. However, they can act as mechanotransducers in sensory neurons as well as in artery endothelial cells (ECs), despite the tethering mechanism is still unclear [44]. The TREK channels are three mechanogated two-pore domain K+ channels activated by membrane tension and expressed in heart, smooth muscle cells and nervous system, in particular at nodes of Ranvier [45]. Large-conductance voltage- and Ca2+- activated K+ channels (BK channels) regulate cell excitability by controlling K+ flux and Ca2+ signaling. They are mainly expressed in muscle cells where they enhance repolarization, in kidney, in vascular muscle cells and in central nervous system (CNS) [46,47,48,49]. Pathological conditions related to BK channel impairment include epilepsy, ataxia, hypertension and erectile dysfunction [50]. Recently, several GPCRs were further described as mechanosensors responsive to cues as shear stress, vibration or mechanical stretch, in order to regulate cell adhesion [51].
In this review, we describe the TRP superfamily and the Piezo channels as pivotal in vasculature, the main focus of our discussion.

2.1. Transient Receptor Potential (TRP) Family Members

Transient receptor potential channels comprise several mechanosensitive cation channels regulated by phosphoinositides and firstly described as secondary mechanotransducers, amplifying Piezo channel-mediated primary mechanotransduction [69]. The TRP channels are classified into 7 large subfamilies, according to sequence homology. However, in mammals TRPs include the TRPC (canonical) subfamily made by 7 members, the TRPM (melastatin) subfamily made by 8 members, the TRPV (vanilloid) subfamily made by 6 members and a single TRPA (ankyrin) member. All these channels are mainly expressed in the cell membrane. The 3 TRPML (mucopilin) and the 3 TRPP (polycystins) receptors, instead, are expressed on the endoplasmic reticulum surface (Figure 2).
The TRPN (no mechanoreceptor potential C-like or NOMPC-like) subfamily has been identified only in fish and invertebrates. The TRP channels contribute to several physiological processes triggered by mechanotransduction, as cell cycle regulation, apoptosis, cell migration, chemo-osmotic stress response [70].
Briefly, the TRPC subfamily consists of Na+ and Ca2+-permeable nonselective cation channels particularly abundant in hippocampal neurons, where they play roles in memory, long-term potentiation, pain perception, anxiety [71]. With the exception of TRPC2 that is a pseudogene [72], the 6 TRPC channel are expressed in humans and, according to structural homology, they are divided into 2 subgroups, TRPC 1/4/5 and TRPC 3/6/7. They are most often activated by phospholipase C (PLC) [73] and trigger cell membrane depolarization, contributing to store-operated Ca2+ entry (SOCE) [74]. However, phenotypes related to TRPC channel disfunction depend on impaired Ca2+ balancing and can affect several organs as kidney, muscle, heart, lung, pancreas [75,76,77,78]. Moreover, TRPC gain of function enhances cell proliferation, migration and invasion in cancer [79,80]. The TRPM subfamily comprises heterogeneous members most often regulating insulin and gastric hormone release [81,82]. Therefore, they are involved in several physio-pathological processes including proliferation and cell death, cancer and neurodegeneration [83,84]. The TRPM4 and TRPM5 channels are activated by Ca2+ but are only permeable to monovalent cations [85]. The TRPM7 is ubiquitously expressed and regulates Mg2+ homeostasis [86]. The TRPM2 acts as osmo-sensor and it was the first TRP channel identified as oxidative stress sensitive [87]. TRPM8 is expressed in sensory neurons where is activated by cold stimuli [88]. Finally, the TRPM3 is activated by heat and inflammatory pain [89].
The unique TRPA member (TRPA1) is a Ca2+ channel, contains 16 ankyrin repeats and is expressed in sensory neurons of skin and lung where it is responsive to mechanical stress, and in the brain where plays a role in nociception directly linked to migraine. It was initially described as a temperature sensor, being active when temperature is lower than 12 °C [90]. Endogenous Ca2+ itself regulates TRPA1 permeability and enhances cytoplasmic Ca2+ release from the endoplasmic reticulum [91]. TRPA1 is expressed in dendritic cells, endothelial cells, macrophages, neutrophils, mast cells, T-lymphocytes and chondrocytes where it responds to reactive oxygen and nitrogen species, irritants and proinflammatory cytokines [92]. Although it mainly triggers inflammation, its protective anti-inflammatory effect in both myocardial and brain ischemia was reported [93]. Furthermore, TRPA1 activation can also induce apoptosis and its dysfunction is associated with migraine, trigeminal nociception, glial inflammation in Alzheimer’s disease, inflammatory bowel disease, peripheral pain, lung disorders [94,95]. However, the rare autosomal dominant Familial Episodic Pain Syndrome (FEPS) arises following germline mutation in the TRPA1 gene [96].
The TRPML subfamily comprises 3 members also known as mucolipins. Among the TRPs, these non-selective ion channels are the only ones expressed on endosomal surface, where they regulate membrane trafficking, vesicle pH and degradation and, then, autophagy [97]. They act as both homomeric and heteromeric complexes. In particular, TRPML1 and TRPML2 are expressed in late endosomes and lysosomes, homomeric TRPML3 is exposed on endoplasmic reticulum surface and early endosomes, while it forms heteromeric complexes in lysosomes. In addition to pH and osmolarity, mechanical tension generated by membrane tubulation also regulates TRPML activity [98]. The three TRPML proteins show different expression patterns, being TRPML1 ubiquitously expressed and, in particular, in brain, kidney, liver spleen and heart. Recessive loss of function mutations in MCOLN1, encoding for TRPML1, lead to mucolipidosis type IV (MLIV), a lysosomal storage disorder characterized by motor neurodegeneration, vision impairment and mental retardation, due to defective biogenesis of lipid-containing vacuoles [99,100]. TRPML3 is mainly expressed in sensory neurons, in the stria vascularis of the cochlea and in the organ of Corti sensory hair cells. Gain of function mutations, indeed, result in hearing loss and vestibular disorders [101]. Finally, TRPML2 expression is restricted to liver, kidney, spleen, heart and immune cells, where enhances viral endocytosis [102]. However, in immune cells as macrophages and natural killer cells, it acts as an antiviral, by promoting interferon release [103]. To date, no genetic diseases were linked to TRPML2 mutations.
Polycystins are the three components of the TRPP subfamily. They were first discovered due to their association with the autosomal dominant polycystic kidney disease (ADPKD). All members are mechanosensitive Ca2+-permeable ion channels and, among these, PC1 and PC2 are the most well characterized. They are encoded by the PKD1 and PKD2, respectively, and form heterocomplexes in the plasma membrane and in the primary cilium. In kidney primary cilium, PC1 is sensitive to urinary flow and its mechanoactivation triggers PC2, that acts as Ca2+ channel responsible for ion entry into the primary cilium and, then, into the cytoplasm [104]. This sequence of events controls tubule morphogenesis and defects of this mechanotransduction pathway result in ciliopathies, such as ADPKD [105]. Further, PC1 colocalizes with focal adhesions, where is sensitive to ECM stiffness, shear stress and adjacent cell tension. In this case, its activation results in a proteolytic cleavage by which the released peptide translocates to the nucleus, to directly regulate gene expression [106].
The TRPV or vanilloid subfamily is the most well characterized and it can be further subdivided into two groups. TRPV5 and TRPV6 are highly Ca2+-selective ion channels mostly expressed in epithelial cells, where they mediate Ca2+ entry in kidney and intestine, in physiological conditions. Germline mutations in TRPV5 and TRPV6 result in chronic kidney disease and nephrolithiasis, and in transient neonatal hyperparathyroidism, respectively [107]. TRPV1, TRPV2, TRPV3 and TRPV4 are non-selective cation channels mostly expressed in afferent neurons, where they function as somatosensors that detect temperature, nociceptive, mechanic and osmotic signals [108]. However, their expression was also confirmed in the endoplasmic reticulum, where they regulate Ca2+ homeostasis [109]. Among all TRPV members, only TRPV2 and TRPV4 have been confirmed as mechanosensors to date [110]. In particular, TRPV4 has been extensively studied due to its involvement in fibrillar collagen remodeling and ECM modulation, in accordance to its link with Charcot-Marie-Tooth disease type 2C and skeletal dysplasia [111,112]. TRPV2 mutations, instead, cause heart failure [113]. Other TRPV-related phenotypes include migraine, neuropathic pain and dry eye disease (TRPV1 gene) and Olmsted syndrome (TRPV3 gene) [114,115].

2.2. Piezo Channels

Not so far, PIEZO1 and PIEZO2 were described as mechanically activated cation permeable channels, highly conserved in Vertebrates. In particular, PIEZO2 has nonselective conductance, while PIEZO1 is mainly Ca2+ selective [116]. They are very large proteins, located in plasma membrane, endoplasmic reticulum and nuclear membrane, differentially expressed across various cell types. During both embryo development and in adulthood they are activated by several stimuli including shear stress, membrane tension, cell compression. PIEZO1 is abundant in epithelial and endothelial tissues, bone and muscle, while PIEZO2 is largely expressed in sensory neurons, dorsal root ganglia and epithelial Merkel cells. However, both channels have baroceptor activity regulating blood pressure in the CNS [117]. Both the force-from-lipids and the force-from-filaments hypotheses have been proposed as mechanisms underlying PIEZO gating [118,119,120]. However, their biological activity is further modulated by several membrane proteins, as cadherins, TRPM4 and TMEM150C, which interact with PIEZO channels enhancing their function [121]; likewise, PECAM1, MTMR2 or PC2 act by reducing PIEZO activity [122,123,124] (Figure 3).
PIEZO1-mediated mechanosensing was initially described as a driver of EC polarity, in response to blood flow and as a key regulator of vascular remodeling [125,126]. However, it also responds to mechanical cues including membrane stretch and stiffness, tissue compression and distension [127,128,129,130]. Following activation, PIEZO1 opening increases intracellular Ca2+, that acts as second messenger [131]. PIEZO1 constitutive activation was shown to trigger mitochondrion-mediated apoptosis due to imbalanced Ca2+ homeostasis and, consequently, cytotoxic cation overload [132]. Likewise, dominant gain of function mutations result in hereditary xerocytosis, a hemolytic anemia characterized by primary erythrocyte dehydration [133]. In contrast, recessive loss of function mutations cause congenital lymphatic dysplasia [134] and, in particular, the lymphatic malformation-6 phenotype [135]. Finally, a novel PIEZO1 missense mutation was recently shown to segregate within a family, together with the cerebral cavernous malformation (CCM) phenotype [136].
PIEZO2 is still poorly characterized. It was shown to be involved in auditory hair cell development [137]. Its expression abounds in sensory neurons, where triggers action potential and depolarization, following mechanogating [138]. Dominant phenotypes linked to PIEZO2 gain of function mutations include Gordon syndrome, Marden-Walker syndrome and distal arthrogryposis type 5 [139]. Neurosensory deficits and skeletal defects, instead, arise following loss of function mutations [140,141].

2.3. Intracellular Mechanotransduction: Receptors for Soluble Ligands as Mechanosensors

Beyond integrins, receptors for extracellular soluble ligands can be mechanically activated, suggesting how biochemical and mechanical signals integrate to induce expression of mechanosensitive genes. Among the activated receptors, TGF-β and Wnt family members are responsive to mechanical stimuli. In many cases, intracellular transduction pathways converge on activation of mechanosensitive genes, to adapt cell homeostasis in response to mechanical force. Among these, SMAD, β-catenin, YAP/TAZ, Klf2/4 and ERK proteins are the most involved. SMAD proteins are activated downstream to the TGF-β signal and their biological activity is essential from early embryo development [142]. In the field of mechanotransduction, the TGF-β signal is involved in cell differentiation and several studies confirmed how extracellular cues can over-activated it, resulting in stemness acquisition or fibrotic phenotype. Differentiation loss was reported in human embryonic stem cells, when the TGF-β/Activin/Nodal signaling is amplified under force application [143]. This cascade was further characterized in kidney, where the primary cilium responds to fluid shear stress, not exclusively through polycystin activation. To regulate glomerular filtration, indeed, also TGF-β1 responds to shear stress and activates SMAD2/3 signal in renal epithelial cells. This signal is antagonized by Notch4 and its perturbation results in fibrotic phenotypes [144]. More in detail, shear stress enhances the expression of ligands for TGF-β and activin receptors. Downstream, SMAD2/3 over-activation results in increased Snai1 and reduced Cdh1 expression, enhancing epithelial differentiation loss and stemness acquisition [145]. Likewise, primary corneal keratocytes can differentiate into myofibroblasts, when the focal adhesion kinase and the TGF-β1 signal are activated. However, also in this case, differentiation rate depends on matrix stiffness and, then, on signal intensity [146]. The focal adhesion kinase complex was also shown to be triggered by matrix stiffness in cardiomyocytes, during cardiac development. Downstream, a multitude of signaling pathways are activated to control cell fate [147]. In detail, the PI3K/AKT and p38/JNK pathways ensure the expression of positive regulators of cardiac morphogenesis. In concerto, the YAP/TAZ, the AKT/TSC2/mTOR and ERK1/2 pathways control proliferation rate [148,149,150]. In cardiomyocytes, expression of mechanosensitive genes is also promoted by the non-canonical Wnt5a/Wnt11 pathway, in presence of pressure overload. In this context, the signal involves the TEAD1-YAP cascade that results in cardiomyocyte failure, suggesting how Wnt5a over-activation due to excessive mechanical stimuli can result in heart contractile dysfunction [151]. Recently, Wnt5a over-expression was reported in human artery ECs, when exposed to disturbed flow. This resulted in over-expression of its receptor frizzled-4 (FZD4) and, downstream, in β-catenin activation, and pro-inflammatory cytokine biosynthesis, stress fiber formation, contractile and atheroprone phenotype acquisition. These results were not observed under normal flow, further suggesting the role of disturbed hemodynamics in vascular phenotypes [152]. Blood flow also controls atrioventricular valve morphogenesis in zebrafish embryo, as shown by Paolini et al. In detail, shear stress activates both Notch and Klf2 signaling in endocardial cells; here, dll4-positive endocardial cells undergo lateral inhibition by Notch, become able to migrate in ECM and respond to paracrine Wnt9a, further produced by the Erk5-Klf2-Wnt9a cascade, upon flow stimulation [153]. The Wnt/β-catenin pathway also controls epithelial-to-mesenchymal transition in liver cancer cells, in a matrix stiffness-dependent manner. In particular, stiffer matrices enhance the expression of the long noncoding RNA (lncRNA) NEAT1, notoriously involved in liver cancer progression. Its role as positive cell cycle regulator and epithelial-to-mesenchymal transition promoter was reported, following its association with increased PCNA, N-cadherin and vimentin expression [154,155]. Interestingly, it was recently shown that NEAT1 overexpression can induce stemness via the canonical Wnt/β-catenin pathway, suggesting as ECM properties can influence liver cancer progression.
Finally, the ERK pathway is also activated within cells, when shear stress, compression or tension are applied. In epithelial cells, increased ERK phosphorylation was observed after activation of the epidermal growth factor receptor by stretching of adjacent cells, resulting in polarization and motile phenotype acquisition [156]. Likewise, tension was shown to induce myosin II-dependent ERK activation, enhancing stress fiber formation in human fibroblasts [157]. Upon tension application, ERK activation can occur by two different mechanisms. The first involves a change in cell shape directly triggered by the cue [158]; the second is linked to the activation of mechanosensitive receptors on the cell surface, as PIEZO1 [159,160].
Therefore, as briefly described, cell response to mechanical cues is not uniform, despite downstream activated pathways most often involve the same proteins. Clearly, force magnitude, duration, intensity and cell type are the main parameters that determine cell fate.

3. Mechanosensing in Embryo Development

Mechanical cues drive cell fate since early stages of embryo development, controlling gastrulation and left-right symmetry breaking, gamete specification, cardiovascular development, kidney and inner ear morphogenesis, neuron migration, hematopoiesis [161,162,163,164].
During gastrulation, ECM forces induce mesodermal specification by activating the VE-cadherin/β-catenin signal, in both Drosophila and zebrafish embryos, in an evolutionary conserved manner [165]. In detail, exposure of the Y654-β-catenin phosphorylation site results from the β-catenin-VE-cadherin interaction, upon mechanical stretching. Beta-catenin phosphorylation by Src42A kinase leads to its release from junctions and its shuttling within the nucleus, where activates genes controlling morphogenic movements during gastrulation and mesoderm induction [166]. In addition, Y654-β-catenin phosphorylation is required for Brachyury expression, which is involved in endothelial-to-mesenchymal transition (EndMT), during mesoderm specification [167]. Within the early somite mesoderm, at the anterior extent of the primitive streak, a group of ciliated cells forms the node, also called the left-right organizer, involved in left-right specification [168,169]. Ciliated cells are divided in central “pit” cells and “crown” cells, exhibiting motile and immotile cilia, respectively [170]. On the crown cell surface, Wnt5A-dependent planar cell polarity drives distribution of posteriorly slanting cilia that express the Pkd2 [171]. Under nodal flow, pkd2 binds pdk1l1 driving embryo asymmetry, according to flow direction, as demonstrated in Medaka fish [172,173]. Furthermore, Pkd1l1 mutant mice exhibited an altered Nodal expression pattern, not restricted to the left side downstream of nodal flow, resulting in loss of left-right asymmetry [174]. On the left side, Nodal induces Pitx2 expression that controls organogenesis [175]. On the other hand, Nodal signaling is inhibited by Dand5. On the left dorsal side, Pkd2 expression allows Ca2+ influx following immotile cilium stimulation, leading to Dand5 degradation and consequent Nodal signal activation [176]. Dand5 further antagonizes the bone morphogenetic protein 4 (BMP) that regulates asymmetric distribution of Pkd2 in nodal immotile cilia, as recently shown by Katoh et al. [177]. Moreover, during zebrafish embryo gastrulation, tight junctions (TJs) respond to actomyosin tension controlling the separation between the enveloping cell layer and the yolk syncytial layer [178].
Embryonic stem cell differentiation is controlled by ECM composition, elasticity and viscosity. For example, fibronectin expression controls axial mesoderm extension and migration, that are enhanced by the fibrillar form of the protein and its interaction with α5β1-integrin, in Xenopus [179,180]. Matrix stiffness is not uniform; rather its gradient depends on cell density, with stiffer areas hosting a higher cell number. Therefore, cells respond to this gradient by migrating from stiffer to softer regions, as shown by neural axon pathfinding during brain development in Xenopus embryo [181,182]. Cranial neural crest migration is mechanically controlled by other several factors including contact inhibition, rear actomyosin contraction and tissue fluidity [183]. However, together with migration, mesodermal stiffness also induces epithelial-to-mesenchymal transition of neural crest cells which remain undifferentiated until they reach their target tissue. Here, ECM stiffness controls neural crest lineage specification [184]. At the membrane level, neural crest cells express mechanosensitive GPCRs and ion channels. Among the GPCRs, the endothelin receptor, the sphingosine 1-phosphate receptor and the parathyroid hormone 1 receptor induce connective tissue differentiation [185,186,187]. Likewise, Piezo1 channel negatively controls cell migration [188] (Figure 4).
In addition, dorsoventral β-actin gradient controls neural tube closure and, in particular, higher tension is observed in the ventral neural tube cells, while greater rigidity occurs in the notochord ventral regions, where Yap activity is enhanced. This activates the Sonic Hedgehog pathway and, downstream, FoxA2 expression [189]. Further, Yap/Taz signaling guarantees proliferation of progenitor hindbrain boundary cells in zebrafish embryo, in response to mechanical cues [190]. In humans, neural stem progenitor cells respond to cell-generated traction forces by PIEZO1, but not PIEZO2, in a substrate stiffness-dependent manner. This results in neural stem progenitor cells differentiation promoting neuron lineage, against the astrocyte one [127]. Likewise, stiffer substrates induces neural crest differentiation into smooth muscle cells, while soft substrates result in acquisition of glial cell phenotype in mice [191]. Finally, ECM elasticity further controls adult mesenchymal stem cell (MSC) lineage specification in-vitro, and cell reprogramming depending on matrix composition is possible during early culture stage [192,193,194].
Musculoskeletal development is also controlled by mechanical stimulation [195,196]. Growth-generated strains lead early differentiation stages, while muscle loading occurs late during tissue development [197]. In the developing muscle, perichondral cells respond by generating forces that induce skeletal biogenesis [198]. At the same time, muscle contractile tension further promotes joint specification by inhibiting expression of Sox9 and Col2a1 chondrogenic markers. After specification, joint cavitation only partially depends on muscle tension [199]. On the other hand, loss of muscle contraction during tissue development perturbs scxa and sox9a expression in developing enthesis of zebrafish embryo [200]. More in detail, Scxa and Sox9 expression increases in mesenchymal stem cells, upon TGF-β2 activation, following mechanical stimulation. The intensity of TGF-β2 response depends on cue properties and results in downstream activation of SMAD proteins and Rho/ROCK/SRF signaling to control MSC differentiation in tenocytes [201]. Together with muscle and tendon, osteoblast mobilization for skeletal development was shown to depend on paracrine signals from ECs, in response to matrix stiffness and mechanical load, in a YAP/TAZ-dependent manner [202,203]. Finally, YAP activation by mechanical cues in smooth muscle cells is responsible for digestive tract elongation [204].
Knowledge about mechanical control of tissue morphogenesis is allowing to apply biomechanics to regenerative medicine. In the last decades, adhesion matrices have been engineered to drive stem cell differentiation and restore damaged tissues, such as osteonecrotic bone or infarcted myocardium [205,206]. It was shown that ECM properties lead stem cell lineage and these findings are allowing to modulate tissue regeneration in-vitro. For examples, increased gain of cartilage phenotype was observed in MSCs-loaded scaffolds, when grown in engineered chondrocyte pericellular matrix, able to trigger the TRPV4-YAP/TAZ-PI3K-Akt signaling within the cell [207]. Likewise, superparamagnetic iron oxide nanoparticles can induce mechanotransduction in human bone marrow-derived MSCs, leading to their osteogenic differentiation, upon external magnetic field application [208]. However, cells seem to maintain a mechanical memory, in response to prolonged persistence of the force. This mechanical memory includes both physical changes as cytoskeleton remodelling and nuclear deformation, and molecular events comprising epigenetic modifications [209,210]. Several factors contribute to mechanical memory acquisition and, among these, stiffness dose, time exposure and cell type [211]. Therefore, further knowledge in this field will allow to modulate mechanical memory and, then, cell plasticity [212,213].

4. Mechanotransduction in Vasculature

4.1. Mechanical Regulation of Endothelial Tip Cell Sprouting

Vasculature morphogenesis begins already during early embryo development and continues throughout adulthood through remodeling phenomena. Correct vessel development requires integration of both biochemical and mechanical factors. Three main moments drive vessel network formation that, chronologically, are vasculogenesis, angiogenesis and arterialization [214] (Figure 5).
Vasculogenesis requires EC phenotype gain from angioblasts, previously differentiated from mesodermal cells. Endothelial progenitor cells are recruited and organized for de-novo formation of the primary capillary plexus [215]. From the primary capillary plexus, EC sprouting is promoted by both angiogenic factors and ECM remodeling. During this process, called angiogenesis, new vessels form from pre-existing capillaries and sprouting direction is driven by endothelial tip cells (ETCs) [216]. Endothelial tip cells are characterized by a protrusive phenotype, aimed to form transversal vessel connections. In contrast, endothelial stalk cells guarantee continuity with the original vessel from which sprout originates. Mechanistically, EC differentiation into the tip phenotype was recently shown to be controlled by several genes including BMPR2 and FBN1. At the cell membrane, activated BMPR2 triggers the PI3K signaling to promote actin polymerization and filopodia formation, by the small Rho GTPase CDC42 [217]. At the angiogenetic front, fibrillin-1, encoded by the FBN1 gene, transmits mechanical forces by binding integrins and syndecans [218]. Knock-out Fbn1 mice exhibit retinal ETCs with disorganized cytoskeleton and reduced migration rate. In detail, fibrillin-1 deficiency was shown to cause impairment of the VEGF-A (vascular endothelial growth factor-A)/Notch/SMAD signal, leading to a compromised stalk-tip phenotype switch [219] (Figure 6).
Biochemically, ETCs show higher oxidative metabolism further enhanced by thicker matrices, due to increased energy demand required for sprouting in dense substrates [220]. Conversely, actin-mediated pulling in ETCs was demonstrated to induce ECM deformation during sprouting [221]. To balance stalk/tip differentiation, BMP9 binds Alk1, activates the SMAD1/5 pathway that induces β-IV-spectrin expression. In differentiating ECs, the β-IV-spectrin promotes vascular endothelial growth factor receptor 2 (VEGFR2) internalization, driving differentiation towards the stalk phenotype [222].
During sprouting angiogenesis, ECs undergo a collective migration program, responsible for the hierarchical vessel organization. In this context, the role of non-canonical Wnt pathway has been established. In particular, Wnt5a is activated by cell tension and stabilizes vinculin binding to α-catenin at adherens junctions, anchoring the cell membrane to the actin cytoskeleton [223]. Alternatively to sprouting, intussusceptive angiogenesis requires EC cytoplasmic protrusions within the lumen of a pre-existing small vessel [224]. At this stage, ECs begin to acquire their identity by expressing specific differentiation markers that drive cell specification. Differential expression of arterial, venous and lymphatic markers is flow-regulated, as shown in chick embryo [225]. However, together with ECs, also mural cells that are progenitors of both vascular smooth muscle cells (VSMCs) and pericytes, contribute to vessel specification as either large or small vessels. Interaction between endothelial and mural cells requires ECM proteins and soluble ligands, as fibroblast growth factor (FGF) and TGF-β, able to bind EC membrane receptors [226,227]. In this context, hemodynamic forces contribute to the endothelial phenotype by further recruiting VSMCs to cover vessels, according to their fate. Adhesion of VSMCs to ECs is encouraged by membrane receptors, including Notch members and chemotactic molecules as Semaphorin 3, released by the endothelium in response to hemodynamic cues. These data were obtained by Padget et al. in Myl7−/− mutant E10.5 yolk sacs, which exhibited reduced VSMC covering the arteries, when compared to wild-type embryos [228].
Finally, vascular remodeling requires both vessel pruning and regression. At this stage, ECs fully acquire their arterial or venous identity, organizing a hierarchical network. Pruning is driven by blood flow velocity, is characterized by EC migration and contributes to vascular hierarchy by controlling vessel caliber [229]. In this context, the role of the TGF-β co-receptor endoglin (ENG) was shown. Specifically, under blood flow, artery diameter is enlarged in eng zebrafish mutants and it depends on increased EC size. This suggests that endoglin acts as mechanosensor, regulating EC shape and volume in response to blood flow [230]. Regression, on the other hand, is based on programmed cell death and EC number reduction, resulting in vessel bed retraction [231].

4.2. Biomechanical Control of Angiogenesis

During angiogenesis, blood vessel development is driven by VEGF family members that bind to their receptors and, in particular VEGFR2, triggering its dimerization and phosphorylation [232]. However, on their surface, ECs also express mechanosensors that can be distinguished as apical or junctional. Apical mechanosensors include ion channels, GPCRs, the primary cilium, the glycocalyx and caveolae [233]. At cell junction, mechanotransduction relies on integrins, PECAM1, vascular endothelial cadherin (VEC) and VEGFR2 [234].
However, PECAM1/VEC/VEGFR2 and caveolae form a multimeric complex capable of responding to shear stress [235], resulting in activation of NADPH oxidase 2 (NOX2) and endothelial nitric oxide synthase (eNOS) [236,237,238,239]. In this context, VEGFR2 phosphorylation is enhanced by mechanical cues including interstitial flow, matrix stiffness, cell curvature and shear stress [240]. When activated, it dissociates from caveolin-1 and becomes phosphorylated [241]. Depending on the phosphorylation site, different downstream cascades involving mechanosensitive proteins can be activated. Among these, the MEK/ERK pathway activates the mechanosensitive transcription factors YAP/TAZ, resulting in a decreased EC proliferation rate, both in-vitro and in zebrafish [242]; the Rho/ROCK cascade leads to focal adhesion kinase activation, actin polymerization and stress fiber formation; the PI3K/Akt and the p38/MAPK pathways [243]. In particular, YAP/TAZ nuclear translocation is enhanced by stiffness and disturbed blood flow, and is mediated by the Dll4-NOTCH1 interaction [244]. NOTCH1 as mechanosensor was recently described [245,246], and it acts by downstream activating SMAD6, which helps maintain EC barrier integrity, under shear stress condition [247]. Moreover, the focal adhesion protein DLC1 was described as a transcriptional target of YAP/TAZ in ECs, and it was recently shown to reduce YAP nuclear localization during sprouting angiogenesis in human umbilical vein endothelial cells (HUVECs), in response to cytoskeletal tension [248].
Under laminar flow conditions, mechanosensing signals originate from ECM proteins and are transmitted to integrins. Downstream of integrin activation, the FAK-ERK1/2, PI3K-AKT-eNOS and Rho/ROCK cascades are activated promoting cell survival, proliferation and migration [249]. However, the key EC mechanosensitive transcription factors activated by ECM stimuli are YAP and TAZ. YAP activation and its nuclear translocation result in a hyperproliferative and pro-inflammatory EC phenotype, commonly observed in vessel pathological conditions, as atherosclerosis [250]. Proliferation depends on ERK1/2 phosphorylation rate that increases in ECs growing in stiffer substrates [251].
Together with ECM-derived cues, several hemodynamic forces act on ECs driving vessel development and remodeling, both in embryo and in adulthood. Their magnitude and duration differ according to developmental stage, body region and physio-pathological condition. Shear stress is the most studied. It is parallel to the endothelium and depends on vessel caliber, blood density and velocity. In response to shear stress, ECs rearrange cytoskeleton and enhance junction stability, by encouraging PECAM1-actin microfilament interaction. Together, these morphological changes allow EC polarization according to flow direction, and this phenomenon is predominant in arteries [252]. In addition, the pivotal role of vinculin localization in junctional fingers during lumen expansion was reported [253]. Upon extracellular mechanical cues, EC cytoskeleton reorganization is driven by interaction between VEC–catenin complex and actin to stabilize AJs and to maintain barrier properties. Vinculin knock-out zebrafish exhibited a more permeable blood barrier, although this effect was limited to small molecules [254].
Blood pressure, instead, influences the vasculature by generating circumferential and axial stresses, that act tangentially and longitudinally on the internal lumen, respectively [255]. Intraluminal cues further result in VSMC stretch and vessel diameter regulation. In particular, VSMCs respond to circumferential stress induced by high blood pressure by adopting a contractile phenotype, that results in vessel caliber reduction. Likewise, under lower blood pressure, VSMC response results in vessel dilatation. This behavior is known as vascular myogenic response, and the main players acting in this mechanotransduction include integrins, cadherins and GPCRs [256]. Recently, latrophilin-2 was described as a novel GPCR activator in ECs. It is activated by flow and triggers pro-angiogenic signals through PECAM1 [257].
Beyond hemodynamic forces, also hydrostatic pressure acts on ECs and enhances tube formation by activating the Ras/ERK pathway, in a VEGFR2-indepenent manner. In detail, hydrostatic pressure does not directly activate the GPCRs/PKC cascade in HUVECs. Rather, its activation occurs downstream of aquaporin-1 mediated water efflux, following cell membrane exposure to the cue [258]. Mechanical cues further modify EC membrane physical and chemical parameters including membrane fluidity, thickness and the lipid order, which refers to the lipid phase transition from a liquid-disordered state to a liquid-ordered state [259,260]. About membrane chemical composition, cholesterol quantity was shown to decrease in response to laminar shear stress [261,262].
As described, membrane proteins act as mechanosensors inducing vascular remodeling, upon activation by ECM components. Cyclic stretch was shown to enhance mural cells to release thrombospondin-1 that binds integrin αvβ1, on EC surface. Downstream, this results in actin stress fiber polymerization and in YAP nuclear translocation [263]. In a recent study Seetharaman and colleagues identified novel mechanosensitive proteins in ECs, responsive to flow, comprising the FHL2 transcription factor, known to be involved in atherosclerosis progression [264]. FHL2 is early activated when flow changes and is capable of binding actin stress fibers to modulate AJ force-adaptation [265]. Other focal adhesion components involved in mechanosensation include the Lin-11, lsl-1 and Mec-3 (LIM) domain-containing proteins, regulating cell motility, apoptosis and proliferation in response to hemodynamic cues. In particular, zyxin is enable to shuttle from focal adhesion to the nucleus, where binds transcription factors and regulates gene expression, promoting VSMC contractility, in response to stretch [266].

4.3. Mechanosensitive Ion Channels in Vascular Development

Nowadays, the role of Ca2+ homeostasis in the behavior of non-contractile cells is plentifully studied. Mechanosensitive channels expressed on the EC membrane contribute to intracellular Ca2+ balance. Downstream, the regulation of Ca2+ release from the endoplasmic reticulum is crucial for cell junction maintaining and vessel permeability, under shear stress condition or increased blood flow [267]. In this context, among the mechanosensitive receptors previously described, several TRP family members and PIEZO1 mainly response to stretching and membrane tension triggering pro-angiogenic responses. Their expression differs throughout the vasculature, according to species, anatomical region and vessel caliber [268].
In mammals, TRPC and TRPV channels consistently contribute to vascular function. As described, during sprouting angiogenesis, mechanical cues cooperate with VEGF to guide ETC pathfinding. In response to VEGF, TRPC1 induces filipodia formation and EC polarization, despite this receptor does not appear to be essential for tube formation [269,270]. Under oxidative stress, TRPC4 was shown to enhance VEGF release in coronary artery ECs [271], as well as it induces retinal neovascularization under hypoxic condition [272]. Together with VEGF, erythropoietin and thrombin can also activate EC mechanosensors and, in particular, the TRPC3, resulting in increased cell proliferation and migration rate [273]. Further, in ECs, zyxin nuclear translocation occurs downstream of TRPC3 activation and contributes to endothelial phenotype maintaining, under increased blood pressure conditions [266]. In addition, TRPC1 and TRPC4 are involved in SOCE; their activation triggers the phospholipase C signal with consequent Ca2+ release from the endoplasmic reticulum [74]. Finally, TRPC6 and TRPC7 control the receptor-operated calcium entry (ROCE) within the cell [274].
About the TRPV members, the TRPV1 and TRPV4 are the main angiogenic regulators further acting on VSMC function [275]. TRPV1 induces vascular relaxation by mediating both eNOS activation and ECM remodeling, in response to shear stress [276]. Although TRPV1 was initially detected in neurons, where regulates synaptic transmission linked to nociception, it is also highly expressed in perivascular nerves and peripheral endothelium of visceral organs as lung. In these tissues TRPV1 responds to oxidative stress and ischemia, suggesting its role in pulmonary arterial hypertension pathogenesis [277,278].
In VSMCs, activated TRPV1 inhibits neointima formation and promotes EC proliferation and migration in a mitofusin 2-dependent manner, involving mitochondrial homeostasis [279]. Also TRPV4 dysfunction can impair mitochondrial homeostasis. In detail, TRPV4 over-activation disrupts the bioenergetic function of mitochondria in pulmonary arterial ECs due to eNOS uncoupling. Additionally, it increases migration and proliferation rates in microvascular ECs [280,281].
It was shown that TRPV1 forms heterodimers with TRPV4, promoting intracellular Ca2+ influx and sprouting angiogenesis in a VEGF-independent manner, in retinal microvascular ECs [282]. In retinal ECs, TRPV4 further contributes to pathological neovascularization due to reduced pericyte coverage, in response to oxidative damage. However, its depletion has not effect under physiological conditions [283].
TRPV4 is expressed early in endothelial progenitor cells [284]. Its down-expression was described in endothelium of several tumors, where ECs appear immature, permeable and covered by a reduced number of pericytes. However, the phenotype ameliorates when VEC expression increases, upon TRPV4 activation by mechanical cues, suggesting that TRPV4 contributes to vascular integrity in response to ECM stiffness [285]. In ECs, TRPV4 is also activated by shear stress and induces neovascularization by promoting VEGFR2 expression. Notably, VEGFR2 phosphorylation increases following TRPV4 silencing due to YAP/TAZ nuclear translocation. Increased VEGFR2 phosphorylation results in its shuttling from the perinuclear region to the plasma membrane, suggesting its gain of function. When phosphorylated, VEGFR2 promotes EC migration and collateral vessel development during arterialization [286,287]. Moreover, it was recently shown as TRPV4 contributes to lung microvessel disfunction in cystic fibrosis patients. In detail, the cystic fibrosis transmembrane conductance regulator (CFTR) protein controls endothelial properties by interacting with PIEZO1 on EC surface; its deficiency impairs PIEZO1 activity, under shear stress condition. As consequence, altered lipid metabolism leads to increased TRVP4 expression with calpain signal amplification and, then, endothelial barrier failure [288]. Regarding PIEZO1, it is expressed in various cell types, particularly in bone, cardiac, kidney and brain endothelium. It responds to shear stress, enhancing Ca2+ intracellular entry. This results in EC polarization and alignment to the flow direction, through protease activation [289]. During embryo development, PIEZO1 expression begins with blood circulation, becoming it abundant in mice ECs since E.9. Shear flow induces actin polymerization and stress fiber formation driving EC elongation and alignment. Piezo1 knock-down was shown to impair EC behavior due to failed mechanotransduction. Moreover, defects of the early vascular plexus were demonstrated in Piezo1 mutant mice, showing loss of large-diameter vessels [125].
In bone tissue, endothelial PIEZO1-mediated mechanotransduction enhances fracture repair by promoting vessel remodeling, to restore blood flow in damaged tissue. Conditional endothelial Piezo1 loss of function, indeed, results in reduced intracellular Ca2+ concentration and failed calpain activation, as well as in reduced PI3K/AKT phosphorylation and NOTCH1 and PECAM1 down-expression. These findings suggest that Piezo1 depletion in ECs can impair angiogenesis during bone repair, in open femoral shaft transverse fracture mice model [290].
Recently, Abello and colleagues described how blood flow regulates the expression of the mechanosensitive transcription factor klf2a in zebrafish, during vascular development. It drives arterial or venous differentiation, in response to pulsatile or laminar hemodynamic cues, respectively, by directing mural cell migration. More in detail, klf2a expression increases or decreases following piezo1 block or activation respectively, suggesting as piezo1 gating in response to shear stress contributes to the cross-talk between endothelial and mural cells [291]. EC behavior control by PIEZO1 depends on different shear stress and stiffness conditions. In detail, on a stiff substrate and under high shear stress, PIEZO1 knock-down results in disoriented stress fiber polymerization and reduced cell and nucleus size. In contrast, on soft matrix, ECs appear biggest and with increased nucleus area, upon PIEZO1 loss of function. Therefore, PIEZO1 drives stress fiber formation and controls cell shape by regulating cytoskeletal organization, according to flow dynamics and ECM rigidity [292]. PIEZO1 pro-angiogenetic function was further confirmed by Kang et al. The authors demonstrated that, following shear stress, PIEZO1-mediated Ca2+ influx modulates matrix metallopeptidase 2 (MMP2) activation to enhance membrane type 1-matrix metalloproteinase (MT1-MMP) translocation from the endoplasmic reticulum to the plasma membrane, to remodel ECM and to promote sprouting angiogenesis [293].
Finally, PIEZO1 expression in mitochondria of ECs was recently shown. Here, its gating enhances glycolysis, oxidative metabolism and ATP synthesis, explaining how the endothelium responds to energy demand, under shear stress conditions [294].

4.4. Mechanotransduction in Lymphangiogenesis

Together with blood vasculature, also lymphatic vessels development is influenced by physical forces. The hierarchical organization of lymphatic tree starts at the tissue interstitium with small caliber vessels, called capillary lymphatics. These consist of a lymphatic EC monolayer lacking of mural cells, continue to collectors and, then, to the thoracic duct. During development, embryonic fluid drainage is sufficient to trigger lymphatic vessel sprouting in-vitro [295]. Exceptionally, under shear flow, lymphatic ECs show reduced Notch1 activity and enhanced sprouting. This is mediated by Orai1, a calcium channel involved in SOCE. Orai1 activity was recently shown to depend on Piezo1, that acts as primary flow mechanosensor [296]. In zebrafish, an increased intracellular Ca2+ results in calmodulin activation and its binding with prox1, key regulator of lymphangiogenesis [297]. By binding Klf2, the Prox1/calmodulin complex forms a trimeric structure that suppresses Notch1 expression, by upregulating Dtx1 and Dtx3l [298]. Likewise, Klf2 and Klf4 expression is induced by Orai1; downstream, they activate expression of Vegfa, Vegfc, Fgfr3 and Cdkn1c, involved in lymphatic ECs proliferation [299].
Recently, Qin and colleagues demonstrated that medium stiff matrices, supplied with VEGF-C, are sufficient to promote sprouting lymphangiogenesis by enhancing the integrin/FAK mechanotransduction pathway, in cultured lymphatic ECs, suggesting new perspectives for lymphatic vessel regeneration [300].

5. Mechanosensing of the Blood-Brain Barrier

Unlike in other anatomical regions, the endothelium of the CNS is characterized by high selectivity, which is ensured by specialized cell junctions and metabolite transport strategies. Across the blood–brain barrier (BBB), molecules can move via three different mechanisms. Two of these require transporters, the ATP-binding cassette transporter (ABC) superfamily and the solute carrier (SLC) superfamily. The third mechanism is based on endocytosis and includes receptor-mediated transcytosis (RMT) and absorptive-mediated transcytosis (AMT). As the name suggests, the ABC transporters rely on ATP hydrolysis to move molecules from the ECs to the blood; the family includes about 50 members, divided into 7 families (ABCA to ABCG). The SLC channels facilitate passive transport of small molecules as ions, organic molecules, metabolites, neurotransmitters and xenobiotics. This superfamily include approximately 500 different transporters organized into 66 families (https://slc.bioparadigms.org/). They can be more or less selective and can mediate either influx or efflux of substances [301]. Finally, RMT requires a highly selective interaction between membrane receptors and macromolecules to be internalized; in contrast, AMT is responsible for cationic substrate absorption onto the caveolae surface. By these types of transcytosis, ECs transport peptides, antibodies, low-density lipoproteins and albumin [302]. These highly selective transcytosis mechanisms can occur from the brain to the bloodstream and vice versa [303]. However, despite these strategies, ECs alone are not sufficient to guarantee this barrier function; rather also pericytes, neurons, astrocytes and ECM contribute to vascular impermeability. For examples, pericytes regulate vascular leakage; likewise astrocytes and neurons release vasoactive molecules to modulate blood flow. For this reason, the term “blood–brain barrier” was coined by Paul Ehrlich, Edwin Goldmann and Lena Stern about a century ago. However, BBB properties are not uniform across different CSN areas, as they vary for capillary density, junction distribution, astrocyte and pericyte number [304]. These differences result from differential gene expression in specific brain areas [305]. Certainly, ECs are the most abundant cells of the BBB, anchored by both TJs and AJs, directly involved in selective permeability. Biogenesis of TJs during development was shown to be driven by VEC. In detail, AJ assembly occurs early during vascular development, as blood flow promotes VEC-mediated cell adhesion. Within ECs, this results in β-catenin stabilization at cell junctions, Akt-mediated FoxO1 phosphorylation and enhanced expression of claudin-5, a key component of TJs [306]. In addition, endothelial β-catenin disruption downregulates claudin-1 and claudin-3 expression in brain ECs, impairing TJ assembly [307]. By β-catenin, claudin-5 regulation is context-dependent, according to differential Wnt family member expression in brain regions. In particular, astrocytes are the primary source of extracellular secretion of Wnt ligands and the cytotype largely involved in generation of extracellular cues [308].
Endothelial cells also join pericytes through adhesion molecules, including N-cadherin and connexin-43, and by TGFβ−1. Therefore, to better describe the close relation underlying reciprocal regulation during neurovascular development, the concept of “neurovascular unit” (NVU) was introduced during the first Stroke Progress Review Group meeting of the National Institute of Neurological Disorders and Stroke (NINDS) of the National Institutes of Health (NIH) in 2001 (https://www.ninds.nih.gov/About-NINDS/Strategic-Plans-Evaluations/Strategic-Plans/Stroke-Progress-Review-Group, accessed on 24 March 2025). This morpho-functional integration leads neurons and vasculature to reciprocally regulate each other and, in particular, to regulate local perfusion in response to neural activity, in a process known as neurovascular coupling. Neurovascular coupling starts with an initiation phase, triggered by hypoxia conditions, chemical mediators as NO, or hemodynamic cues [309,310]. This early signal is dosed during a modulation phase, before being transmitted to the astrocytes and, then, to pericytes and ECs surrounding arterioles and capillaries (neurovascular transmission phase). In astrocytes, depolarization is promoted by the BK channels [311]. At EC monolayer, neuronal signal is elaborated as a vasomotor response and transmitted to mural cells, responsible for vascular tone and flow regulation (retrograde propagation/implementation phases) [312,313]. Neurovascular coupling is driven by hemodynamic forces also in astrocytes. They own mechanosensitive properties mediated by the TRPV4 receptor. Following a decrease in cerebral perfusion pressure, TRPV4/Cx43 coupling results in intracellular Ca2+ mobilization, aimed to increase systemic arterial blood pressure and heart rate [314,315,316] (Figure 7).
During neurovascular coupling, ECs respond to hemodynamic cues through the primary cilium. It is a sensory organelle exposed on the apical surface of the ECs and directly linked to the cytoskeletal microtubules, where modulate EC shape, in response to blood flow [317]. During zebrafish development, cilium morphogenesis in brain ECs begins during early vasculogenesis, preceding both flow induction and cardiac contractions, and continues until late-capillaries formation, contributing to vascular integrity. Cilium disruption following shear stress results in increased vessel permeability and intracerebral hemorrhage (ICH) [318]. Moreover, zebrafish mutants for the intraflagellar transport genes exhibit defective cilia and spontaneous ICH, most likely due to impaired sonic hedgehog (shh) signaling [319]. Likewise, Pkd1 mutant mice showed vascular leakage, hemorrhage and failed embryo development [320]. Moreover, flow was shown to activate Notch signal and, downstream, foxc1b in endothelial primary cilia, driving mural cell recruitment to arteries during vessel specification in zebrafish [321]. Together with the primary cilium, PIEZO1 and several TRPV family members drive BBB development and function. PIEZO1 expression is greater in ECs compared to other NVU cell types [322]. Its activation by blood flow during brain vasculature development triggers NOTCH signaling in ECs, enhancing EC/pericyte cross-talk and promoting pericyte proliferation and vessel coverage to ensure BBB properties [323]. By electrophysiological assays, it was shown that ETC branching depends on piezo1-mediated Ca2+ flux and piezo1 loss of function causes branch retraction in zebrafish larvae [324]. Calpain triggers ETC branch retraction downstream Ca2+ transient perturbation. In contrast, NOS regulates ETC branch extension [325]. In the same study, Liu and colleagues demonstrated that piezo1 mutant zebrafish larvae show enlarged brain vessels and increased number of vascular segments, suggesting the pivotal role of this mechanosensitive channel in early angiogenesis. In adulthood, ECs of brain arteries, veins and capillaries express PIEZO1, that remains responsive to blood flow and pressure. In addition, it induces NO-mediated vasodilatation in small diameter vessels, upon activation by blood cells. A similar response is observed in ECs, following neuronal depolarization [326]. In the retinal vasculature, Piezo1 was shown to colocalize with Pecam1 at AJs. Under shear stress conditions, Piezo1 over-activation disrupts Pecam1 structural organization and drives Cdh5 recruitment at AJs to maintain BBB integrity [122]. beyond its role in blood vasculature, Piezo1 further drives development of meningeal lymphatic vessels, where it regulates cerebrospinal fluid drainage. Piezo1 loss of function results in slower drainage and ventricular fluid accumulation [327]. In the CNS, also neurons express PIEZO1. Here, it contributes to neuroinflammation after ICH. In particular, Piezo1 expression increases following ICH, when promotes IL-6 production and NLRP3-mediated inflammasome activation [328]. Conversely, Piezo1 block decreases Bcl2-mediated neuronal apoptosis and reduces brain edema by down-regulating AQP4 expression, suggesting as PIEZO1 inhibition can contribute to neuronal tissue rescue, in an ICH mouse model [329].
Among the TRP family, TRPM4, TRPC3, TRPC6 and TRPA1 are involved in myogenic constriction and arteriolar contractility, regulating cerebral blood flow in response to pressure [330,331,332]. The TRPV members mainly regulate neurovascular coupling. TRPV2 is the most well characterized TRP channel in brain ECs, where it controls proliferation, migration and tubulogenesis [333]. Recently, Ramos et al. demonstrated heterozygous TrpV2 mutant rats exhibit enlarged retinal vessels, covered by thinner endothelium and increased inflammatory and oxidative marker expression, suggesting that this MC may play a protective role in diabetic retinopathy [334]. TRPV4 is expressed in all neurovascular cell types. In arteriolar and capillary ECs, it controls vasodilatation. In detail, in arterioles TRPV4-induced relaxation requires the activation of the endothelial muscarinic acetylcholine receptor and, for this reason, its role in vascular dementia onset is strongly hypothesized [335,336]. Moreover, when activated after ischemic stroke, TRPV4 enhances brain EC proliferation, angiogenesis and neurogenesis, thereby restoring brain functional properties [337,338]. In contrast, microglia-derived TNF-α induces increased TRPV4 expression in multiple sclerosis. In this context, TRPV4 contributes to BBB damage by increasing proinflammatory cytokine production and by down-regulating Cldn5, Cdh5 and Tjp1 expression [339]. Finally, in brain vasculature, TRPV1 controls vessel permeability and its expression increases after ischemic damage, contributing to brain edema, EC apoptosis and neuroinflammation [340,341].

6. Hemodynamics in Pathological Vascular Phenotypes

Perturbed hemodynamics can result in vasculature defects. For instance, the role of ENG and ALK1 as mechanosensors and regulators of vessel diameter was previously described [230]. They are BMP receptors and germline loss of function mutations in their coding genes cause the autosomal dominant hereditary hemorrhagic telangiectasia (HHT) phenotype, also known as Osler-Weber-Rendu syndrome [342]. Patients with HHT show focal vascular lesions featured by direct arteriovenous shunt, due to the lack of the capillary bed, and so called arteriovenous malformations (AVM). Sporadic AVM can also occur due to impaired expression of endothelial differentiation markers [343,344,345]. ALK1 controls arteriovenous specification and EC polarization against blood flow direction, by mediating integrin signaling with VEGFR2. Downstream, YAP/TAZ nuclear translocation controls cell migration [346]. Evidence of disturbed shear stress in HHT development was proposed by Baeyens et al., based on studies performed on mouse retina. In detail, they showed that BMP expression increased under shear stress condition and this condition enhanced Alk1-Eng coupling. Downstream, this response resulted in EC proliferation arrest and mural cell recruitment, suggesting that this mechanism contributes to EC specification and vascular stability maintaining. Therefore, perturbation of this signal may contribute to the HHT phenotype [347]. In addition, it was recently shown that shear stress induces BMP9/Alk1 interaction and, downstream, the activation of SMAD1/5 cascade, both in-vitro and in-vivo. Perturbation of this cascade results in the AVM phenotype. More in detail, SMAD1/5 signal activates the transcription of the GJA4 gene, encoding for the connexin 37 (Cx37). Cx37 was shown to negatively control EC migration in response to blood flow, suggesting its role in shunt regulation [348].
Together with HHT, also pulmonary arterial hypertension (PAH) arises following perturbation of TGF-β signal and, in particular, due to BMPR2 gene mutation, encoding for the BMP type 2 receptor. In PAH, BMPR2 loss of function mutations result in permeable lung vessels and increased pulmonary artery pressure, due to amplification of TGFβ-SMAD signaling [349]. However, mechanobiological factors, comprising EC contractile phenotype, ECM remodeling and stiffness, also contribute to the phenotype. In this field, formation of mixed canonical TGFβ-SMAD2/3 and lateral TGFβ-SMAD1/5 complexes was shown in-vitro, upon BMPR2 loss of function. As a consequence, ECs undergo EndMT, down-expressing endothelial specific markers as PECAM1 and CDH5. In contrast, increased expression of β1-integrin, integrin-linked kinase and fibrillin-1 was reported, suggesting BMPR2 as antagonist of the TGFβ-SMAD signaling, in response to ECM and AJ stiffness [350].
Again, TGF-β1 activation in stiffer matrices results in valve stiffening, a condition in which valvular ECs undergo EndMT, leading to aortic valve stenosis. However, in this case, downstream signal involves the Wnt/β-catenin pathway [351].

7. Discussion

Discovery of cell ability to be responsive to mechanical cues allowed the recent development of a novel research field known as mechanobiology. Broadly, mechanobiology encompasses all mechanisms and proteins involved in mechanotransduction, defined as the process by which a cell converts mechanical forces into biological responses. External mechanical stimuli that drive cell fate include, but are not limited to, ECM stiffness, rigidity, density, hemodynamic forces, flow direction, hydrostatic pressure [4,147,220]. Likewise, adherens junctions ensure the transmission of cytoskeletal mechanics between adjacent cells and, in this field, tension and compression are the most common forces [8].
Mechanosensitive proteins include focal adhesion proteins and mechanosensitive ion channels. The large number of mechanotransducers are heterogeneously expressed in different tissues, making cells responsive to specific mechanical stimuli. For this reason, the mechanical control of cell fate has been shown to be crucial during both development and remodeling phenomena. During development, ECM forces control mesoderm specification, gastrulation and morphogenetic movements by activating integrins and cadherins [166]. Organogenesis, in turn, further requires the activation of mechanosensitive channels, resulting in their gating and ion influx within the cell. In this context, the TRP and PIEZO families are the most well characterized. Notably, the role of polycystins in primary cilium and kidney development has been largely studied, showing as PKD1 and PKD2 mutations result in the autosomal dominant polycystic kidney disease [104,105,106]. Likewise, PIEZO1 involvement in muscle and bone development was confirmed. More in detail, in bone tissue PIEZO1 activation by blood flow induces tissue repair after fracture [202].
However, mechanical control of cell function is even more evident during vascular development, when hemodynamic cues drive EC differentiation, polarization and migration. In this context, receptors for soluble growth factors also act as mechanosensors [217,230]. The endothelial cell response to blood flow further involves the VEGF signaling and promotes cell adhesion. In particular, this phenomenon is crucial during BBB development, when expression of TJ proteins is promoted by VEC-mediated cell adhesion, when blood starts to flow [306,307,308].
The importance of mechanosensation is proven by pathological phenotypes arising due to mutations in genes encoding mechanoreceptors. Among these, failed mechanotransduction was linked to fibrosis and congenital heart defects [352,353,354]. For this reason, targeting mechanosensors is a novel and promising therapeutic strategy in the field of regenerative medicine.

8. Conclusions

Cell responses to external mechanical cues drive intracellular biological processes aimed to control tissue development, remodeling and physio-pathological conditions. In the last few years, attention to cell mechanotransduction has been rapidly increasing, due to the large number of biological processes regulated by mechanical stimuli. As discussed, proteins acting as mechanosensor include focal adhesion components as integrins, vinculin and cytoskeleton elements, responsive to ECM mechanical properties. The discovery of epigenetic regulation as a final event of mechanotransduction is increasing perspectives on the possibility of regulating gene expression by modulating ECM composition, stiffness and rigidity. This strategy is currently considered very promising for tissue regeneration. On the other hand, the identification of ion channels that gain gating activity upon mechanical stimulation has clarified several aspects of development and remodeling in various tissues, including bone, teeth, muscle and vessels. In the field of vascular biology, mechanotransduction drives endothelial differentiation and vessel morphogenesis since the early stages of development, as mechanosensor expression precedes blood flow. In particular, PIEZO1 and TRPV4 mechanosensitive ion channels control endothelial progenitor cell differentiation and capillary plexus formation. Interestingly, defects in mechanotransduction result in aberrant angiogenetic sprouting due to impaired endothelial tip/stalk phenotype acquisition. In addition, blood flow and shears stress promote cell adhesion and pericyte recruitment during BBB development, and aberrant mechanotransduction has been confirmed to result in enlarged vessels and aneurysm. Recent studies aim to develop modulable mechanofluidic devices to control fluid dynamics in-vitro, for endothelial and myocardial regeneration after injury.

Author Contributions

Conceptualization, S.A.; methodology, C.R.; resources, S.L.V. and A.C.; data curation, L.D.; writing—original draft preparation, C.S.; writing—review and editing, A.S.; supervision, R.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AJAdherens Junction
AVMArterioVenous Malformation
BBBBlood-Brain Barrier
BKBig Potassium
BMPBone Morphogenetic Protein
CNSCentral Nervous System
ECEndothelial Cell
ECMExtracellular Matrix
ENaCEpithelial Sodium Channel
EndMTEndothelial-to-Mesenchymal Transition
ENGEndoglin
ETCEndothelial Tip Cell
FAKFocal Adhesion Kinase
GPCRG-Protein Coupled Receptor
HHTHereditary Hemorrhagic Telangiectasia
ICHIntracerebral Haemorrhage
KLFKrüppel-Like Factor
LINCLinker of Nucleoskeleton and Cytoskeleton
LTPLong Term Potentiation
MCMechanosensitive Channel
MSCMesenchymal Stem Cell
NONitric Oxide
NVUNeuroVascular Unit
SOCEStore Operated Ca2+ Entry
TAZTranscriptional coactivator with PDZ-binding motif
TJTight junction
TREKTWIK-Related K+
TRPTransient Receptor Potential
VECVascular Endothelial Cadherin
VEGFVascular Endothelial Growth Factor
VSMCVascular Smooth Muscle Cell
YAPYes-Associated Protein

References

  1. Cai, J.; Deng, Y.; Min, Z.; Li, C.; Zhao, Z.; Jing, D. Deciphering the dynamics: Exploring the impact of mechanical forces on histone acetylation. FASEB J. 2024, 38, e23849. [Google Scholar] [CrossRef] [PubMed]
  2. Qin, R.; Schmid, H.; Münzberg, C.; Maass, U.; Krndija, D.; Adler, G.; Seufferlein, T.; Liedert, A.; Ignatius, A.; Oswald, F.; et al. Phosphorylation and turnover of paxillin in focal contacts is controlled by force and defines the dynamic state of the adhesion site. Cytoskeleton 2015, 72, 101–112. [Google Scholar] [CrossRef] [PubMed]
  3. Panagaki, F.; Tapia-Rojo, R.; Zhu, T.; Milmoe, N.; Paracuellos, P.; Board, S.; Mora, M.; Walker, J.; Rostkova, E.; Stannard, A.; et al. Structural anisotropy results in mechano-directional transport of proteins across nuclear pores. Nat. Phys. 2024, 20, 1180–1193. [Google Scholar] [CrossRef] [PubMed]
  4. Bajpai, S.K.; Bae, Y.; Balakrishnan, S.; Kim, D.H. Editorial: Mechanical coupling between extracellular and intracellular microenvironment. Front. Cell Dev. Biol. 2024, 12, 1427439. [Google Scholar] [CrossRef]
  5. Do, T.D.; Katsuyoshi, J.; Cai, H.; Ohashi, T. Mechanical Properties of Isolated Primary Cilia Measured by Micro-tensile Test and Atomic Force Microscopy. Front. Bioeng. Biotechnol. 2021, 9, 753805. [Google Scholar] [CrossRef]
  6. Nishizawa, K.; Lin, S.Z.; Chardès, C.; Rupprecht, J.F.; Lenne, P.F. Two-point optical manipulation reveals mechanosensitive remodeling of cell-cell contacts in vivo. Proc. Natl. Acad. Sci. USA 2023, 120, e2212389120. [Google Scholar] [CrossRef]
  7. McCormick, L.E.; Gupton, S.L. Mechanistic advances in axon pathfinding. Curr. Opin. Cell Biol. 2020, 63, 11–19. [Google Scholar] [CrossRef]
  8. Morales-Camilo, N.; Liu, J.; Ramírez, M.J.; Canales-Salgado, P.; Alegría, J.J.; Liu, X.; Ong, H.T.; Barrera, N.P.; Fierro, A.; Toyama, Y.; et al. Alternative molecular mechanisms for force transmission at adherens junctions via β-catenin-vinculin interaction. Nat. Commun. 2024, 15, 5608. [Google Scholar] [CrossRef]
  9. Chen, K.; Kwon, S.H.; Henn, D.; Kuehlmann, B.A.; Tevlin, R.; Bonham, C.A.; Griffin, M.; Trotsyuk, A.A.; Borrelli, M.R.; Noishiki, C.; et al. Disrupting biological sensors of force promotes tissue regeneration in large organisms. Nat. Commun. 2021, 12, 5256. [Google Scholar] [CrossRef]
  10. Liu, L.; Zheng, W.; Wei, Y.; Li, Q.; Chen, N.; Xia, Q.; Wang, L.; Hu, J.; Zhou, X.; Sun, Y.; et al. Mechanical stress-induced autophagy is cytoskeleton dependent. Cell Prolif. 2024, 57, e13728. [Google Scholar] [CrossRef]
  11. Bays, J.L.; Campbell, H.K.; Heidema, C.; Sebbagh, M.; DeMali, K.A. Linking E-cadherin mechanotransduction to cell metabolism through force-mediated activation of AMPK. Nat. Cell Biol. 2017, 19, 724–731. [Google Scholar] [CrossRef] [PubMed]
  12. Welhaven, H.D.; McCutchen, C.N.; June, R.K. Effects of mechanical stimulation on metabolomic profiles of SW1353 chondrocytes: Shear and compression. Biol. Open 2022, 11, bio058895. [Google Scholar] [CrossRef] [PubMed]
  13. Phang, J.M.; Liu, W.; Zabirnyk, O. Proline metabolism and microenvironmental stress. Annu. Rev. Nutr. 2010, 30, 441–463. [Google Scholar] [CrossRef]
  14. Arnadóttir, J.; Chalfie, M. Eukaryotic mechanosensitive channels. Annu. Rev. Biophys. 2010, 39, 111–137. [Google Scholar] [CrossRef] [PubMed]
  15. Hamill, O.P.; Martinac, B. Molecular basis of mechanotransduction in living cells. Physiol. Rev. 2001, 81, 685–740. [Google Scholar] [CrossRef]
  16. Anishkin, A.; Loukin, S.H.; Teng, J.; Kung, C. Feeling the hidden mechanical forces in lipid bilayer is an original sense. Proc. Natl. Acad. Sci. USA 2014, 111, 7898–7905. [Google Scholar] [CrossRef]
  17. Hu, X.; Margadant, F.M.; Yao, M.; Sheetz, M.P. Molecular stretching modulates mechanosensing pathways. Protein Sci. 2017, 26, 1337–1351. [Google Scholar] [CrossRef]
  18. Fu, Y.; Jing, Z.; Chen, T.; Xu, X.; Wang, X.; Ren, M.; Wu, Y.; Wu, T.; Li, Y.; Zhang, H.; et al. Nanotube patterning reduces macrophage inflammatory response via nuclear mechanotransduction. J. Nanobiotechnol. 2023, 21, 229. [Google Scholar] [CrossRef]
  19. Vivo, M.; Rosti, V.; Cervone, S.; Lanzuolo, C. Chromatin plasticity in mechanotransduction. Curr. Opin. Cell Biol. 2024, 88, 102376. [Google Scholar] [CrossRef]
  20. Grudtsyna, V.; Packirisamy, S.; Bidone, T.C.; Swaminathan, V. Extracellular matrix sensing via modulation of orientational order of integrins and F-actin in focal adhesions. Life Sci. Alliance 2023, 6, e202301898. [Google Scholar] [CrossRef]
  21. Benke, K.; Ágg, B.; Szilveszter, B.; Tarr, F.; Nagy, Z.B.; Pólos, M.; Daróczi, L.; Merkely, B.; Szabolcs, Z. The role of transforming growth factor-beta in Marfan syndrome. Cardiol. J. 2013, 20, 227–234. [Google Scholar] [CrossRef]
  22. Pei, F.; Guo, T.; Zhang, M.; Ma, L.; Jing, J.; Feng, J.; Ho, T.V.; Wen, Q.; Chai, Y. FGF signaling modulates mechanotransduction/WNT signaling in progenitors during tooth root development. Bone Res. 2024, 12, 37. [Google Scholar] [CrossRef]
  23. Steinberg, T.; Dieterle, M.P.; Ramminger, I.; Klein, C.; Brossette, J.; Husari, A.; Tomakidi, P. On the Value of In Vitro Cell Systems for Mechanobiology from the Perspective of Yes-Associated Protein/Transcriptional Co-Activator with a PDZ-Binding Motif and Focal Adhesion Kinase and Their Involvement in Wound Healing, Cancer, Aging, and Senescence. Int. J. Mol. Sci. 2023, 24, 12677. [Google Scholar] [CrossRef]
  24. Gautel, M. Cytoskeletal protein kinases: Titin and its relations in mechanosensing. Pflugers Arch. 2011, 462, 119–134. [Google Scholar] [CrossRef]
  25. Yao, M.; Goult, B.T.; Klapholz, B.; Hu, X.; Toseland, C.P.; Guo, Y.; Cong, P.; Sheetz, M.P.; Yan, J. The mechanical response of talin. Nat. Commun. 2016, 7, 11966. [Google Scholar] [CrossRef]
  26. Takata, T.; Matsumura, M. The LINC Complex Assists the Nuclear Import of Mechanosensitive Transcriptional Regulators. Results Probl. Cell Differ. 2022, 70, 315–337. [Google Scholar] [CrossRef]
  27. García-González, A.; Jacchetti, E.; Marotta, R.; Tunesi, M.; Rodríguez Matas, J.F.; Raimondi, M.T. The Effect of Cell Morphology on the Permeability of the Nuclear Envelope to Diffusive Factors. Front. Physiol. 2018, 9, 925. [Google Scholar] [CrossRef]
  28. Galbraith, C.G.; Yamada, K.M.; Sheetz, M.P. The relationship between force and focal complex development. J. Cell Biol. 2002, 159, 695–705. [Google Scholar] [CrossRef]
  29. Harrison, O.J.; Jin, X.; Hong, S.; Bahna, F.; Ahlsen, G.; Brasch, J.; Wu, Y.; Vendome, J.; Felsovalyi, K.; Hampton, C.M.; et al. The extracellular architecture of adherens junctions revealed by crystal structures of type I cadherins. Structure 2011, 19, 244–256. [Google Scholar] [CrossRef]
  30. Meili, R.; Alonso-Latorre, B.; del Alamo, J.C.; Firtel, R.A.; Lasheras, J.C. Myosin II is essential for the spatiotemporal organization of traction forces during cell motility. Mol. Biol. Cell 2010, 21, 405–417. [Google Scholar] [CrossRef]
  31. Jacquemet, G.; Stubb, A.; Saup, R.; Miihkinen, M.; Kremneva, E.; Hamidi, H.; Ivaska, J. Filopodome Mapping Identifies p130Cas as a Mechanosensitive Regulator of Filopodia Stability. Curr. Biol. 2019, 29, 202–216.e7. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, X.; Yang, Y.; Wang, Y.; Lu, C.; Hu, X.; Kawazoe, N.; Yang, Y.; Chen, G. Focal adhesion and actin orientation regulated by cellular geometry determine stem cell differentiation via mechanotransduction. Acta Biomater. 2024, 182, 81–92. [Google Scholar] [CrossRef] [PubMed]
  33. McGillivary, R.M.; Starr, D.A.; Luxton, G.W.G. Building and breaking mechanical bridges between the nucleus and cytoskeleton: Regulation of LINC complex assembly and disassembly. Curr. Opin. Cell Biol. 2023, 85, 102260. [Google Scholar] [CrossRef]
  34. Miralles, F.; Posern, G.; Zaromytidou, A.I.; Treisman, R. Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell 2003, 113, 329–342. [Google Scholar] [CrossRef]
  35. Maciejowski, J.; Hatch, E.M. Nuclear Membrane Rupture and Its Consequences. Annu. Rev. Cell Dev. Biol. 2020, 36, 85–114. [Google Scholar] [CrossRef]
  36. Ahmed, M.S.; Ikram, S.; Bibi, N.; Mir, A. Hutchinson-Gilford Progeria Syndrome: A Premature Aging Disease. Mol. Neurobiol. 2018, 55, 4417–4427. [Google Scholar] [CrossRef]
  37. Greenberg, C.R.; Rimoin, D.L.; Gruber, H.E.; DeSa, D.J.; Reed, M.; Lachman, R.S. A new autosomal recessive lethal chondrodystrophy with congenital hydrops. Am. J. Med. Genet. 1988, 29, 623–632. [Google Scholar] [CrossRef]
  38. Eriksson, M.; Brown, W.T.; Gordon, L.B.; Glynn, M.W.; Singer, J.; Scott, L.; Erdos, M.R.; Robbins, C.M.; Moses, T.Y.; Berglund, P.; et al. Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature 2003, 423, 293–298. [Google Scholar] [CrossRef]
  39. Li, L.; Du, Y.; Kong, X.; Li, Z.; Jia, Z.; Cui, J.; Gao, J.; Wang, G.; Xie, K. Lamin B1 is a novel therapeutic target of betulinic acid in pancreatic cancer. Clin. Cancer Res. 2013, 19, 4651–4661. [Google Scholar] [CrossRef]
  40. Moss, S.F.; Krivosheyev, V.; de Souza, A.; Chin, K.; Gaetz, H.P.; Chaudhary, N.; Worman, H.J.; Holt, P.R. Decreased and aberrant nuclear lamin expression in gastrointestinal tract neoplasms. Gut 1999, 45, 723–729. [Google Scholar] [CrossRef]
  41. Chuang, Y.C.; Chen, C.C. Force from Filaments: The Role of the Cytoskeleton and Extracellular Matrix in the Gating of Mechanosensitive Channels. Front. Cell Dev. Biol. 2022, 10, 886048. [Google Scholar] [CrossRef]
  42. Buncha, V.; Cherezova, A.; Alexander, S.; Baranovskaya, I.; Coleman, K.A.; Cherian-Shaw, M.; Brands, M.W.; Sullivan, J.C.; O’Connor, P.M.; Mamenko, M. Aldosterone Antagonism Is More Effective at Reducing Blood Pressure and Excessive Renal ENaC Activity in AngII-Infused Female Rats Than in Males. Hypertension 2023, 80, 2196–2208. [Google Scholar] [CrossRef]
  43. Nickerson, A.J.; Sheng, S.; Cox, N.A.; Szekely, K.G.; Marciszyn, A.L.; Lam, T.; Chen, J.; Gingras, S.; Kashlan, O.B.; Kirabo, A.; et al. Loss of the alpha subunit distal furin cleavage site blunts ENaC activation following Na+ restriction. J. Physiol. 2024, 602, 4309–4326. [Google Scholar] [CrossRef]
  44. Cheng, Y.R.; Jiang, B.Y.; Chen, C.C. Acid-sensing ion channels: Dual function proteins for chemo-sensing and mechano-sensing. J. Biomed. Sci. 2018, 25, 46. [Google Scholar] [CrossRef]
  45. Sorum, B.; Docter, T.; Panico, V.; Rietmeijer, R.A.; Brohawn, S.G. Tension activation of mechanosensitive two-pore domain K+ channels TRAAK, TREK-1, and TREK-2. Nat. Commun. 2024, 15, 3142. [Google Scholar] [CrossRef]
  46. Dupont, C.; Blake, B.; Voss, A.A.; Rich, M.M. BK channels promote action potential repolarization in skeletal muscle but contribute little to myotonia. Pflugers Arch. 2024, 476, 1693–1702. [Google Scholar] [CrossRef]
  47. Wang, Y.; Wang, M.; Ning, F.; Ren, D.; Tao, J.; Xie, W.; Eaton, D.C.; Jiang, G.; Farris, A.B.; Xin, H.; et al. A novel role of BK potassium channel activity in preventing the development of kidney fibrosis. Kidney Int. 2022, 101, 945–962. [Google Scholar] [CrossRef]
  48. Zhang, C.; Xiao, M.; Cao, N.; Zhang, L.; He, Q.; Wang, J.; Wang, R.; Wang, L.; Zhao, L.; Si, J. Emodin activates BK channel in vascular smooth muscle cells and relaxes the interlobar renal artery of rat. Biomed. Pharmacother. 2022, 153, 113452. [Google Scholar] [CrossRef]
  49. Contet, C.; Goulding, S.P.; Kuljis, D.A.; Barth, A.L. BK Channels in the Central Nervous System. Int. Rev. Neurobiol. 2016, 128, 281–342. [Google Scholar] [CrossRef]
  50. Echeverría, F.; Gonzalez-Sanabria, N.; Alvarado-Sanchez, R.; Fernández, M.; Castillo, K.; Latorre, R. Large conductance voltage-and calcium-activated K+ (BK) channel in health and disease. Front. Pharmacol. 2024, 15, 1373507. [Google Scholar] [CrossRef]
  51. Lin, H.H.; Ng, K.F.; Chen, T.C.; Tseng, W.Y. Ligands and Beyond: Mechanosensitive Adhesion GPCRs. Pharmaceuticals 2022, 15, 219. [Google Scholar] [CrossRef] [PubMed]
  52. Dupont, S. Role of YAP/TAZ in cell-matrix adhesion-mediated signalling and mechanotransduction. Exp. Cell Res. 2016, 343, 42–53. [Google Scholar] [CrossRef]
  53. Pang, V.Y.; Yang, Z.; Wu, S.M.; Pang, J.J. The co-expression of the depolarizing and hyperpolarizing mechanosensitive ion channels in mammalian retinal neurons. Front. Med. 2024, 11, 1463898. [Google Scholar] [CrossRef]
  54. Edosuyi, O.; Adesuyi, A.; Choi, M.; Igbe, I.; Oyekan, A. Malate reduced blood pressure and exerted differential effects on renal hemodynamics; role of the nitric oxide system and renal epithelial sodium channels (ENaC). Eur. J. Pharmacol. 2023, 938, 175441. [Google Scholar] [CrossRef]
  55. Sandoz, G.; Lesage, F. Protein complex analysis of native brain potassium channels by proteomics. Methods Mol. Biol. 2008, 491, 113–123. [Google Scholar] [CrossRef]
  56. Sandoz, G.; Tardy, M.P.; Thümmler, S.; Feliciangeli, S.; Lazdunski, M.; Lesage, F. Mtap2 is a constituent of the protein network that regulates twik-related K+ channel expression and trafficking. J. Neurosci. 2008, 28, 8545–8552. [Google Scholar] [CrossRef]
  57. Li, Z.; Yu, Y.; Cao, X.; Wang, Y.; Lu, J.; Feng, Y.; Jiang, Y.; Lu, Y. Mechanism of Ca2+ overload caused by STIM1/ORAI1 activation of store-operated Ca2+ entry (SOCE) in hydrogen peroxide-induced mitochondrial damage and apoptosis in human primary melanocytes. Mol. Biol. Rep. 2025, 52, 223. [Google Scholar] [CrossRef]
  58. Storch, U.; Mederos y Schnitzler, M.; Gudermann, T. G protein-mediated stretch reception. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H1241–H1249. [Google Scholar] [CrossRef]
  59. Gualdani, R.; Gailly, P. How TRPC Channels Modulate Hippocampal Function. Int. J. Mol. Sci. 2020, 21, 3915. [Google Scholar] [CrossRef]
  60. Rosenberg, P.; Hawkins, A.; Stiber, J.; Shelton, J.M.; Hutcheson, K.; Bassel-Duby, R.; Shin, D.M.; Yan, Z.; Williams, R.S. TRPC3 channels confer cellular memory of recent neuromuscular activity. Proc. Natl. Acad. Sci. USA 2004, 101, 9387–9392. [Google Scholar] [CrossRef]
  61. Higashida, H.; Yuhi, T.; Akther, S.; Amina, S.; Zhong, J.; Liang, M.; Nishimura, T.; Liu, H.X.; Lopatina, O. Oxytocin release via activation of TRPM2 and CD38 in the hypothalamus during hyperthermia in mice: Implication for autism spectrum disorder. Neurochem. Int. 2018, 119, 42–48. [Google Scholar] [CrossRef] [PubMed]
  62. Tong, X.; Tong, Y.; Zheng, J.; Shi, R.; Liang, H.; Li, M.; Meng, Y.; Shi, J.; Zhao, D.; Seehus, C.R.; et al. TRPM7 contributes to pyroptosis and its involvement in status epilepticus. J. Neuroinflammation 2024, 21, 315. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, Y.; Shi, Y.; Zuo, M.; Yu, Y.; Huang, X. Advances in the Study for Modulators of Transient Receptor Potential Vanilloid (TRPV) Channel Family. Curr. Top Med. Chem. 2025, 25, 1–48. [Google Scholar] [CrossRef]
  64. Wang, X.D.; Lin, Z.K.; Ji, S.X.; Bi, S.Y.; Liu, W.X.; Zhang, G.F.; Wan, F.H.; Lü, Z.C. Molecular Characterization of TRPA Subfamily Genes and Function in Temperature Preference in Tuta absoluta (Meyrick) (Lepidoptera: Gelechiidae). Int. J. Mol. Sci. 2021, 22, 7157. [Google Scholar] [CrossRef]
  65. Huang, P.; Dong, R.Y.; Wang, P.; Xu, M.; Sun, X.; Dong, X.P. MCOLN/TRPML channels in the regulation of MTORC1 and autophagy. Autophagy 2024, 20, 1203–1204. [Google Scholar] [CrossRef]
  66. Lakhia, R.; Ramalingam, H.; Chang, C.M.; Cobo-Stark, P.; Biggers, L.; Flaten, A.; Alvarez, J.; Valencia, T.; Wallace, D.P.; Lee, E.C.; et al. PKD1 and PKD2 mRNA cis-inhibition drives polycystic kidney disease progression. Nat. Commun. 2022, 13, 4765. [Google Scholar] [CrossRef]
  67. Zhang, Y.; Zou, W.; Dou, W.; Luo, H.; Ouyang, X. Pleiotropic physiological functions of Piezo1 in human body and its effect on malignant behavior of tumors. Front. Physiol. 2024, 15, 1377329. [Google Scholar] [CrossRef]
  68. Wang, Y.; Zhang, Y.; Leung, V.; Seradj, S.H.; Sonmez, U.; Servin-Vences, R.; Lipomi, D.; Ye, L.; Patapoutian, A. A key role of PIEZO2 mechanosensitive ion channel in adipose sensory innervation. bioRxiv 2024. [Google Scholar] [CrossRef]
  69. Cox, C.D.; Poole, K.; Martinac, B. Re-evaluating TRP channel mechanosensitivity. Trends Biochem. Sci. 2024, 49, 693–702. [Google Scholar] [CrossRef]
  70. Huang, J.; Korsunsky, A.; Yazdani, M.; Chen, J. Targeting TRP channels: Recent advances in structure, ligand binding, and molecular mechanisms. Front. Mol. Neurosci. 2024, 16, 1334370. [Google Scholar] [CrossRef]
  71. Wang, Y.; Bu, J.; Shen, H.; Li, H.; Wang, Z.; Chen, G. Targeting Transient Receptor Potential Canonical Channels for Diseases of the Nervous System. Curr. Drug Targets 2017, 18, 1460–1465. [Google Scholar] [CrossRef]
  72. Wes, P.D.; Chevesich, J.; Jeromin, A.; Rosenberg, C.; Stetten, G.; Montell, C. TRPC1, a human homolog of a Drosophila store-operated channel. Proc. Natl. Acad. Sci. USA 1995, 92, 9652–9656. [Google Scholar] [CrossRef] [PubMed]
  73. Obukhov, A.G.; Harteneck, C.; Zobel, A.; Harhammer, R.; Kalkbrenner, F.; Leopoldt, D.; Lückhoff, A.; Nürnberg, B.; Schultz, G. Direct activation of trpl cation channels by G alpha11 subunits. EMBO J. 1996, 15, 5833–5838. [Google Scholar] [CrossRef] [PubMed]
  74. Lopez, J.J.; Jardin, I.; Sanchez-Collado, J.; Salido, G.M.; Smani, T.; Rosado, J.A. TRPC Channels in the SOCE Scenario. Cells 2020, 9, 126. [Google Scholar] [CrossRef]
  75. Wang, M.; Wen, W.; Chen, Y.; Yishajiang, S.; Li, Y.; Li, Z.; Zhang, X. TRPC5 channel participates in myocardial injury in chronic intermittent hypoxia. Clinics 2024, 79, 100368. [Google Scholar] [CrossRef]
  76. Yang, L.; Peng, Z.; Gong, F.; Yan, W.; Shi, Y.; Li, H.; Zhou, C.; Yao, H.; Yuan, M.; Yu, F.; et al. TRPC4 aggravates hypoxic pulmonary hypertension by promoting pulmonary endothelial cell apoptosis. Free Radic. Biol. Med. 2024, 219, 141–152. [Google Scholar] [CrossRef]
  77. Zong, P.; Li, C.X.; Feng, J.; Cicchetti, M.; Yue, L. TRP Channels in Stroke. Neurosci. Bull. 2024, 40, 1141–1159. [Google Scholar] [CrossRef]
  78. Bröker-Lai, J.; Rego Terol, J.; Richter, C.; Mathar, I.; Wirth, A.; Kopf, S.; Moreno-Pérez, A.; Büttner, M.; Tan, L.L.; Makke, M.; et al. TRPC5 controls the adrenaline-mediated counter regulation of hypoglycemia. EMBO J. 2024, 43, 5813–5836. [Google Scholar] [CrossRef]
  79. Wang, S.; Li, X.; Hu, Y.; Wang, L.; Lv, G.; Feng, Y.; Sun, Z.; Cao, Z.; Liu, Y.; Wang, H. Discovery of N-alkyl-N-benzyl thiazoles as novel TRPC antagonists for the treatment of glioblastoma multiforme. Eur. J. Med. Chem. 2024, 265, 116066. [Google Scholar] [CrossRef]
  80. Qi, H.; Wu, F.; Wang, H. Function of TRPC1 in modulating hepatocellular carcinoma progression. Med. Oncol. 2023, 40, 97. [Google Scholar] [CrossRef]
  81. Becker, A.; Mannebach, S.; Mathar, I.; Weissgerber, P.; Freichel, M.; Loodin, A.P.; Fecher-Trost, C.; Belkacemi, A.; Beck, A.; Philipp, S.E. Control of Insulin Release by Transient Receptor Potential Melastatin 3 (TRPM3) Ion Channels. Cell Physiol. Biochem. 2020, 54, 1115–1131. [Google Scholar] [CrossRef] [PubMed]
  82. Becker, A.; Götz, C.; Montenarh, M.; Philipp, S.E. Control of TRPM3 Ion Channels by Protein Kinase CK2-Mediated Phosphorylation in Pancreatic β-Cells of the Line INS-1. Int. J. Mol. Sci. 2021, 22, 13133. [Google Scholar] [CrossRef] [PubMed]
  83. do Nascimento, T.H.O.; Pereira-Figueiredo, D.; Veroneze, L.; Nascimento, A.A.; De Logu, F.; Nassini, R.; Campello-Costa, P.; Faria-Melibeu, A.D.C.; Souza Monteiro de Araújo, D.; Calaza, K.C. Functions of TRPs in retinal tissue in physiological and pathological conditions. Front. Mol. Neurosci. 2024, 17, 1459083. [Google Scholar] [CrossRef]
  84. Liu, D.Q.; Mei, W.; Zhou, Y.Q.; Xi, H. Targeting TRPM channels for cerebral ischemia-reperfusion injury. Trends Pharmacol. Sci. 2024, 45, 862–867. [Google Scholar] [CrossRef]
  85. Liman, E.R. The Ca2+-Activated TRP Channels: TRPM4 and TRPM5. In TRP Ion Channel Function in Sensory Transduction and Cellular Signaling Cascades; Chapter 15; Liedtke, W.B., Heller, S., Eds.; CRC Press: Boca Raton, FL, USA, 2007. [Google Scholar]
  86. Holderby, K.G.; Kozak, J.A. Use of tetraethylammonium (TEA) and Tris loading for blocking TRPM7 channels in intact cells. Front. Pharmacol. 2024, 15, 1341799. [Google Scholar] [CrossRef]
  87. Huang, P.; Qu, C.; Rao, Z.; Wu, D.; Zhao, J. Bidirectional regulation mechanism of TRPM2 channel: Role in oxidative stress, inflammation and ischemia-reperfusion injury. Front. Immunol. 2024, 15, 1391355. [Google Scholar] [CrossRef]
  88. Landen, J.G.; Vandendoren, M.; Killmer, S.; Bedford, N.L.; Nelson, A.C. Huddling substates in mice facilitate dynamic changes in body temperature and are modulated by Shank3b and Trpm8 mutation. Commun. Biol. 2024, 7, 1186. [Google Scholar] [CrossRef]
  89. King, J.W.; Bennett, A.S.W.; Wood, H.M.; Baker, C.C.; Alsaadi, H.; Topley, M.; Vanner, S.A.; Reed, D.E.; Lomax, A.E. Expression and function of transient receptor potential melastatin 3 in the spinal afferent innervation of the mouse colon. Am. J. Physiol. Gastrointest. Liver Physiol. 2024, 326, G176–G186. [Google Scholar] [CrossRef]
  90. Sinica, V.; Vlachová, V. Transient receptor potential ankyrin 1 channel: An evolutionarily tuned thermosensor. Physiol. Res. 2021, 70, 363–381. [Google Scholar] [CrossRef]
  91. Stueber, T.; Eberhardt, M.J.; Caspi, Y.; Lev, S.; Binshtok, A.; Leffler, A. Differential cytotoxicity and intracellular calcium-signalling following activation of the calcium-permeable ion channels TRPV1 and TRPA1. Cell Calcium 2017, 68, 34–44. [Google Scholar] [CrossRef]
  92. Fila, M.; Przyslo, L.; Derwich, M.; Sobczuk, P.; Pawlowska, E.; Blasiak, J. The TRPA1 Ion Channel Mediates Oxidative Stress-Related Migraine Pathogenesis. Molecules 2024, 29, 3385. [Google Scholar] [CrossRef]
  93. Pires, P.W.; Earley, S. Neuroprotective effects of TRPA1 channels in the cerebral endothelium following ischemic stroke. Elife 2018, 7, e35316. [Google Scholar] [CrossRef] [PubMed]
  94. Faris, P.; Rumolo, A.; Pellavio, G.; Tanzi, M.; Vismara, M.; Berra-Romani, R.; Gerbino, A.; Corallo, S.; Pedrazzoli, P.; Laforenza, U.; et al. Transient receptor potential ankyrin 1 (TRPA1) mediates reactive oxygen species-induced Ca2+ entry, mitochondrial dysfunction, and caspase-3/7 activation in primary cultures of metastatic colorectal carcinoma cells. Cell Death Discov. 2023, 9, 213. [Google Scholar] [CrossRef]
  95. Li, J.; Zhang, H.; Du, Q.; Gu, J.; Wu, J.; Liu, Q.; Li, Z.; Zhang, T.; Xu, J.; Xie, R. Research Progress on TRPA1 in Diseases. J. Membr. Biol. 2023, 256, 301–316. [Google Scholar] [CrossRef] [PubMed]
  96. Kremeyer, B.; Lopera, F.; Cox, J.J.; Momin, A.; Rugiero, F.; Marsh, S.; Woods, C.G.; Jones, N.G.; Paterson, K.J.; Fricker, F.R.; et al. A gain-of-function mutation in TRPA1 causes familial episodic pain syndrome. Neuron 2010, 66, 671–680. [Google Scholar] [CrossRef]
  97. Rosato, A.S.; Tang, R.; Grimm, C. Two-pore and TRPML cation channels: Regulators of phagocytosis, autophagy and lysosomal exocytosis. Pharmacol. Ther. 2021, 220, 107713. [Google Scholar] [CrossRef]
  98. Chen, C.C.; Krogsaeter, E.; Grimm, C. Two-pore and TRP cation channels in endolysosomal osmo-/mechanosensation and volume regulation. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 118921. [Google Scholar] [CrossRef]
  99. Bargal, R.; Avidan, N.; Ben-Asher, E.; Olender, Z.; Zeigler, M.; Frumkin, A.; Raas-Rothschild, A.; Glusman, G.; Lancet, D.; Bach, G. Identification of the gene causing mucolipidosis type IV. Nat. Genet. 2000, 26, 118–123. [Google Scholar] [CrossRef]
  100. Sun, M.; Goldin, E.; Stahl, S.; Falardeau, J.L.; Kennedy, J.C.; Acierno, J.S., Jr.; Bove, C.; Kaneski, C.R.; Nagle, J.; Bromley, M.C.; et al. Mucolipidosis type IV is caused by mutations in a gene encoding a novel transient receptor potential channel. Hum. Mol. Genet. 2000, 9, 2471–2478. [Google Scholar] [CrossRef]
  101. Grimm, C.; Cuajungco, M.P.; van Aken, A.F.; Schnee, M.; Jörs, S.; Kros, C.J.; Ricci, A.J.; Heller, S. A helix-breaking mutation in TRPML3 leads to constitutive activity underlying deafness in the varitint-waddler mouse. Proc. Natl. Acad. Sci. USA 2007, 104, 19583–19588. [Google Scholar] [CrossRef]
  102. Rinkenberger, N.; Schoggins, J.W. Mucolipin-2 Cation Channel Increases Trafficking Efficiency of Endocytosed Viruses. mBio 2018, 9, e02314-17. [Google Scholar] [CrossRef] [PubMed]
  103. Schoggins, J.W. Interferon-stimulated genes: Roles in viral pathogenesis. Curr. Opin. Virol. 2014, 6, 40–46. [Google Scholar] [CrossRef] [PubMed]
  104. Sudarikova, A.V.; Vasileva, V.Y.; Sultanova, R.F.; Ilatovskaya, D.V. Recent advances in understanding ion transport mechanisms in polycystic kidney disease. Clin. Sci. 2021, 135, 2521–2540. [Google Scholar] [CrossRef]
  105. Kotsis, F.; Boehlke, C.; Kuehn, E.W. The ciliary flow sensor and polycystic kidney disease. Nephrol. Dial. Transplant. 2013, 28, 518–526. [Google Scholar] [CrossRef]
  106. Nigro, E.A.; Distefano, G.; Chiaravalli, M.; Matafora, V.; Castelli, M.; Pesenti Gritti, A.; Bachi, A.; Boletta, A. Polycystin-1 Regulates Actomyosin Contraction and the Cellular Response to Extracellular Stiffness. Sci. Rep. 2019, 9, 16640. [Google Scholar] [CrossRef]
  107. Peng, J.B.; Suzuki, Y.; Gyimesi, G.; Hediger, M.A. TRPV5 and TRPV6 Calcium-Selective Channels. In Calcium Entry Channels in Non-Excitable Cells; Chapter 13; Kozak, J.A., Putney, J.W., Jr., Eds.; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
  108. Ma, L.; Lee, B.H.; Clifton, H.; Schaefer, S.; Zheng, J. Nicotinic acid is a common regulator of heat-sensing TRPV1-4 ion channels. Sci. Rep. 2015, 5, 8906. [Google Scholar] [CrossRef]
  109. Haustrate, A.; Prevarskaya, N.; Lehen’kyi, V. Role of the TRPV Channels in the Endoplasmic Reticulum Calcium Homeostasis. Cells 2020, 9, 317. [Google Scholar] [CrossRef]
  110. Nakazawa, Y.; Petrova, R.S.; Sugiyama, Y.; Nagai, N.; Tamura, H.; Donaldson, P.J. Regulation of the Membrane Trafficking of the Mechanosensitive Ion Channels TRPV1 and TRPV4 by Zonular Tension, Osmotic Stress and Activators in the Mouse Lens. Int. J. Mol. Sci. 2021, 22, 12658. [Google Scholar] [CrossRef]
  111. Ji, C.; Wang, Y.; Wang, Q.; Wang, A.; Ali, A.; McCulloch, C.A. TRPV4 regulates β1 integrin-mediated cell-matrix adhesions and collagen remodeling. FASEB J. 2023, 37, e22946. [Google Scholar] [CrossRef]
  112. Berciano, J.; Baets, J.; Gallardo, E.; Zimoń, M.; García, A.; López-Laso, E.; Combarros, O.; Infante, J.; Timmerman, V.; Jordanova, A.; et al. Reduced penetrance in hereditary motor neuropathy caused by TRPV4 Arg269Cys mutation. J. Neurol. 2011, 258, 1413–1421. [Google Scholar] [CrossRef]
  113. Lorin, C.; Vögeli, I.; Niggli, E. Dystrophic cardiomyopathy: Role of TRPV2 channels in stretch-induced cell damage. Cardiovasc. Res. 2015, 106, 153–162. [Google Scholar] [CrossRef] [PubMed]
  114. Katz, B.; Zaguri, R.; Edvardson, S.; Maayan, C.; Elpeleg, O.; Lev, S.; Davidson, E.; Peters, M.; Kfir-Erenfeld, S.; Berger, E.; et al. Nociception and pain in humans lacking a functional TRPV1 channel. J. Clin. Investig. 2023, 133, e153558. [Google Scholar] [CrossRef] [PubMed]
  115. Lin, Z.; Chen, Q.; Lee, M.; Cao, X.; Zhang, J.; Ma, D.; Chen, L.; Hu, X.; Wang, H.; Wang, X.; et al. Exome sequencing reveals mutations in TRPV3 as a cause of Olmsted syndrome. Am. J. Hum. Genet. 2012, 90, 558–564. [Google Scholar] [CrossRef]
  116. Coste, B.; Mathur, J.; Schmidt, M.; Earley, T.J.; Ranade, S.; Petrus, M.J.; Dubin, A.E.; Patapoutian, A. Piezo1 and Piezo2 are essential components of distinct mechanically activated cation channels. Science 2010, 330, 55–60. [Google Scholar] [CrossRef]
  117. Zeng, W.Z.; Marshall, K.L.; Min, S.; Daou, I.; Chapleau, M.W.; Abboud, F.M.; Liberles, S.D.; Patapoutian, A. PIEZOs mediate neuronal sensing of blood pressure and the baroreceptor reflex. Science 2018, 362, 464–467. [Google Scholar] [CrossRef]
  118. Cox, C.D.; Bae, C.; Ziegler, L.; Hartley, S.; Nikolova-Krstevski, V.; Rohde, P.R.; Ng, C.A.; Sachs, F.; Gottlieb, P.A.; Martinac, B. Removal of the mechanoprotective influence of the cytoskeleton reveals PIEZO1 is gated by bilayer tension. Nat. Commun. 2016, 7, 10366. [Google Scholar] [CrossRef]
  119. Verkest, C.; Schaefer, I.; Nees, T.A.; Wang, N.; Jegelka, J.M.; Taberner, F.J.; Lechner, S.G. Intrinsically disordered intracellular domains control key features of the mechanically-gated ion channel PIEZO2. Nat. Commun. 2022, 13, 1365. [Google Scholar] [CrossRef]
  120. Wang, J.; Jiang, J.; Yang, X.; Zhou, G.; Wang, L.; Xiao, B. Tethering Piezo channels to the actin cytoskeleton for mechanogating via the cadherin-β-catenin mechanotransduction complex. Cell Rep. 2022, 38, 110342. [Google Scholar] [CrossRef]
  121. Yu, Z.Y.; Gong, H.; Kesteven, S.; Guo, Y.; Wu, J.; Li, J.V.; Cheng, D.; Zhou, Z.; Iismaa, S.E.; Kaidonis, X.; et al. Piezo1 is the cardiac mechanosensor that initiates the cardiomyocyte hypertrophic response to pressure overload in adult mice. Nat. Cardiovasc. Res. 2022, 1, 577–591. [Google Scholar] [CrossRef]
  122. Chuntharpursat-Bon, E.; Povstyan, O.V.; Ludlow, M.J.; Carrier, D.J.; Debant, M.; Shi, J.; Gaunt, H.J.; Bauer, C.C.; Curd, A.; Simon Futers, T.; et al. PIEZO1 and PECAM1 interact at cell-cell junctions and partner in endothelial force sensing. Commun. Biol. 2023, 6, 358. [Google Scholar] [CrossRef]
  123. Peyronnet, R.; Martins, J.R.; Duprat, F.; Demolombe, S.; Arhatte, M.; Jodar, M.; Tauc, M.; Duranton, C.; Paulais, M.; Teulon, J.; et al. Piezo1-dependent stretch-activated channels are inhibited by Polycystin-2 in renal tubular epithelial cells. EMBO Rep. 2013, 14, 1143–1148. [Google Scholar] [CrossRef] [PubMed]
  124. Narayanan, P.; Hütte, M.; Kudryasheva, G.; Taberner, F.J.; Lechner, S.G.; Rehfeldt, F.; Gomez-Varela, D.; Schmidt, M. Myotubularin related protein-2 and its phospholipid substrate PIP2 control Piezo2-mediated mechanotransduction in peripheral sensory neurons. Elife 2018, 7, e32346. [Google Scholar] [CrossRef] [PubMed]
  125. Ranade, S.S.; Qiu, Z.; Woo, S.H.; Hur, S.S.; Murthy, S.E.; Cahalan, S.M.; Xu, J.; Mathur, J.; Bandell, M.; Coste, B.; et al. Piezo1, a mechanically activated ion channel, is required for vascular development in mice. Proc. Natl. Acad. Sci. USA. 2014, 111, 10347–10352. [Google Scholar] [CrossRef]
  126. Retailleau, K.; Duprat, F.; Arhatte, M.; Ranade, S.S.; Peyronnet, R.; Martins, J.R.; Jodar, M.; Moro, C.; Offermanns, S.; Feng, Y.; et al. Piezo1 in Smooth Muscle Cells Is Involved in Hypertension-Dependent Arterial Remodeling. Cell Rep. 2015, 13, 1161–1171. [Google Scholar] [CrossRef]
  127. Pathak, M.M.; Nourse, J.L.; Tran, T.; Hwe, J.; Arulmoli, J.; Le, D.T.; Bernardis, E.; Flanagan, L.A.; Tombola, F. Stretch-activated ion channel Piezo1 directs lineage choice in human neural stem cells. Proc. Natl. Acad. Sci. USA 2014, 111, 16148–16153. [Google Scholar] [CrossRef]
  128. Jin, Y.; Li, J.; Wang, Y.; Ye, R.; Feng, X.; Jing, Z.; Zhao, Z. Functional role of mechanosensitive ion channel Piezo1 in human periodontal ligament cells. Angle Orthod. 2015, 85, 87–94. [Google Scholar] [CrossRef]
  129. Michishita, M.; Yano, K.; Tomita, K.I.; Matsuzaki, O.; Kasahara, K.I. Piezo1 expression increases in rat bladder after partial bladder outlet obstruction. Life Sci. 2016, 166, 1–7. [Google Scholar] [CrossRef]
  130. Syeda, R.; Florendo, M.N.; Cox, C.D.; Kefauver, J.M.; Santos, J.S.; Martinac, B.; Patapoutian, A. Piezo1 Channels Are Inherently Mechanosensitive. Cell Rep. 2016, 17, 1739–1746. [Google Scholar] [CrossRef]
  131. Procès, A.; Luciano, M.; Kalukula, Y.; Ris, L.; Gabriele, S. Multiscale Mechanobiology in Brain Physiology and Diseases. Front. Cell Dev. Biol. 2022, 10, 823857. [Google Scholar] [CrossRef]
  132. Wang, B.; Ke, W.; Wang, K.; Li, G.; Ma, L.; Lu, S.; Xiang, Q.; Liao, Z.; Luo, R.; Song, Y.; et al. Mechanosensitive Ion Channel Piezo1 Activated by Matrix Stiffness Regulates Oxidative Stress-Induced Senescence and Apoptosis in Human Intervertebral Disc Degeneration. Oxid. Med. Cell Longev. 2021, 2021, 8884922. [Google Scholar] [CrossRef]
  133. Zarychanski, R.; Schulz, V.P.; Houston, B.L.; Maksimova, Y.; Houston, D.S.; Smith, B.; Rinehart, J.; Gallagher, P.G. Mutations in the mechanotransduction protein PIEZO1 are associated with hereditary xerocytosis. Blood 2012, 120, 1908–1915. [Google Scholar] [CrossRef] [PubMed]
  134. Lukacs, V.; Mathur, J.; Mao, R.; Bayrak-Toydemir, P.; Procter, M.; Cahalan, S.M.; Kim, H.J.; Bandell, M.; Longo, N.; Day, R.W.; et al. Impaired PIEZO1 function in patients with a novel autosomal recessive congenital lymphatic dysplasia. Nat. Commun. 2015, 6, 8329. [Google Scholar] [CrossRef] [PubMed]
  135. Han, S.; Guo, X.; Wang, X.; Lin, H.; Yu, Y.; Shu, J.; Dong, M.; Yang, L. A Novel Homozygous Missense Mutation of PIEZO1 Leading to Lymphatic Malformation-6 Identified in a Family with Three Adverse Pregnancy Outcomes due to Nonimmune Fetal Hydrops. Front. Genet. 2022, 13, 856046. [Google Scholar] [CrossRef]
  136. Scimone, C.; Donato, L.; Alibrandi, S.; D’Angelo, R.; Sidoti, A. Evidences of PIEZO1 involvement in cerebral cavernous malformation pathogenesis. Microvasc. Res. 2022, 141, 104342. [Google Scholar] [CrossRef]
  137. Beurg, M.; Fettiplace, R. PIEZO2 as the anomalous mechanotransducer channel in auditory hair cells. J. Physiol. 2017, 595, 7039–7048. [Google Scholar] [CrossRef]
  138. von Buchholtz, L.J.; Ghitani, N.; Lam, R.M.; Licholai, J.A.; Chesler, A.T.; Ryba, N.J.P. Decoding Cellular Mechanisms for Mechanosensory Discrimination. Neuron 2021, 109, 285–298.e5. [Google Scholar] [CrossRef]
  139. McMillin, M.J.; Beck, A.E.; Chong, J.X.; Shively, K.M.; Buckingham, K.J.; Gildersleeve, H.I.; Aracena, M.I.; Aylsworth, A.S.; Bitoun, P.; Carey, J.C.; et al. Mutations in PIEZO2 cause Gordon syndrome, Marden-Walker syndrome, and distal arthrogryposis type 5. Am. J. Hum. Genet. 2014, 94, 734–744. [Google Scholar] [CrossRef]
  140. Chesler, A.T.; Szczot, M.; Bharucha-Goebel, D.; Čeko, M.; Donkervoort, S.; Laubacher, C.; Hayes, L.H.; Alter, K.; Zampieri, C.; Stanley, C.; et al. The Role of PIEZO2 in Human Mechanosensation. N. Engl. J. Med. 2016, 375, 1355–1364. [Google Scholar] [CrossRef]
  141. Delle Vedove, A.; Storbeck, M.; Heller, R.; Hölker, I.; Hebbar, M.; Shukla, A.; Magnusson, O.; Cirak, S.; Girisha, K.M.; O’Driscoll, M.; et al. Biallelic Loss of Proprioception-Related PIEZO2 Causes Muscular Atrophy with Perinatal Respiratory Distress, Arthrogryposis, and Scoliosis. Am. J. Hum. Genet. 2016, 99, 1206–1216. [Google Scholar] [CrossRef]
  142. Miyazawa, K.; Itoh, Y.; Fu, H.; Miyazono, K. Receptor-activated transcription factors and beyond: Multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J. Biol. Chem. 2024, 300, 107256. [Google Scholar] [CrossRef]
  143. Saha, S.; Ji, L.; de Pablo, J.J.; Palecek, S.P. TGFbeta/Activin/Nodal pathway in inhibition of human embryonic stem cell differentiation by mechanical strain. Biophys. J. 2008, 94, 4123–4133. [Google Scholar] [CrossRef] [PubMed]
  144. Grabias, B.M.; Konstantopoulos, K. Notch4-dependent antagonism of canonical TGF-β1 signaling defines unique temporal fluctuations of SMAD3 activity in sheared proximal tubular epithelial cells. Am. J. Physiol. Renal Physiol. 2013, 305, F123–F133. [Google Scholar] [CrossRef]
  145. Kunnen, S.J.; Leonhard, W.N.; Semeins, C.; Hawinkels, L.J.A.C.; Poelma, C.; Ten Dijke, P.; Bakker, A.; Hierck, B.P.; Peters, D.J.M. Fluid shear stress-induced TGF-β/ALK5 signaling in renal epithelial cells is modulated by MEK1/2. Cell Mol. Life Sci. 2017, 74, 2283–2298. [Google Scholar] [CrossRef] [PubMed]
  146. Maruri, D.P.; Iyer, K.S.; Schmidtke, D.W.; Petroll, W.M.; Varner, V.D. Signaling Downstream of Focal Adhesions Regulates Stiffness-Dependent Differences in the TGF-β1-Mediated Myofibroblast Differentiation of Corneal Keratocytes. Front. Cell Dev. Biol. 2022, 10, 886759. [Google Scholar] [CrossRef]
  147. Stutchbury, B.; Atherton, P.; Tsang, R.; Wang, D.Y.; Ballestrem, C. Distinct focal adhesion protein modules control different aspects of mechanotransduction. J. Cell Sci. 2017, 130, 1612–1624. [Google Scholar] [CrossRef]
  148. Boogerd, C.J.; Perini, I.; Kyriakopoulou, E.; Han, S.J.; La, P.; van der Swaan, B.; Berkhout, J.B.; Versteeg, D.; Monshouwer-Kloots, J.; van Rooij, E. Cardiomyocyte proliferation is suppressed by ARID1A-mediated YAP inhibition during cardiac maturation. Nat. Commun. 2023, 14, 4716. [Google Scholar] [CrossRef]
  149. Marin, T.M.; Clemente, C.F.; Santos, A.M.; Picardi, P.K.; Pascoal, V.D.; Lopes-Cendes, I.; Saad, M.J.; Franchini, K.G. Shp2 negatively regulates growth in cardiomyocytes by controlling focal adhesion kinase/Src and mTOR pathways. Circ. Res. 2008, 103, 813–824. [Google Scholar] [CrossRef]
  150. Baba, H.A.; Stypmann, J.; Grabellus, F.; Kirchhof, P.; Sokoll, A.; Schäfers, M.; Takeda, A.; Wilhelm, M.J.; Scheld, H.H.; Takeda, N.; et al. Dynamic regulation of MEK/Erks and Akt/GSK-3beta in human end-stage heart failure after left ventricular mechanical support: Myocardial mechanotransduction-sensitivity as a possible molecular mechanism. Cardiovasc. Res. 2003, 59, 390–399. [Google Scholar] [CrossRef]
  151. Kishimoto, H.; Iwasaki, M.; Wada, K.; Horitani, K.; Tsukamoto, O.; Kamikubo, K.; Nomura, S.; Matsumoto, S.; Harada, T.; Motooka, D.; et al. Wnt5a-YAP signaling axis mediates mechanotransduction in cardiac myocytes and contributes to contractile dysfunction induced by pressure overload. iScience 2023, 26, 107146. [Google Scholar] [CrossRef]
  152. Rickman, M.; Ghim, M.; Pang, K.; von Huelsen Rocha, A.C.; Drudi, E.M.; Sureda-Vives, M.; Ayoub, N.; Tajadura-Ortega, V.; George, S.J.; Weinberg, P.D.; et al. Disturbed flow increases endothelial inflammation and permeability via a Frizzled-4-β-catenin-dependent pathway. J. Cell Sci. 2023, 136, jcs260449. [Google Scholar] [CrossRef]
  153. Paolini, A.; Fontana, F.; Pham, V.C.; Rödel, C.J.; Abdelilah-Seyfried, S. Mechanosensitive Notch-Dll4 and Klf2-Wnt9 signaling pathways intersect in guiding valvulogenesis in zebrafish. Cell Rep. 2021, 37, 109782. [Google Scholar] [CrossRef] [PubMed]
  154. Saleh, R.O.; Alkhafaji, A.T.; Mohammed, J.S.; Bansal, P.; Kaur, H.; Ahmad, I.; Hjazi, A.; Mohammed, I.H.; Jawad, M.A.; Zwamel, A.H. LncRNA NEAT1 in the pathogenesis of liver-related diseases. Cell Biochem. Funct. 2024, 42, e4006. [Google Scholar] [CrossRef] [PubMed]
  155. Xu, X.; Zhang, Y.; Wang, X.; Li, S.; Tang, L. Substrate Stiffness Drives Epithelial to Mesenchymal Transition and Proliferation through the NEAT1-Wnt/β-Catenin Pathway in Liver Cancer. Int. J. Mol. Sci. 2021, 22, 12066. [Google Scholar] [CrossRef]
  156. Hino, N.; Rossetti, L.; Marín-Llauradó, A.; Aoki, K.; Trepat, X.; Matsuda, M.; Hirashima, T. ERK-Mediated Mechanochemical Waves Direct Collective Cell Polarization. Dev. Cell 2020, 53, 646–660.e8. [Google Scholar] [CrossRef] [PubMed]
  157. Hirata, H.; Gupta, M.; Vedula, S.R.; Lim, C.T.; Ladoux, B.; Sokabe, M. Actomyosin bundles serve as a tension sensor and a platform for ERK activation. EMBO Rep. 2015, 16, 250–257. [Google Scholar] [CrossRef]
  158. Shaya, O.; Binshtok, U.; Hersch, M.; Rivkin, D.; Weinreb, S.; Amir-Zilberstein, L.; Khamaisi, B.; Oppenheim, O.; Desai, R.A.; Goodyear, R.J.; et al. Cell-Cell Contact Area Affects Notch Signaling and Notch-Dependent Patterning. Dev. Cell 2017, 40, 505–511.e6. [Google Scholar] [CrossRef]
  159. He, X.; Liu, Y.; Dai, Z.; Chen, Y.; Liu, W.; Dai, H.; Hu, Y. Yoda1 pretreated BMSC derived exosomes accelerate osteogenesis by activating phospho-ErK signaling via Yoda1-mediated signal transmission. J. Nanobiotechnol. 2024, 22, 407. [Google Scholar] [CrossRef]
  160. Zheng, M.; Yao, Y.; Borkar, N.A.; Thompson, M.A.; Zhang, E.; Drake, L.Y.; Ye, X.; Vogel, E.R.; Pabelick, C.M.; Prakash, Y.S. Piezo channels modulate human lung fibroblast function. Am. J. Physiol. Lung Cell Mol. Physiol. 2024, 327, L547–L556. [Google Scholar] [CrossRef]
  161. Siddiqui, H.B.; Golcez, T.; Çelik, M.; Sevgin, B.; Çoban, M.; Süder, İ.; Kaya, Ö.; Özören, N.; Pekkan, K. Modulation of mechanosensitive genes during embryonic aortic arch development. Dev. Dyn. 2024, 254, 222–239. [Google Scholar] [CrossRef]
  162. Okamoto, S.; Chaya, T.; Omori, Y.; Kuwahara, R.; Kubo, S.; Sakaguchi, H.; Furukawa, T. Ick Ciliary Kinase Is Essential for Planar Cell Polarity Formation in Inner Ear Hair Cells and Hearing Function. J. Neurosci. 2017, 37, 2073–2085. [Google Scholar] [CrossRef]
  163. Takacs, Z.; Proikas-Cezanne, T. Primary cilia mechanosensing triggers autophagy-regulated cell volume control. Nat. Cell Biol. 2016, 18, 591–592. [Google Scholar] [CrossRef] [PubMed]
  164. Krenn, P.W.; Montanez, E.; Costell, M.; Fässler, R. Integrins, anchors and signal transducers of hematopoietic stem cells during development and in adulthood. Curr. Top Dev. Biol. 2022, 149, 203–261. [Google Scholar] [CrossRef] [PubMed]
  165. Brunet, T.; Bouclet, A.; Ahmadi, P.; Mitrossilis, D.; Driquez, B.; Brunet, A.C.; Henry, L.; Serman, F.; Béalle, G.; Ménager, C.; et al. Evolutionary conservation of early mesoderm specification by mechanotransduction in Bilateria. Nat. Commun. 2013, 4, 2821. [Google Scholar] [CrossRef] [PubMed]
  166. Röper, J.C.; Mitrossilis, D.; Stirnemann, G.; Waharte, F.; Brito, I.; Fernandez-Sanchez, M.E.; Baaden, M.; Salamero, J.; Farge, E. The major β-catenin/E-cadherin junctional binding site is a primary molecular mechano-transductor of differentiation in vivo. Elife 2018, 7, e33381. [Google Scholar] [CrossRef]
  167. Ayad, N.M.E.; Lakins, J.N.; Ghagre, A.; Ehrlicher, A.J.; Weaver, V.M. Tissue tension permits β-catenin phosphorylation to drive mesoderm specification in human embryonic stem cells. bioRxiv 2023. [Google Scholar] [CrossRef]
  168. Blum, M.; Andre, P.; Muders, K.; Schweickert, A.; Fischer, A.; Bitzer, E.; Bogusch, S.; Beyer, T.; van Straaten, H.W.; Viebahn, C. Ciliation and gene expression distinguish between node and posterior notochord in the mammalian embryo. Differentiation 2007, 75, 133–146. [Google Scholar] [CrossRef]
  169. Sampaio, P.; Ferreira, R.R.; Guerrero, A.; Pintado, P.; Tavares, B.; Amaro, J.; Smith, A.A.; Montenegro-Johnson, T.; Smith, D.J.; Lopes, S.S. Left-right organizer flow dynamics: How much cilia activity reliably yields laterality? Dev. Cell 2014, 29, 716–728. [Google Scholar] [CrossRef]
  170. Nonaka, S.; Tanaka, Y.; Okada, Y.; Takeda, S.; Harada, A.; Kanai, Y.; Kido, M.; Hirokawa, N. Randomization of left-right asymmetry due to loss of nodal cilia generating leftward flow of extraembryonic fluid in mice lacking KIF3B motor protein. Cell 1998, 95, 829–837. [Google Scholar] [CrossRef]
  171. Axelrod, J.D. Planar cell polarity signaling in the development of left-right asymmetry. Curr. Opin. Cell Biol. 2020, 62, 61–69. [Google Scholar] [CrossRef]
  172. Kamura, K.; Kobayashi, D.; Uehara, Y.; Koshida, S.; Iijima, N.; Kudo, A.; Yokoyama, T.; Takeda, H. Pkd1l1 complexes with Pkd2 on motile cilia and functions to establish the left-right axis. Development 2011, 138, 1121–1129. [Google Scholar] [CrossRef]
  173. Yoshiba, S.; Shiratori, H.; Kuo, I.Y.; Kawasumi, A.; Shinohara, K.; Nonaka, S.; Asai, Y.; Sasaki, G.; Belo, J.A.; Sasaki, H.; et al. Cilia at the node of mouse embryos sense fluid flow for left-right determination via Pkd2. Science 2012, 338, 226–231. [Google Scholar] [CrossRef] [PubMed]
  174. Grimes, D.T.; Keynton, J.L.; Buenavista, M.T.; Jin, X.; Patel, S.H.; Kyosuke, S.; Vibert, J.; Williams, D.J.; Hamada, H.; Hussain, R.; et al. Genetic Analysis Reveals a Hierarchy of Interactions between Polycystin-Encoding Genes and Genes Controlling Cilia Function during Left-Right Determination. PLoS Genet. 2016, 12, e1006070. [Google Scholar] [CrossRef] [PubMed]
  175. Logan, M.; Pagán-Westphal, S.M.; Smith, D.M.; Paganessi, L.; Tabin, C.J. The transcription factor Pitx2 mediates situs-specific morphogenesis in response to left-right asymmetric signals. Cell 1998, 94, 307–317. [Google Scholar] [CrossRef]
  176. Katoh, T.A.; Omori, T.; Mizuno, K.; Sai, X.; Minegishi, K.; Ikawa, Y.; Nishimura, H.; Itabashi, T.; Kajikawa, E.; Hiver, S.; et al. Immotile cilia mechanically sense the direction of fluid flow for left-right determination. Science 2023, 379, 66–71. [Google Scholar] [CrossRef]
  177. Katoh, T.A.; Lange, T.; Nakajima, Y.; Yashiro, K.; Okada, Y.; Hamada, H. BMP4 regulates asymmetric Pkd2 distribution in mouse nodal immotile cilia and ciliary mechanosensing required for left-right determination. Dev. Dyn. 2024, 10, 1–14. [Google Scholar] [CrossRef]
  178. Schwayer, C.; Shamipour, S.; Pranjic-Ferscha, K.; Schauer, A.; Balda, M.; Tada, M.; Matter, K.; Heisenberg, C.P. Mechanosensation of Tight Junctions Depends on ZO-1 Phase Separation and Flow. Cell 2019, 179, 937–952.e18. [Google Scholar] [CrossRef]
  179. Davidson, L.A.; Marsden, M.; Keller, R.; Desimone, D.W. Integrin alpha5beta1 and fibronectin regulate polarized cell protrusions required for Xenopus convergence and extension. Curr. Biol. 2006, 16, 833–844. [Google Scholar] [CrossRef]
  180. Marsden, M.; DeSimone, D.W. Integrin-ECM interactions regulate cadherin-dependent cell adhesion and are required for convergent extension in Xenopus. Curr. Biol. 2003, 13, 1182–1191. [Google Scholar] [CrossRef]
  181. Thompson, A.J.; Pillai, E.K.; Dimov, I.B.; Foster, S.K.; Holt, C.E.; Franze, K. Rapid changes in tissue mechanics regulate cell behaviour in the developing embryonic brain. Elife 2019, 8, e39356. [Google Scholar] [CrossRef]
  182. Barriga, E.H.; Franze, K.; Charras, G.; Mayor, R. Tissue stiffening coordinates morphogenesis by triggering collective cell migration in vivo. Nature 2018, 554, 523–527. [Google Scholar] [CrossRef]
  183. Shellard, A.; Mayor, R. Integrating chemical and mechanical signals in neural crest cell migration. Curr. Opin. Genet. Dev. 2019, 57, 16–24. [Google Scholar] [CrossRef] [PubMed]
  184. Canales Coutiño, B.; Mayor, R. Neural crest mechanosensors: Seeing old proteins in a new light. Dev. Cell 2022, 57, 1792–1801. [Google Scholar] [CrossRef] [PubMed]
  185. Liu, C.; Li, Q.; Xiao, Q.; Gong, P.; Kang, N. CHD7 Regulates Osteogenic Differentiation of Human Dental Follicle Cells via PTH1R Signaling. Stem Cells Int. 2020, 2020, 8882857. [Google Scholar] [CrossRef] [PubMed]
  186. Camargo-Sosa, K.; Colanesi, S.; Müller, J.; Schulte-Merker, S.; Stemple, D.; Patton, E.E.; Kelsh, R.N. Endothelin receptor Aa regulates proliferation and differentiation of Erb-dependent pigment progenitors in zebrafish. PLoS Genet. 2019, 15, e1007941. [Google Scholar] [CrossRef]
  187. Balczerski, B.; Matsutani, M.; Castillo, P.; Osborne, N.; Stainier, D.Y.; Crump, J.G. Analysis of sphingosine-1-phosphate signaling mutants reveals endodermal requirements for the growth but not dorsoventral patterning of jaw skeletal precursors. Dev. Biol. 2012, 362, 230–241. [Google Scholar] [CrossRef]
  188. Canales Coutiño, B.; Mayor, R. The mechanosensitive channel Piezo1 cooperates with semaphorins to control neural crest migration. Development 2021, 148, dev200001. [Google Scholar] [CrossRef]
  189. Cheng, C.; Cong, Q.; Liu, Y.; Hu, Y.; Liang, G.; Dioneda, K.M.M.; Yang, Y. Yap controls notochord formation and neural tube patterning by integrating mechanotransduction with FoxA2 and Shh expression. Sci. Adv. 2023, 9, eadf6927. [Google Scholar] [CrossRef]
  190. Voltes, A.; Hevia, C.F.; Engel-Pizcueta, C.; Dingare, C.; Calzolari, S.; Terriente, J.; Norden, C.; Lecaudey, V.; Pujades, C. Yap/Taz-TEAD activity links mechanical cues to progenitor cell behavior during zebrafish hindbrain segmentation. Development 2019, 146, dev176735. [Google Scholar] [CrossRef]
  191. Zhu, Y.; Li, X.; Janairo, R.R.R.; Kwong, G.; Tsou, A.D.; Chu, J.S.; Wang, A.; Yu, J.; Wang, D.; Li, S. Matrix stiffness modulates the differentiation of neural crest stem cells in vivo. J. Cell Physiol. 2019, 234, 7569–7578. [Google Scholar] [CrossRef]
  192. Engler, A.J.; Sen, S.; Sweeney, H.L.; Discher, D.E. Matrix elasticity directs stem cell lineage specification. Cell 2006, 126, 677–689. [Google Scholar] [CrossRef]
  193. Wilson, R.E.; Denisin, A.K.; Dunn, A.R.; Pruitt, B.L. 3D Microwell Platforms for Control of Single Cell 3D Geometry and Intracellular Organization. Cell Mol. Bioeng. 2020, 14, 1–14. [Google Scholar] [CrossRef] [PubMed]
  194. Saez, P.; Shirke, P.U.; Seth, J.R.; Alegre-Cebollada, J.; Majumder, A. Competing elastic and viscous gradients determine directional cell migration. Math Biosci. 2025, 380, 109362. [Google Scholar] [CrossRef]
  195. Rolfe, R.A.; Kenny, E.M.; Cormican, P.; Murphy, P. Transcriptome analysis of the mouse E14.5 (TS23) developing humerus and differential expression in muscle-less mutant embryos lacking mechanical stimulation. Genom. Data 2014, 2, 32–36. [Google Scholar] [CrossRef]
  196. Arvind, V.; Huang, A.H. Mechanobiology of limb musculoskeletal development. Ann. N. Y. Acad. Sci. 2017, 1409, 18–32. [Google Scholar] [CrossRef] [PubMed]
  197. Henderson, J.H.; Carter, D.R. Mechanical induction in limb morphogenesis: The role of growth-generated strains and pressures. Bone 2002, 31, 645–653. [Google Scholar] [CrossRef]
  198. Foolen, J.; van Donkelaar, C.C.; Ito, K. Intracellular tension in periosteum/perichondrium cells regulates long bone growth. J. Orthop. Res. 2011, 29, 84–91. [Google Scholar] [CrossRef]
  199. Mikic, B.; Johnson, T.L.; Chhabra, A.B.; Schalet, B.J.; Wong, M.; Hunziker, E.B. Differential effects of embryonic immobilization on the development of fibrocartilaginous skeletal elements. J. Rehabil. Res. Dev. 2000, 37, 127–133. [Google Scholar]
  200. Subramanian, A.; Kanzaki, L.F.; Schilling, T.F. Mechanical force regulates Sox9 expression at the developing enthesis. Development 2023, 150, dev201141. [Google Scholar] [CrossRef]
  201. Vermeulen, S.; Roumans, N.; Honig, F.; Carlier, A.; Hebels, D.G.A.J.; Eren, A.D.; Dijke, P.T.; Vasilevich, A.; de Boer, J. Mechanotransduction is a context-dependent activator of TGF-β signaling in mesenchymal stem cells. Biomaterials 2020, 259, 120331. [Google Scholar] [CrossRef]
  202. Langen, U.H.; Pitulescu, M.E.; Kim, J.M.; Enriquez-Gasca, R.; Sivaraj, K.K.; Kusumbe, A.P.; Singh, A.; Di Russo, J.; Bixel, M.G.; Zhou, B.; et al. Cell-matrix signals specify bone endothelial cells during developmental osteogenesis. Nat. Cell Biol. 2017, 19, 189–201. [Google Scholar] [CrossRef]
  203. Wein, M.N. Blood and bones: Mechanical cues and Hippo signaling drive vascular invasion during limb formation. Dev. Cell 2024, 59, 173–174. [Google Scholar] [CrossRef] [PubMed]
  204. Yang, Y.; Paivinen, P.; Xie, C.; Krup, A.L.; Makela, T.P.; Mostov, K.E.; Reiter, J.F. Ciliary Hedgehog signaling patterns the digestive system to generate mechanical forces driving elongation. Nat. Commun. 2021, 12, 7186. [Google Scholar] [CrossRef] [PubMed]
  205. Gargalionis, A.N.; Adamopoulos, C.; Vottis, C.T.; Papavassiliou, A.G.; Basdra, E.K. Runx2 and Polycystins in Bone Mechanotransduction: Challenges for Therapeutic Opportunities. Int. J. Mol. Sci. 2024, 25, 5291. [Google Scholar] [CrossRef] [PubMed]
  206. Shewale, B.; Ebrahim, T.; Samal, A.; Dubois, N. Molecular Regulation of Cardiomyocyte Maturation. Curr. Cardiol. Rep. 2025, 27, 32. [Google Scholar] [CrossRef]
  207. Yang, Y.; Xu, Z.; He, S.; Wang, C.; Li, R.; Zhang, R.; Li, J.; Yang, Z.; Li, H.; Liu, S.; et al. Developmental dynamics mimicking inversely engineered pericellular matrix for articular cartilage regeneration. Biomaterials 2025, 317, 123066. [Google Scholar] [CrossRef]
  208. Estévez, M.; Cicuéndez, M.; Colilla, M.; Vallet-Regí, M.; González, B.; Izquierdo-Barba, I. Magnetic colloidal nanoformulations to remotely trigger mechanotransduction for osteogenic differentiation. J. Colloid Interface Sci. 2024, 664, 454–468. [Google Scholar] [CrossRef]
  209. Ihalainen, T.O.; Aires, L.; Herzog, F.A.; Schwartlander, R.; Moeller, J.; Vogel, V. Differential basal-to-apical accessibility of lamin A/C epitopes in the nuclear lamina regulated by changes in cytoskeletal tension. Nat. Mater. 2015, 14, 1252–1261. [Google Scholar] [CrossRef]
  210. Sturm, G.; Cardenas, A.; Bind, M.A.; Horvath, S.; Wang, S.; Wang, Y.; Hägg, S.; Hirano, M.; Picard, M. Human aging DNA methylation signatures are conserved but accelerated in cultured fibroblasts. Epigenetics 2019, 14, 961–976. [Google Scholar] [CrossRef]
  211. Beedle, A.E.; Roca-Cusachs, P. The reversibility of cellular mechano-activation. Curr. Opin. Cell Biol. 2023, 84, 102229. [Google Scholar] [CrossRef]
  212. Dudaryeva, O.Y.; Bernhard, S.; Tibbitt, M.W.; Labouesse, C. Implications of Cellular Mechanical Memory in Bioengineering. ACS Biomater. Sci. Eng. 2023, 9, 5985–5998. [Google Scholar] [CrossRef]
  213. Bakhshandeh, B.; Sorboni, S.G.; Ranjbar, N.; Deyhimfar, R.; Abtahi, M.S.; Izady, M.; Kazemi, N.; Noori, A.; Pennisi, C.P. Mechanotransduction in tissue engineering: Insights into the interaction of stem cells with biomechanical cues. Exp. Cell Res. 2023, 431, 113766. [Google Scholar] [CrossRef] [PubMed]
  214. Zakrzewicz, A.; Secomb, T.W.; Pries, A.R. Angioadaptation: Keeping the vascular system in shape. News Physiol. Sci. 2002, 17, 197–201. [Google Scholar] [CrossRef] [PubMed]
  215. Risau, W. Angiogenesis and endothelial cell function. Arzneimittelforschung 1994, 44, 416–417. [Google Scholar]
  216. Bentley, K.; Gerhardt, H.; Bates, P.A. Agent-based simulation of notch-mediated tip cell selection in angiogenic sprout initialisation. J. Theor. Biol. 2008, 250, 25–36. [Google Scholar] [CrossRef]
  217. Hiepen, C.; Benamar, M.; Barrasa-Fano, J.; Condor, M.; Ilhan, M.; Münch, J.; Hastar, N.; Kerkhoff, Y.; Harms, G.S.; Mielke, T.; et al. Endothelial tip-cell position, filopodia formation and biomechanics require BMPR2 expression and signaling. Commun. Biol. 2025, 8, 21. [Google Scholar] [CrossRef]
  218. Ramirez, F.; Caescu, C.; Wondimu, E.; Galatioto, J. Marfan syndrome; A connective tissue disease at the crossroads of mechanotransduction, TGFβ signaling and cell stemness. Matrix Biol. 2018, 71, 82–89. [Google Scholar] [CrossRef]
  219. Alonso, F.; Dong, Y.; Li, L.; Jahjah, T.; Dupuy, J.W.; Fremaux, I.; Reinhardt, D.P.; Génot, E. Fibrillin-1 regulates endothelial sprouting during angiogenesis. Proc. Natl. Acad. Sci. USA 2023, 120, e2221742120. [Google Scholar] [CrossRef]
  220. Wang, W.; Zanotelli, M.R.; Sabo, L.N.; Fabiano, E.D.; Goldfield, N.M.; Le, C.; Techasiriwan, E.P.; Lopez, S.; Berestesky, E.D.; Reinhart-King, C.A. Collagen density regulates tip-stalk cell rearrangement during angiogenesis via cellular bioenergetics. APL Bioeng. 2024, 8, 026120. [Google Scholar] [CrossRef]
  221. Vaeyens, M.M.; Jorge-Peñas, A.; Barrasa-Fano, J.; Steuwe, C.; Heck, T.; Carmeliet, P.; Roeffaers, M.; Van Oosterwyck, H. Matrix deformations around angiogenic sprouts correlate to sprout dynamics and suggest pulling activity. Angiogenesis 2020, 23, 315–324. [Google Scholar] [CrossRef]
  222. Ahmed, T.; Ramonett, A.; Kwak, E.A.; Kumar, S.; Flores, P.C.; Ortiz, H.R.; Langlais, P.R.; Hund, T.J.; Mythreye, K.; Lee, N.Y. Endothelial tip/stalk cell selection requires BMP9-induced βIV-spectrin expression during sprouting angiogenesis. Mol. Biol. Cell 2023, 34, ar72. [Google Scholar] [CrossRef]
  223. Carvalho, J.R.; Fortunato, I.C.; Fonseca, C.G.; Pezzarossa, A.; Barbacena, P.; Dominguez-Cejudo, M.A.; Vasconcelos, F.F.; Santos, N.C.; Carvalho, F.A.; Franco, C.A. Non-canonical Wnt signaling regulates junctional mechanocoupling during angiogenic collective cell migration. Elife 2019, 8, e45853. [Google Scholar] [CrossRef] [PubMed]
  224. Burri, P.H.; Djonov, V. Intussusceptive angiogenesis--the alternative to capillary sprouting. Mol. Asp. Med. 2002, 23, S1–S27. [Google Scholar] [CrossRef] [PubMed]
  225. le Noble, F.; Moyon, D.; Pardanaud, L.; Yuan, L.; Djonov, V.; Matthijsen, R.; Bréant, C.; Fleury, V.; Eichmann, A. Flow regulates arterial-venous differentiation in the chick embryo yolk sac. Development 2004, 131, 361–375. [Google Scholar] [CrossRef]
  226. Perryn, E.D.; Czirók, A.; Little, C.D. Vascular sprout formation entails tissue deformations and VE-cadherin-dependent cell-autonomous motility. Dev. Biol. 2008, 313, 545–555. [Google Scholar] [CrossRef] [PubMed]
  227. Gajdusek, C.M.; Luo, Z.; Mayberg, M.R. Basic fibroblast growth factor and transforming growth factor beta-1: Synergistic mediators of angiogenesis in vitro. J. Cell Physiol. 1993, 157, 133–144. [Google Scholar] [CrossRef] [PubMed]
  228. Padget, R.L.; Mohite, S.S.; Hoog, T.G.; Justis, B.S.; Green, B.E.; Udan, R.S. Hemodynamic force is required for vascular smooth muscle cell recruitment to blood vessels during mouse embryonic development. Mech. Dev. 2019, 156, 8–19. [Google Scholar] [CrossRef]
  229. Lenard, A.; Daetwyler, S.; Betz, C.; Ellertsdottir, E.; Belting, H.G.; Huisken, J.; Affolter, M. Endothelial cell self-fusion during vascular pruning. PLoS Biol. 2015, 13, e1002126. [Google Scholar] [CrossRef]
  230. Sugden, W.W.; Meissner, R.; Aegerter-Wilmsen, T.; Tsaryk, R.; Leonard, E.V.; Bussmann, J.; Hamm, M.J.; Herzog, W.; Jin, Y.; Jakobsson, L.; et al. Endoglin controls blood vessel diameter through endothelial cell shape changes in response to haemodynamic cues. Nat. Cell Biol. 2017, 19, 653–665. [Google Scholar] [CrossRef]
  231. Meeson, A.P.; Argilla, M.; Ko, K.; Witte, L.; Lang, R.A. VEGF deprivation-induced apoptosis is a component of programmed capillary regression. Development 1999, 126, 1407–1415. [Google Scholar] [CrossRef]
  232. Nieminen, T.; Toivanen, P.I.; Rintanen, N.; Heikura, T.; Jauhiainen, S.; Airenne, K.J.; Alitalo, K.; Marjomäki, V.; Ylä-Herttuala, S. The impact of the receptor binding profiles of the vascular endothelial growth factors on their angiogenic features. Biochim. Biophys. Acta 2014, 1840, 454–463. [Google Scholar] [CrossRef]
  233. Echarri, A.; Del Pozo, M.A. Caveolae—Mechanosensitive membrane invaginations linked to actin filaments. J. Cell Sci. 2015, 128, 2747–2758. [Google Scholar] [CrossRef] [PubMed]
  234. Givens, C.; Tzima, E. Endothelial Mechanosignaling: Does One Sensor Fit All? Antioxid. Redox Signal 2016, 25, 373–388. [Google Scholar] [CrossRef] [PubMed]
  235. Chatterjee, S.; Browning, E.A.; Hong, N.; DeBolt, K.; Sorokina, E.M.; Liu, W.; Birnbaum, M.J.; Fisher, A.B. Membrane depolarization is the trigger for PI3K/Akt activation and leads to the generation of ROS. Am. J. Physiol. Heart Circ. Physiol. 2012, 302, H105–H114. [Google Scholar] [CrossRef] [PubMed]
  236. Chatterjee, S.; Fisher, A.B. Mechanotransduction in the endothelium: Role of membrane proteins and reactive oxygen species in sensing, transduction, and transmission of the signal with altered blood flow. Antioxid. Redox Signal. 2014, 20, 899–913. [Google Scholar] [CrossRef]
  237. Takeda, H.; Komori, K.; Nishikimi, N.; Nimura, Y.; Sokabe, M.; Naruse, K. Bi-phasic activation of eNOS in response to uni-axial cyclic stretch is mediated by differential mechanisms in BAECs. Life Sci. 2006, 79, 233–239. [Google Scholar] [CrossRef]
  238. Gomes, A.M.; Pinto, T.S.; da Costa Fernandes, C.J.; da Silva, R.A.; Zambuzzi, W.F. Wortmannin targeting phosphatidylinositol 3-kinase suppresses angiogenic factors in shear-stressed endothelial cells. J. Cell Physiol. 2020, 235, 5256–5269. [Google Scholar] [CrossRef]
  239. Cronin, N.M.; Dawson, L.W.; DeMali, K.A. Mechanical activation of VE-cadherin stimulates AMPK to increase endothelial cell metabolism and vasodilation. bioRxiv 2024. [Google Scholar] [CrossRef]
  240. LaValley, D.J.; Zanotelli, M.R.; Bordeleau, F.; Wang, W.; Schwager, S.C.; Reinhart-King, C.A. Matrix Stiffness Enhances VEGFR-2 Internalization, Signaling, and Proliferation in Endothelial Cells. Converg. Sci. Phys. Oncol. 2017, 3, 044001. [Google Scholar] [CrossRef]
  241. Labrecque, L.; Royal, I.; Surprenant, D.S.; Patterson, C.; Gingras, D.; Béliveau, R. Regulation of vascular endothelial growth factor receptor-2 activity by caveolin-1 and plasma membrane cholesterol. Mol. Biol. Cell. 2003, 14, 334–347. [Google Scholar] [CrossRef]
  242. Shin, H.; Haga, J.H.; Kosawada, T.; Kimura, K.; Li, Y.S.; Chien, S.; Schmid-Schönbein, G.W. Fine control of endothelial VEGFR-2 activation: Caveolae as fluid shear stress shelters for membrane receptors. Biomech. Model Mechanobiol. 2019, 18, 5–16. [Google Scholar] [CrossRef]
  243. Zhao, C.; Kam, H.T.; Chen, Y.; Gong, G.; Hoi, M.P.; Skalicka-Woźniak, K.; Dias, A.C.P.; Lee, S.M. Crocetin and Its Glycoside Crocin, Two Bioactive Constituents from Crocus sativus L. (Saffron), Differentially Inhibit Angiogenesis by Inhibiting Endothelial Cytoskeleton Organization and Cell Migration Through VEGFR2/SRC/FAK and VEGFR2/MEK/ERK Signaling Pathways. Front. Pharmacol. 2021, 12, 675359. [Google Scholar] [CrossRef]
  244. Matsuo, E.; Okamoto, T.; Ito, A.; Kawamoto, E.; Asanuma, K.; Wada, K.; Shimaoka, M.; Takao, M.; Shimamoto, A. Substrate stiffness modulates endothelial cell function via the YAP-Dll4-Notch1 pathway. Exp. Cell Res. 2021, 408, 112835. [Google Scholar] [CrossRef] [PubMed]
  245. Kretschmer, M.; Mamistvalov, R.; Sprinzak, D.; Vollmar, A.M.; Zahler, S. Matrix stiffness regulates Notch signaling activity in endothelial cells. J. Cell Sci. 2023, 136, jcs260442. [Google Scholar] [CrossRef] [PubMed]
  246. Mack, J.J.; Mosqueiro, T.S.; Archer, B.J.; Jones, W.M.; Sunshine, H.; Faas, G.C.; Briot, A.; Aragón, R.L.; Su, T.; Romay, M.C.; et al. NOTCH1 is a mechanosensor in adult arteries. Nat. Commun. 2017, 8, 1620. [Google Scholar] [CrossRef]
  247. Ruter, D.L.; Liu, Z.; Ngo, K.M.; Shaka, X.; Marvin, A.; Buglak, D.B.; Kidder, E.J.; Bautch, V.L. SMAD6 transduces endothelial cell flow responses required for blood vessel homeostasis. Angiogenesis 2021, 24, 387–398. [Google Scholar] [CrossRef]
  248. Hooglugt, A.; van der Stoel, M.M.; Shapeti, A.; Neep, B.F.; de Haan, A.; van Oosterwyck, H.; Boon, R.A.; Huveneers, S. DLC1 promotes mechanotransductive feedback for YAP via RhoGAP-mediated focal adhesion turnover. J. Cell Sci. 2024, 137, jcs261687. [Google Scholar] [CrossRef]
  249. Wang, X.; Bove, A.M.; Simone, G.; Ma, B. Molecular Bases of VEGFR-2-Mediated Physiological Function and Pathological Role. Front. Cell Dev. Biol. 2020, 8, 599281. [Google Scholar] [CrossRef]
  250. Wang, K.C.; Yeh, Y.T.; Nguyen, P.; Limqueco, E.; Lopez, J.; Thorossian, S.; Guan, K.L.; Li, Y.J.; Chien, S. Flow-dependent YAP/TAZ activities regulate endothelial phenotypes and atherosclerosis. Proc. Natl. Acad. Sci. USA 2016, 113, 11525–11530. [Google Scholar] [CrossRef]
  251. Rüdiger, D.; Kick, K.; Goychuk, A.; Vollmar, A.M.; Frey, E.; Zahler, S. Cell-Based Strain Remodeling of a Nonfibrous Matrix as an Organizing Principle for Vasculogenesis. Cell Rep. 2020, 32, 108015. [Google Scholar] [CrossRef]
  252. Kwon, H.B.; Wang, S.; Helker, C.S.; Rasouli, S.J.; Maischein, H.M.; Offermanns, S.; Herzog, W.; Stainier, D.Y. In vivo modulation of endothelial polarization by Apelin receptor signalling. Nat. Commun. 2016, 7, 11805. [Google Scholar] [CrossRef]
  253. Hoefer, I.E.; den Adel, B.; Daemen, M.J. Biomechanical factors as triggers of vascular growth. Cardiovasc. Res. 2013, 99, 276–283. [Google Scholar] [CrossRef] [PubMed]
  254. Hong, K.S.; Kim, K.; Hill, M.A. Regulation of blood flow in small arteries: Mechanosensory events underlying myogenic vasoconstriction. J. Exerc. Rehabil. 2020, 16, 207–215. [Google Scholar] [CrossRef] [PubMed]
  255. Tanaka, K.; Chen, M.; Prendergast, A.; Zhuang, Z.; Nasiri, A.; Joshi, D.; Hintzen, J.; Chung, M.; Kumar, A.; Mani, A.; et al. Latrophilin-2 mediates fluid shear stress mechanotransduction at endothelial junctions. EMBO J. 2024, 43, 3175–3191. [Google Scholar] [CrossRef]
  256. Yoshino, D.; Funamoto, K.; Sato, K.; Kenry Sato, M.; Lim, C.T. Hydrostatic pressure promotes endothelial tube formation through aquaporin 1 and Ras-ERK signaling. Commun. Biol. 2020, 3, 152. [Google Scholar] [CrossRef]
  257. Yamamoto, K.; Ando, J. Endothelial cell and model membranes respond to shear stress by rapidly decreasing the order of their lipid phases. J. Cell Sci. 2013, 126, 1227–1234. [Google Scholar] [CrossRef]
  258. Lundbaek, J.A.; Collingwood, S.A.; Ingólfsson, H.I.; Kapoor, R.; Andersen, O.S. Lipid bilayer regulation of membrane protein function: Gramicidin channels as molecular force probes. J. R. Soc. Interface 2010, 7, 373–395. [Google Scholar] [CrossRef]
  259. Sun, X.; Fu, Y.; Gu, M.; Zhang, L.; Li, D.; Li, H.; Chien, S.; Shyy, J.Y.; Zhu, Y. Activation of integrin α5 mediated by flow requires its translocation to membrane lipid rafts in vascular endothelial cells. Proc. Natl. Acad. Sci. USA 2016, 113, 769–774. [Google Scholar] [CrossRef]
  260. Yamamoto, K.; Ando, J. Emerging Role of Plasma Membranes in Vascular Endothelial Mechanosensing. Circ. J. 2018, 82, 2691–2698. [Google Scholar] [CrossRef]
  261. Yamashiro, Y.; Thang, B.Q.; Ramirez, K.; Shin, S.J.; Kohata, T.; Ohata, S.; Nguyen, T.A.V.; Ohtsuki, S.; Nagayama, K.; Yanagisawa, H. Matrix mechanotransduction mediated by thrombospondin-1/integrin/YAP in the vascular remodeling. Proc. Natl. Acad. Sci. USA 2020, 117, 9896–9905. [Google Scholar] [CrossRef]
  262. Kotini, M.P.; van der Stoel, M.M.; Yin, J.; Han, M.K.; Kirchmaier, B.; de Rooij, J.; Affolter, M.; Huveneers, S.; Belting, H.G. Vinculin controls endothelial cell junction dynamics during vascular lumen formation. Cell Rep. 2022, 39, 110658. [Google Scholar] [CrossRef]
  263. van der Stoel, M.M.; Kotini, M.P.; Schoon, R.M.; Affolter, M.; Belting, H.G.; Huveneers, S. Vinculin strengthens the endothelial barrier during vascular development. Vasc. Biol. 2023, 5, e220012. [Google Scholar] [CrossRef] [PubMed]
  264. Chen, C.Y.; Tsai, H.Y.; Tsai, S.H.; Chu, P.H.; Huang, P.H.; Chen, J.W.; Lin, S.J. Deletion of the FHL2 gene attenuates intima-media thickening in a partially ligated carotid artery ligated mouse model. J. Cell Mol. Med. 2020, 24, 160–173. [Google Scholar] [CrossRef] [PubMed]
  265. Seetharaman, S.; Devany, J.; Kim, H.R.; van Bodegraven, E.; Chmiel, T.; Tzu-Pin, S.; Chou, W.H.; Fang, Y.; Gardel, M.L. Mechanosensitive FHL2 tunes endothelial function. bioRxiv 2024. [Google Scholar] [CrossRef]
  266. Suresh Babu, S.; Wojtowicz, A.; Freichel, M.; Birnbaumer, L.; Hecker, M.; Cattaruzza, M. Mechanism of stretch-induced activation of the mechanotransducer zyxin in vascular cells. Sci. Signal 2012, 5, ra91. [Google Scholar] [CrossRef]
  267. Smyth, J.T.; Hwang, S.Y.; Tomita, T.; DeHaven, W.I.; Mercer, J.C.; Putney, J.W. Activation and regulation of store-operated calcium entry. J. Cell Mol. Med. 2010, 14, 2337–2349. [Google Scholar] [CrossRef]
  268. Thakore, P.; Earley, S. Transient Receptor Potential Channels and Endothelial Cell Calcium Signaling. Compr. Physiol. 2019, 9, 1249–1277. [Google Scholar] [CrossRef]
  269. Savage, A.M.; Kurusamy, S.; Chen, Y.; Jiang, Z.; Chhabria, K.; MacDonald, R.B.; Kim, H.R.; Wilson, H.L.; van Eeden, F.J.M.; Armesilla, A.L.; et al. tmem33 is essential for VEGF-mediated endothelial calcium oscillations and angiogenesis. Nat. Commun. 2019, 10, 732. [Google Scholar] [CrossRef]
  270. Antigny, F.; Girardin, N.; Frieden, M. Transient receptor potential canonical channels are required for in vitro endothelial tube formation. J. Biol. Chem. 2012, 287, 5917–5927. [Google Scholar] [CrossRef]
  271. Qin, W.; Xie, W.; Xia, N.; He, Q.; Sun, T. Silencing of Transient Receptor Potential Channel 4 Alleviates oxLDL-induced Angiogenesis in Human Coronary Artery Endothelial Cells by Inhibition of VEGF and NF-κB. Med. Sci. Monit. 2016, 22, 930–936. [Google Scholar] [CrossRef]
  272. Song, H.B.; Jun, H.O.; Kim, J.H.; Fruttiger, M.; Kim, J.H. Suppression of transient receptor potential canonical channel 4 inhibits vascular endothelial growth factor-induced retinal neovascularization. Cell Calcium 2015, 57, 101–108. [Google Scholar] [CrossRef]
  273. Maltaneri, R.E.; Schiappacasse, A.; Chamorro, M.E.; Nesse, A.B.; Vittori, D.C. Participation of membrane calcium channels in erythropoietin-induced endothelial cell migration. Eur. J. Cell Biol. 2018, 97, 411–421. [Google Scholar] [CrossRef] [PubMed]
  274. Weber, E.W.; Han, F.; Tauseef, M.; Birnbaumer, L.; Mehta, D.; Muller, W.A. TRPC6 is the endothelial calcium channel that regulates leukocyte transendothelial migration during the inflammatory response. J. Exp. Med. 2015, 212, 1883–1899. [Google Scholar] [CrossRef]
  275. Solano, A.S.; Lavanderos, B.; Metwally, E.; Earley, S. Transient Receptor Potential Channels in Vascular Mechanotransduction. Am. J. Hypertens. 2024, 23, hpae134. [Google Scholar] [CrossRef] [PubMed]
  276. Yang, D.; Luo, Z.; Ma, S.; Wong, W.T.; Ma, L.; Zhong, J.; He, H.; Zhao, Z.; Cao, T.; Yan, Z.; et al. Activation of TRPV1 by dietary capsaicin improves endothelium-dependent vasorelaxation and prevents hypertension. Cell Metab. 2010, 12, 130–141. [Google Scholar] [CrossRef] [PubMed]
  277. Lee, B.H.; Zheng, J. Proton block of proton-activated TRPV1 current. J. Gen. Physiol. 2015, 146, 147–159. [Google Scholar] [CrossRef]
  278. Parpaite, T.; Cardouat, G.; Mauroux, M.; Gillibert-Duplantier, J.; Robillard, P.; Quignard, J.F.; Marthan, R.; Savineau, J.P.; Ducret, T. Effect of hypoxia on TRPV1 and TRPV4 channels in rat pulmonary arterial smooth muscle cells. Pflugers Arch. 2016, 468, 111–130. [Google Scholar] [CrossRef]
  279. Su, L.; Zhang, Y.; He, K.; Wei, S.; Pei, H.; Wang, Q.; Yang, D.; Yang, Y. Activation of transient receptor potential vanilloid 1 accelerates re-endothelialization and inhibits neointimal formation after vascular injury. J. Vasc. Surg. 2017, 65, 197–205.e2. [Google Scholar] [CrossRef]
  280. Lu, Q.; Zemskov, E.A.; Sun, X.; Wang, H.; Yegambaram, M.; Wu, X.; Garcia-Flores, A.; Song, S.; Tang, H.; Kangath, A.; et al. Activation of the mechanosensitive Ca2+ channel TRPV4 induces endothelial barrier permeability via the disruption of mitochondrial bioenergetics. Redox Biol. 2021, 38, 101785. [Google Scholar] [CrossRef]
  281. Suresh, K.; Servinsky, L.; Jiang, H.; Bigham, Z.; Yun, X.; Kliment, C.; Huetsch, J.; Damarla, M.; Shimoda, L.A. Reactive oxygen species induced Ca2+ influx via TRPV4 and microvascular endothelial dysfunction in the SU5416/hypoxia model of pulmonary arterial hypertension. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 314, L893–L907. [Google Scholar] [CrossRef]
  282. O’Leary, C.; McGahon, M.K.; Ashraf, S.; McNaughten, J.; Friedel, T.; Cincolà, P.; Barabas, P.; Fernandez, J.A.; Stitt, A.W.; McGeown, J.G.; et al. Involvement of TRPV1 and TRPV4 Channels in Retinal Angiogenesis. Investig. Ophthalmol. Vis. Sci. 2019, 60, 3297–3309. [Google Scholar] [CrossRef]
  283. Cappelli, H.C.; Guarino, B.D.; Kanugula, A.K.; Adapala, R.K.; Perera, V.; Smith, M.A.; Paruchuri, S.; Thodeti, C.K. Transient receptor potential vanilloid 4 channel deletion regulates pathological but not developmental retinal angiogenesis. J. Cell Physiol. 2021, 236, 3770–3779. [Google Scholar] [CrossRef] [PubMed]
  284. Dragoni, S.; Guerra, G.; Fiorio Pla, A.; Bertoni, G.; Rappa, A.; Poletto, V.; Bottino, C.; Aronica, A.; Lodola, F.; Cinelli, M.P.; et al. A functional transient receptor potential vanilloid 4 (TRPV4) channel is expressed in human endothelial progenitor cells. J. Cell Physiol. 2015, 230, 95–104. [Google Scholar] [CrossRef] [PubMed]
  285. Cappelli, H.C.; Kanugula, A.K.; Adapala, R.K.; Amin, V.; Sharma, P.; Midha, P.; Paruchuri, S.; Thodeti, C.K. Mechanosensitive TRPV4 channels stabilize VE-cadherin junctions to regulate tumor vascular integrity and metastasis. Cancer Lett. 2019, 442, 15–20. [Google Scholar] [CrossRef]
  286. Lamalice, L.; Houle, F.; Huot, J. Phosphorylation of Tyr1214 within VEGFR-2 triggers the recruitment of Nck and activation of Fyn leading to SAPK2/p38 activation and endothelial cell migration in response to VEGF. J. Biol. Chem. 2006, 281, 34009–34020. [Google Scholar] [CrossRef]
  287. Li, X.; Li, X.; Chen, Y.; Wang, Y.; Li, X.; Hao, A.; Hu, Y.; Li, X. Correlation of vascular change with TRPV1, TRPV4, and TRPA1 in a rat model of inferior gluteal artery perforator flap. Wound Repair Regen. 2022, 30, 365–375. [Google Scholar] [CrossRef]
  288. Amoakon, J.P.; Lee, J.; Liyanage, P.; Arora, K.; Karlstaedt, A.; Mylavarapu, G.; Amin, R.; Naren, A.P. Defective CFTR modulates mechanosensitive channels TRPV4 and PIEZO1 and drives endothelial barrier failure. iScience 2024, 27, 110703. [Google Scholar] [CrossRef]
  289. Li, J.; Hou, B.; Tumova, S.; Muraki, K.; Bruns, A.; Ludlow, M.J.; Sedo, A.; Hyman, A.J.; McKeown, L.; Young, R.S.; et al. Piezo1 integration of vascular architecture with physiological force. Nature 2014, 515, 279–282. [Google Scholar] [CrossRef]
  290. Chen, P.; Zhang, G.; Jiang, S.; Ning, Y.; Deng, B.; Pan, X.; Liu, S.; He, Y.; Zhang, L.; Wan, R.; et al. The mechanosensitive Piezo1 orchestrating angiogenesis is essential in bone fracture repair. biorXiv 2019. [Google Scholar] [CrossRef]
  291. Abello, J.; Yin, Y.; Zhao, Y.; Maurer, J.; Lee, J.; Bodell, C.; Clevenger, A.J.; Burton, Z.; Goeckel, M.E.; Lin, M.; et al. Endothelial cell Piezo1 promotes vascular smooth muscle cell differentiation on large arteries. bioRxiv 2024. [Google Scholar] [CrossRef]
  292. Lai, A.; Zhou, Y.; Thurgood, P.; Chheang, C.; Chandra Sekar, N.; Nguyen, N.; Peter, K.; Khoshmanesh, K.; Baratchi, S. Endothelial Response to the Combined Biomechanics of Vessel Stiffness and Shear Stress Is Regulated via Piezo1. ACS Appl. Mater. Interfaces 2023, 15, 59103–59116. [Google Scholar] [CrossRef]
  293. Kang, H.; Hong, Z.; Zhong, M.; Klomp, J.; Bayless, K.J.; Mehta, D.; Karginov, A.V.; Hu, G.; Malik, A.B. Piezo1 mediates angiogenesis through activation of MT1-MMP signaling. Am. J. Physiol. Cell Physiol. 2019, 316, C92–C103. [Google Scholar] [CrossRef] [PubMed]
  294. Jiang, M.; Zhang, Y.X.; Bu, W.J.; Li, P.; Chen, J.H.; Cao, M.; Dong, Y.C.; Sun, Z.J.; Dong, D.L. Piezo1 channel activation stimulates ATP production through enhancing mitochondrial respiration and glycolysis in vascular endothelial cells. Br. J. Pharmacol. 2023, 180, 1862–1877. [Google Scholar] [CrossRef] [PubMed]
  295. Kim, S.; Chung, M.; Jeon, N.L. Three-dimensional biomimetic model to reconstitute sprouting lymphangiogenesis in vitro. Biomaterials 2016, 78, 115–128. [Google Scholar] [CrossRef] [PubMed]
  296. Choi, D.; Park, E.; Yu, R.P.; Cooper, M.N.; Cho, I.T.; Choi, J.; Yu, J.; Zhao, L.; Yum, J.I.; Yu, J.S.; et al. Piezo1-Regulated Mechanotransduction Controls Flow-Activated Lymphatic Expansion. Circ. Res. 2022, 131, e2–e21. [Google Scholar] [CrossRef]
  297. Grimm, L.; Mason, E.; Yu, H.; Dudczig, S.; Panara, V.; Chen, T.; Bower, N.I.; Paterson, S.; Galeano, M.R.; Kobayashi, S.; et al. Single-cell analysis of lymphatic endothelial cell fate specification and differentiation during zebrafish development. EMBO J. 2023, 42, e112590. [Google Scholar] [CrossRef]
  298. Choi, D.; Park, E.; Jung, E.; Seong, Y.J.; Yoo, J.; Lee, E.; Hong, M.; Lee, S.; Ishida, H.; Burford, J.; et al. Laminar flow downregulates Notch activity to promote lymphatic sprouting. J. Clin. Investig. 2017, 127, 1225–1240. [Google Scholar] [CrossRef]
  299. Choi, D.; Park, E.; Jung, E.; Seong, Y.J.; Hong, M.; Lee, S.; Burford, J.; Gyarmati, G.; Peti-Peterdi, J.; Srikanth, S.; et al. ORAI1 Activates Proliferation of Lymphatic Endothelial Cells in Response to Laminar Flow Through Krüppel-Like Factors 2 and 4. Circ. Res. 2017, 120, 1426–1439. [Google Scholar] [CrossRef]
  300. Qin, Z.; Chen, H.; Fang, Y.; Wu, G.; Chen, Q.; Xue, B.; Xu, R.; Zheng, K.; Jiang, H. Matrix Stiffness of GelMA Hydrogels Regulates Lymphatic Endothelial Cells toward Enhanced Lymphangiogenesis. ACS Appl. Mater. Interfaces 2024, 16, 55130–55141. [Google Scholar] [CrossRef]
  301. Ronaldson, P.T.; Davis, T.P. Transport Mechanisms at the Blood-Brain Barrier and in Cellular Compartments of the Neurovascular Unit: Focus on CNS Delivery of Small Molecule Drugs. Pharmaceutics 2022, 14, 1501. [Google Scholar] [CrossRef]
  302. Zhang, Q.; Wang, J.; Chen, Z.; Qin, H.; Zhang, Q.; Tian, B.; Li, X. Transcytosis: An effective mechanism to enhance nanoparticle extravasation and infiltration through biological barriers. Biomed. Mater. 2025, 20, 022003. [Google Scholar] [CrossRef]
  303. Gu, Y.; Cai, R.; Zhang, C.; Xue, Y.; Pan, Y.; Wang, J.; Zhang, Z. miR-132-3p boosts caveolae-mediated transcellular transport in glioma endothelial cells by targeting PTEN/PI3K/PKB/Src/Cav-1 signaling pathway. FASEB J. 2019, 33, 441–454. [Google Scholar] [CrossRef] [PubMed]
  304. Wilhelm, I.; Nyúl-Tóth, Á.; Suciu, M.; Hermenean, A.; Krizbai, I.A. Heterogeneity of the blood-brain barrier. Tissue Barriers 2016, 4, e1143544. [Google Scholar] [CrossRef] [PubMed]
  305. Liebner, S.; Dijkhuizen, R.M.; Reiss, Y.; Plate, K.H.; Agalliu, D.; Constantin, G. Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol. 2018, 135, 311–336. [Google Scholar] [CrossRef] [PubMed]
  306. Taddei, A.; Giampietro, C.; Conti, A.; Orsenigo, F.; Breviario, F.; Pirazzoli, V.; Potente, M.; Daly, C.; Dimmeler, S.; Dejana, E. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat. Cell Biol. 2008, 10, 923–934. [Google Scholar] [CrossRef] [PubMed]
  307. Tran, K.A.; Zhang, X.; Predescu, D.; Huang, X.; Machado, R.F.; Göthert, J.R.; Malik, A.B.; Valyi-Nagy, T.; Zhao, Y.Y. Endothelial β-Catenin Signaling Is Required for Maintaining Adult Blood-Brain Barrier Integrity and Central Nervous System Homeostasis. Circulation 2016, 133, 177–186. [Google Scholar] [CrossRef]
  308. Stenman, J.M.; Rajagopal, J.; Carroll, T.J.; Ishibashi, M.; McMahon, J.; McMahon, A.P. Canonical Wnt signaling regulates organ-specific assembly and differentiation of CNS vasculature. Science 2008, 322, 1247–1250. [Google Scholar] [CrossRef]
  309. Dormanns, K.; Brown, R.G.; David, T. The role of nitric oxide in neurovascular coupling. J. Theor. Biol. 2016, 394, 1–17. [Google Scholar] [CrossRef]
  310. Cheng, S.; Xia, I.F.; Wanner, R.; Abello, J.; Stratman, A.N.; Nicoli, S. Hemodynamics regulate spatiotemporal artery muscularization in the developing circle of Willis. Elife 2024, 13, RP94094. [Google Scholar] [CrossRef]
  311. Filosa, J.A.; Bonev, A.D.; Straub, S.V.; Meredith, A.L.; Wilkerson, M.K.; Aldrich, R.W.; Nelson, M.T. Local potassium signaling couples neuronal activity to vasodilation in the brain. Nat. Neurosci. 2006, 9, 1397–1403. [Google Scholar] [CrossRef]
  312. Iadecola, C. The Neurovascular Unit Coming of Age: A Journey through Neurovascular Coupling in Health and Disease. Neuron 2017, 96, 17–42. [Google Scholar] [CrossRef]
  313. Kim, K.J.; Filosa, J.A. Advanced in vitro approach to study neurovascular coupling mechanisms in the brain microcirculation. J. Physiol. 2012, 590, 1757–1770. [Google Scholar] [CrossRef] [PubMed]
  314. Turovsky, E.A.; Braga, A.; Yu, Y.; Esteras, N.; Korsak, A.; Theparambil, S.M.; Hadjihambi, A.; Hosford, P.S.; Teschemacher, A.G.; Marina, N.; et al. Mechanosensory Signaling in Astrocytes. J. Neurosci. 2020, 40, 9364–9371. [Google Scholar] [CrossRef] [PubMed]
  315. Turovsky, E.A.; Plotnikov, E.Y.; Simakin, A.V.; Gudkov, S.V.; Varlamova, E.G. New magnetic iron nanoparticle doped with selenium nanoparticles and the mechanisms of their cytoprotective effect on cortical cells under ischemia-like conditions. Arch. Biochem. Biophys. 2025, 764, 110241. [Google Scholar] [CrossRef]
  316. Marina, N.; Turovsky, E.; Christie, I.N.; Hosford, P.S.; Hadjihambi, A.; Korsak, A.; Ang, R.; Mastitskaya, S.; Sheikhbahaei, S.; Theparambil, S.M.; et al. Brain metabolic sensing and metabolic signaling at the level of an astrocyte. Glia 2018, 66, 1185–1199. [Google Scholar] [CrossRef]
  317. Goetz, J.G.; Steed, E.; Ferreira, R.R.; Roth, S.; Ramspacher, C.; Boselli, F.; Charvin, G.; Liebling, M.; Wyart, C.; Schwab, Y.; et al. Endothelial cilia mediate low flow sensing during zebrafish vascular development. Cell Rep. 2014, 6, 799–808. [Google Scholar] [CrossRef]
  318. Eisa-Beygi, S.; Benslimane, F.M.; El-Rass, S.; Prabhudesai, S.; Abdelrasoul, M.K.A.; Simpson, P.M.; Yalcin, H.C.; Burrows, P.E.; Ramchandran, R. Characterization of Endothelial Cilia Distribution During Cerebral-Vascular Development in Zebrafish (Danio rerio). Arterioscler. Thromb. Vasc. Biol. 2018, 38, 2806–2818. [Google Scholar] [CrossRef]
  319. Pollock, L.M.; Perkins, B.; Anand-Apte, B. Primary cilia are present on endothelial cells of the hyaloid vasculature but are not required for the development of the blood-retinal barrier. PLoS ONE 2020, 15, e0225351. [Google Scholar] [CrossRef]
  320. Kallakuri, S.; Yu, J.A.; Li, J.; Li, Y.; Weinstein, B.M.; Nicoli, S.; Sun, Z. Endothelial cilia are essential for developmental vascular integrity in zebrafish. J. Am. Soc. Nephrol. 2015, 26, 864–875. [Google Scholar] [CrossRef]
  321. Chen, X.; Gays, D.; Milia, C.; Santoro, M.M. Cilia Control Vascular Mural Cell Recruitment in Vertebrates. Cell Rep. 2017, 18, 1033–1047. [Google Scholar] [CrossRef]
  322. Harraz, O.F.; Klug, N.R.; Senatore, A.J.; Hill-Eubanks, D.C.; Nelson, M.T. Piezo1 Is a Mechanosensor Channel in Central Nervous System Capillaries. Circ. Res. 2022, 130, 1531–1546. [Google Scholar] [CrossRef]
  323. Zi, H.; Peng, X.; Cao, J.; Xie, T.; Liu, T.; Li, H.; Bu, J.; Du, J.; Li, J. Piezo1-dependent regulation of pericyte proliferation by blood flow during brain vascular development. Cell Rep. 2024, 43, 113652. [Google Scholar] [CrossRef] [PubMed]
  324. Zhao, Z.; Zlokovic, B.V. Endothelial Tip Cell Finds Its Way with Piezo1. Neuron 2020, 108, 5–7. [Google Scholar] [CrossRef] [PubMed]
  325. Liu, T.T.; Du, X.F.; Zhang, B.B.; Zi, H.X.; Yan, Y.; Yin, J.A.; Hou, H.; Gu, S.Y.; Chen, Q.; Du, J.L. Piezo1-Mediated Ca2+ Activities Regulate Brain Vascular Pathfinding during Development. Neuron 2020, 108, 180–192.e5. [Google Scholar] [CrossRef] [PubMed]
  326. Longden, T.A.; Mughal, A.; Hennig, G.W.; Harraz, O.F.; Shui, B.; Lee, F.K.; Lee, J.C.; Reining, S.; Kotlikoff, M.I.; König, G.M.; et al. Local IP3 receptor-mediated Ca2+ signals compound to direct blood flow in brain capillaries. Sci. Adv. 2021, 7, eabh0101. [Google Scholar] [CrossRef]
  327. Choi, D.; Park, E.; Choi, J.; Lu, R.; Yu, J.S.; Kim, C.; Zhao, L.; Yu, J.; Nakashima, B.; Lee, S.; et al. Piezo1 regulates meningeal lymphatic vessel drainage and alleviates excessive CSF accumulation. Nat. Neurosci. 2024, 27, 913–926. [Google Scholar] [CrossRef]
  328. Guo, T.; Chen, G.; Yang, L.; Deng, J.; Pan, Y. Piezo1 inhibitor isoquercitrin rescues neural impairment mediated by NLRP3 after intracerebral hemorrhage. Exp. Neurol. 2024, 379, 114852. [Google Scholar] [CrossRef]
  329. Qi, M.; Liu, R.; Zhang, F.; Yao, Z.; Zhou, M.L.; Jiang, X.; Ling, S. Roles of mechanosensitive ion channel PIEZO1 in the pathogenesis of brain injury after experimental intracerebral hemorrhage. Neuropharmacology 2024, 251, 109896. [Google Scholar] [CrossRef]
  330. Li, Y.; Baylie, R.L.; Tavares, M.J.; Brayden, J.E. TRPM4 channels couple purinergic receptor mechanoactivation and myogenic tone development in cerebral parenchymal arterioles. J. Cereb. Blood Flow Metab. 2014, 34, 1706–1714. [Google Scholar] [CrossRef]
  331. Toth, L.; Czigler, A.; Szarka, N.; Toth, P. The role of transient receptor potential channels in cerebral myogenic autoregulation in hypertension and aging. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H159–H161. [Google Scholar] [CrossRef]
  332. Alvarado, M.G.; Thakore, P.; Earley, S. Transient Receptor Potential Channel Ankyrin 1: A Unique Regulator of Vascular Function. Cells 2021, 10, 1167. [Google Scholar] [CrossRef]
  333. Luo, H.; Rossi, E.; Saubamea, B.; Chasseigneaux, S.; Cochois, V.; Choublier, N.; Smirnova, M.; Glacial, F.; Perrière, N.; Bourdoulous, S.; et al. Cannabidiol Increases Proliferation, Migration, Tubulogenesis, and Integrity of Human Brain Endothelial Cells through TRPV2 Activation. Mol. Pharm. 2019, 16, 1312–1326. [Google Scholar] [CrossRef] [PubMed]
  334. Ramos, H.; Augustine, J.; Karan, B.M.; Hernández, C.; Stitt, A.W.; Curtis, T.M.; Simó, R. Sitagliptin eye drops prevent the impairment of retinal neurovascular unit in the new Trpv2+/− rat model. J. Neuroinflammation 2024, 21, 312. [Google Scholar] [CrossRef] [PubMed]
  335. Zhang, L.; Papadopoulos, P.; Hamel, E. Endothelial TRPV4 channels mediate dilation of cerebral arteries: Impairment and recovery in cerebrovascular pathologies related to Alzheimer’s disease. Br. J. Pharmacol. 2013, 170, 661–670. [Google Scholar] [CrossRef]
  336. Diaz-Otero, J.M.; Yen, T.C.; Ahmad, A.; Laimon-Thomson, E.; Abolibdeh, B.; Kelly, K.; Lewis, M.T.; Wiseman, R.W.; Jackson, W.F.; Dorrance, A.M. Transient receptor potential vanilloid 4 channels are important regulators of parenchymal arteriole dilation and cognitive function. Microcirculation 2019, 26, e12535. [Google Scholar] [CrossRef] [PubMed]
  337. Hatano, N.; Suzuki, H.; Itoh, Y.; Muraki, K. TRPV4 partially participates in proliferation of human brain capillary endothelial cells. Life Sci. 2013, 92, 317–324. [Google Scholar] [CrossRef]
  338. Chen, C.K.; Hsu, P.Y.; Wang, T.M.; Miao, Z.F.; Lin, R.T.; Juo, S.H. TRPV4 Activation Contributes Functional Recovery from Ischemic Stroke via Angiogenesis and Neurogenesis. Mol. Neurobiol. 2018, 55, 4127–4135. [Google Scholar] [CrossRef]
  339. Hansen, C.E.; Kamermans, A.; Mol, K.; Berve, K.; Rodriguez-Mogeda, C.; Fung, W.K.; van Het Hof, B.; Fontijn, R.D.; van der Pol, S.M.A.; Michalick, L.; et al. Inflammation-induced TRPV4 channels exacerbate blood-brain barrier dysfunction in multiple sclerosis. J. Neuroinflammation 2024, 21, 72. [Google Scholar] [CrossRef]
  340. Hakimizadeh, E.; Shamsizadeh, A.; Roohbakhsh, A.; Arababadi, M.K.; Hajizadeh, M.R.; Shariati, M.; Fatemi, I.; Moghadam-Ahmadi, A.; Bazmandegan, G.; Rezazadeh, H.; et al. TRPV1 receptor-mediated expression of Toll-like receptors 2 and 4 following permanent middle cerebral artery occlusion in rats. Iran J. Basic Med. Sci. 2017, 20, 863–869. [Google Scholar] [CrossRef]
  341. Miyanohara, J.; Shirakawa, H.; Sanpei, K.; Nakagawa, T.; Kaneko, S. A pathophysiological role of TRPV1 in ischemic injury after transient focal cerebral ischemia in mice. Biochem. Biophys. Res. Commun. 2015, 467, 478–483. [Google Scholar] [CrossRef]
  342. Al Tabosh, T.; Al Tarrass, M.; Tourvieilhe, L.; Guilhem, A.; Dupuis-Girod, S.; Bailly, S. Hereditary hemorrhagic telangiectasia: From signaling insights to therapeutic advances. J. Clin. Investig. 2024, 134, e176379. [Google Scholar] [CrossRef]
  343. Scimone, C.; Donato, L.; Alibrandi, S.; Conti, A.; Bortolotti, C.; Germanò, A.; Alafaci, C.; Vinci, S.L.; D’Angelo, R.; Sidoti, A. Methylome analysis of endothelial cells suggests new insights on sporadic brain arteriovenous malformation. Heliyon 2024, 10, e35126. [Google Scholar] [CrossRef] [PubMed]
  344. Scimone, C.; Granata, F.; Longo, M.; Mormina, E.; Turiaco, C.; Caragliano, A.A.; Donato, L.; Sidoti, A.; D’Angelo, R. Germline Mutation Enrichment in Pathways Controlling Endothelial Cell Homeostasis in Patients with Brain Arteriovenous Malformation: Implication for Molecular Diagnosis. Int. J. Mol. Sci. 2020, 21, 4321. [Google Scholar] [CrossRef] [PubMed]
  345. Scimone, C.; Donato, L.; Alafaci, C.; Granata, F.; Rinaldi, C.; Longo, M.; D’Angelo, R.; Sidoti, A. High-Throughput Sequencing to Detect Novel Likely Gene-Disrupting Variants in Pathogenesis of Sporadic Brain Arteriovenous Malformations. Front. Genet. 2020, 11, 46. [Google Scholar] [CrossRef]
  346. Park, H.; Furtado, J.; Poulet, M.; Chung, M.; Yun, S.; Lee, S.; Sessa, W.C.; Franco, C.A.; Schwartz, M.A.; Eichmann, A. Defective Flow-Migration Coupling Causes Arteriovenous Malformations in Hereditary Hemorrhagic Telangiectasia. Circulation 2021, 144, 805–822. [Google Scholar] [CrossRef]
  347. Baeyens, N.; Larrivée, B.; Ola, R.; Hayward-Piatkowskyi, B.; Dubrac, A.; Huang, B.; Ross, T.D.; Coon, B.G.; Min, E.; Tsarfati, M.; et al. Defective fluid shear stress mechanotransduction mediates hereditary hemorrhagic telangiectasia. J. Cell Biol. 2016, 214, 807–816. [Google Scholar] [CrossRef]
  348. Peacock, H.M.; Tabibian, A.; Criem, N.; Caolo, V.; Hamard, L.; Deryckere, A.; Haefliger, J.A.; Kwak, B.R.; Zwijsen, A.; Jones, E.A.V. Impaired SMAD1/5 Mechanotransduction and Cx37 (Connexin37) Expression Enable Pathological Vessel Enlargement and Shunting. Arterioscler. Thromb. Vasc. Biol. 2020, 40, e87–e104. [Google Scholar] [CrossRef]
  349. Li, W.; Quigley, K. Bone morphogenetic protein signalling in pulmonary arterial hypertension: Revisiting the BMPRII connection. Biochem. Soc. Trans. 2024, 52, 1515–1528. [Google Scholar] [CrossRef]
  350. Hiepen, C.; Jatzlau, J.; Hildebrandt, S.; Kampfrath, B.; Goktas, M.; Murgai, A.; Cuellar Camacho, J.L.; Haag, R.; Ruppert, C.; Sengle, G.; et al. BMPR2 acts as a gatekeeper to protect endothelial cells from increased TGFβ responses and altered cell mechanics. PLoS Biol. 2019, 17, e3000557. [Google Scholar] [CrossRef]
  351. Zhong, A.; Mirzaei, Z.; Simmons, C.A. The Roles of Matrix Stiffness and ß-Catenin Signaling in Endothelial-to-Mesenchymal Transition of Aortic Valve Endothelial Cells. Cardiovasc. Eng. Technol. 2018, 9, 158–167. [Google Scholar] [CrossRef]
  352. Ragazzini, S.; Scocozza, F.; Bernava, G.; Auricchio, F.; Colombo, G.I.; Barbuto, M.; Conti, M.; Pesce, M.; Garoffolo, G. Mechanosensor YAP cooperates with TGF-β1 signaling to promote myofibroblast activation and matrix stiffening in a 3D model of human cardiac fibrosis. Acta Biomater. 2022, 152, 300–312. [Google Scholar] [CrossRef]
  353. Rolle, I.G.; Crivellari, I.; Zanello, A.; Mazzega, E.; Dalla, E.; Bulfoni, M.; Avolio, E.; Battistella, A.; Lazzarino, M.; Cellot, A.; et al. Heart failure impairs the mechanotransduction properties of human cardiac pericytes. J. Mol. Cell. Cardiol. 2021, 151, 15–30. [Google Scholar] [CrossRef] [PubMed]
  354. Zhao, Y.Q.; Deng, X.W.; Xu, G.Q.; Lin, J.; Lu, H.Z.; Chen, J. Mechanical homeostasis imbalance in hepatic stellate cells activation and hepatic fibrosis. Front. Mol. Biosci. 2023, 10, 1183808. [Google Scholar] [CrossRef]
Figure 1. Mechanical signals from the ECM to the nucleus. Extracellular matrix mechanical cues are captured on cell surface by integrins and transmitted to cytoskeletal actin by the focal adhesion proteins. Likewise, tension from adjacent cell is transmitted by adherens junctions. Actin directly continues with nesprin 1/2, on nuclear envelope. By the SUN proteins, actin mechanical remodelling is trasmitted by the nesprins to the nuclear lamins, triggering chromatin remodelling and modification of the epigenetic pattern. ECM: extracellular matrix; FAK: focal adhesion kinase. Image created by the BioRender tool (https://www.biorender.com/).
Figure 1. Mechanical signals from the ECM to the nucleus. Extracellular matrix mechanical cues are captured on cell surface by integrins and transmitted to cytoskeletal actin by the focal adhesion proteins. Likewise, tension from adjacent cell is transmitted by adherens junctions. Actin directly continues with nesprin 1/2, on nuclear envelope. By the SUN proteins, actin mechanical remodelling is trasmitted by the nesprins to the nuclear lamins, triggering chromatin remodelling and modification of the epigenetic pattern. ECM: extracellular matrix; FAK: focal adhesion kinase. Image created by the BioRender tool (https://www.biorender.com/).
Biology 14 00346 g001
Figure 2. Mammalian TRP channels. The six mammal TRP channel classes include TRPC, TRPM, TRPA, TRPML, TRPP, TRPV. (a) TRPC channels show 4 ankyrin domains at the N-terminus that are recognized by the activated phospholipase C, upon stimulation. Downstream, intracellular calcium is mobilized. (b) TRPM channels exhibit 4 melastatin homology domains at the N-terminus; cation gating upon activation results in regulation of gene expression. (c) The TRPA1 channel has 16 ankyrin repeats at the N-terminus and contributes to calcium release from the endoplasmic reticulum. (d) The TRPML channels are expressed on endosomal and lysosomal membranes, forming either heterodimers (TRPML1/2) or homodimers (TRPML2/2). (e) The TRPP, also known as polycystins, are the most important mechanosensors of the primary cilium. (f) The TRPV channels are divided into two subclasses, the calcium-selective TRPV5/6 activate the MAPK-p38 cascade, regulating ECM remodeling and cell adhesion; the cation-aspecific TRPV1/2/3/4 activate the phospholipase cascades. ANK: ankyrin; CC: coiled-coil; CTD: carboxyl-terminal domain; DAG: diacyl-glycerol; ER: endoplasmic reticulum; IP3: inositol trisphosphate; MAPK: MAP-kinase; MHR: melastatin homology domains; eNOS: nitric oxide synthase; PC: polycystin; PL: phospholipase; RNS: reactive nitrogenous species; ROS: reactive oxygen species. Image created by the BioRender tool.
Figure 2. Mammalian TRP channels. The six mammal TRP channel classes include TRPC, TRPM, TRPA, TRPML, TRPP, TRPV. (a) TRPC channels show 4 ankyrin domains at the N-terminus that are recognized by the activated phospholipase C, upon stimulation. Downstream, intracellular calcium is mobilized. (b) TRPM channels exhibit 4 melastatin homology domains at the N-terminus; cation gating upon activation results in regulation of gene expression. (c) The TRPA1 channel has 16 ankyrin repeats at the N-terminus and contributes to calcium release from the endoplasmic reticulum. (d) The TRPML channels are expressed on endosomal and lysosomal membranes, forming either heterodimers (TRPML1/2) or homodimers (TRPML2/2). (e) The TRPP, also known as polycystins, are the most important mechanosensors of the primary cilium. (f) The TRPV channels are divided into two subclasses, the calcium-selective TRPV5/6 activate the MAPK-p38 cascade, regulating ECM remodeling and cell adhesion; the cation-aspecific TRPV1/2/3/4 activate the phospholipase cascades. ANK: ankyrin; CC: coiled-coil; CTD: carboxyl-terminal domain; DAG: diacyl-glycerol; ER: endoplasmic reticulum; IP3: inositol trisphosphate; MAPK: MAP-kinase; MHR: melastatin homology domains; eNOS: nitric oxide synthase; PC: polycystin; PL: phospholipase; RNS: reactive nitrogenous species; ROS: reactive oxygen species. Image created by the BioRender tool.
Biology 14 00346 g002
Figure 3. PIEZO channels. (a) PIEZO1 activation results in intracellular calcium influx; within the cell, protein kinase C can be activated. Downstream responses depend on stimulus properties and cell type. (b) PIEZO2 activation results in cation gating and cell depolarization. PKC: protein-kinase C. Image created by the BioRender tool.
Figure 3. PIEZO channels. (a) PIEZO1 activation results in intracellular calcium influx; within the cell, protein kinase C can be activated. Downstream responses depend on stimulus properties and cell type. (b) PIEZO2 activation results in cation gating and cell depolarization. PKC: protein-kinase C. Image created by the BioRender tool.
Biology 14 00346 g003
Figure 4. Mechanical regulation during embryo development. Extracellular matrix stiffness and cell density lead migration of neural crest cells; in particular, GPCRs and PIEZO1 drive mechanotransduction. Neural tube closure, instead, is driven by the β-actin gradient, increasing from dorsal to ventral cells. Likewise, differentiation of the cells of the ventral notochord region is driven by higher rigidity. ECM: extracellular matrix; GPCR: G-protein coupled receptor; PLC: phospholipase C. Image created by the BioRender tool.
Figure 4. Mechanical regulation during embryo development. Extracellular matrix stiffness and cell density lead migration of neural crest cells; in particular, GPCRs and PIEZO1 drive mechanotransduction. Neural tube closure, instead, is driven by the β-actin gradient, increasing from dorsal to ventral cells. Likewise, differentiation of the cells of the ventral notochord region is driven by higher rigidity. ECM: extracellular matrix; GPCR: G-protein coupled receptor; PLC: phospholipase C. Image created by the BioRender tool.
Biology 14 00346 g004
Figure 5. Mechanical regulation of vessel development. Vascular development can be divided into three moments: vasculogenesis, angiogenesis and specification. During vasculogenesis (a), mechanical stimuli generated by early blood flow act on progenitor endothelial cells and are mainly transduced by the TRPV4/PIEZO1 mechanosensitive ion channels, driving capillary plexus formation. During sprouting angiogenesis (b) both external and intracellular cues control endothelial tip cell protrusion. During late angiogenesis, mechanical properties of ECM surrounding mural cells control the early stages of arteriovenous differentiation. Finally, during specification (c), EC acquire their final identity as lymphatic ECs by expressing Prox1; likewise, blood flow velocity and direction drive vessel caliber during pruning. Finally, vascular regression contributes to the formation of the final vascular network. EC: endothelial cell; ECM: extracellular matrix; ETC: endothelial tip cell. Image created by the BioRender tool.
Figure 5. Mechanical regulation of vessel development. Vascular development can be divided into three moments: vasculogenesis, angiogenesis and specification. During vasculogenesis (a), mechanical stimuli generated by early blood flow act on progenitor endothelial cells and are mainly transduced by the TRPV4/PIEZO1 mechanosensitive ion channels, driving capillary plexus formation. During sprouting angiogenesis (b) both external and intracellular cues control endothelial tip cell protrusion. During late angiogenesis, mechanical properties of ECM surrounding mural cells control the early stages of arteriovenous differentiation. Finally, during specification (c), EC acquire their final identity as lymphatic ECs by expressing Prox1; likewise, blood flow velocity and direction drive vessel caliber during pruning. Finally, vascular regression contributes to the formation of the final vascular network. EC: endothelial cell; ECM: extracellular matrix; ETC: endothelial tip cell. Image created by the BioRender tool.
Biology 14 00346 g005
Figure 6. Mechanical signals in tip/stalk phenotype acquisition. At the angiogenetic front, during the angiogenic phase, EC can differentiate into either the stalk or tip phenotype. Tip cells exhibit by a protrusive behavior, driven by the BMPR2, activated by blood flow. Likewise, fibrillin binding to the integrin-syndecan complex directs differentiation toward the tip phenotype. In stalk cells, instead, the BMP9/Alk9 interaction promotes the β-IV-spectrin expression, responsible for VEGFR2 internalization. BMP: bone morphogenetic protein; BMPR2: bone morphogenetic protein receptor 2; FBN: fibrillin; VEGFR2: vascular endothelial growth factor receptor 2. Image created by the BioRender tool.
Figure 6. Mechanical signals in tip/stalk phenotype acquisition. At the angiogenetic front, during the angiogenic phase, EC can differentiate into either the stalk or tip phenotype. Tip cells exhibit by a protrusive behavior, driven by the BMPR2, activated by blood flow. Likewise, fibrillin binding to the integrin-syndecan complex directs differentiation toward the tip phenotype. In stalk cells, instead, the BMP9/Alk9 interaction promotes the β-IV-spectrin expression, responsible for VEGFR2 internalization. BMP: bone morphogenetic protein; BMPR2: bone morphogenetic protein receptor 2; FBN: fibrillin; VEGFR2: vascular endothelial growth factor receptor 2. Image created by the BioRender tool.
Biology 14 00346 g006
Figure 7. Mechanics of the neurovascular development. During early embryo development, blood flow drives endothelial cell (EC) adhesion by enhancing adherens junction-mediated claudin 5 expression. Likewise, PIEZO1 responds to flow by stabilizing adherens junctions between endothelial cells. The Notch1-Jag1-Notch3 signals encourages EC/pericyte adhesion. During neurovascular coupling, not only chemical but also hemodynamic stimuli activate both neurons and astrocytes. Signals are transmitted to ECs by the pericytes. In ECs, a vasomotor response in generated and transmitted to pericyte, regulating vascular tone and blood pressure. Image created by the BioRender tool.
Figure 7. Mechanics of the neurovascular development. During early embryo development, blood flow drives endothelial cell (EC) adhesion by enhancing adherens junction-mediated claudin 5 expression. Likewise, PIEZO1 responds to flow by stabilizing adherens junctions between endothelial cells. The Notch1-Jag1-Notch3 signals encourages EC/pericyte adhesion. During neurovascular coupling, not only chemical but also hemodynamic stimuli activate both neurons and astrocytes. Signals are transmitted to ECs by the pericytes. In ECs, a vasomotor response in generated and transmitted to pericyte, regulating vascular tone and blood pressure. Image created by the BioRender tool.
Biology 14 00346 g007
Table 1. Mechanosensitive ion channels in mammalian cells. The table lists the main classes of proteins acting as MCs in mammalian cells. For each family (Column I), the activating stimuli (Column II), the intracellular effector (Column III) and the biological response within the cell (Column IV) are listed. References are indicated in the reference list. BK: big potassium; CaM: calmodulin; ENaC: epithelial sodium channel; GPCR: G-protein coupled receptor; NO: nitric oxide; TREK: TWIK-Related K+; TRP: transient receptor potential.
Table 1. Mechanosensitive ion channels in mammalian cells. The table lists the main classes of proteins acting as MCs in mammalian cells. For each family (Column I), the activating stimuli (Column II), the intracellular effector (Column III) and the biological response within the cell (Column IV) are listed. References are indicated in the reference list. BK: big potassium; CaM: calmodulin; ENaC: epithelial sodium channel; GPCR: G-protein coupled receptor; NO: nitric oxide; TREK: TWIK-Related K+; TRP: transient receptor potential.
MechanosensorStimulusDownstream EffectorCell ResponseReference
ECM-integrin-cytoskeleton proteinsStiffness, tension, KLF2/4, YAP/TAZDifferentiation, apoptosis, proliferation[52]
ENaCPressure, shear forcesNONeurosensation, blood pressure regulation[53,54]
TREKMembrane tensionAKAP150, β-COP, Mtap2, sortilinK+ flow regulation[55,56]
BKMembrane electric potentialK+Cell hyperpolarization, smooth muscle tone[57]
GPCRsShear stress, mechanical stretchHeterogeneous Cell adhesion[58]
TRPCOxidative damageCalcineurin Memory, LTP, pain[59,60]
TRPMOxidative stress, inflammation, temperatureHeterogeneousHormone release, apoptosis [61,62]
TRPVMechanical and osmolar stimuliPhosphatidyl-inositol cascade ECM remodelling[63]
TRPAMechanical stress Ca2+Nociception, temperature sensation[64]
TRPMLMembrane tubulation, pHCaM, calcineurinAutophagy [65]
TRPPStiffness, cell tension, shear stressCa2+Tubule morphogenesis[66]
PIEZO1Shear stress, stiffnessKLF2/4, YAP/TAZVessel morphogenesis, bone repair[67]
PIEZO2Touch, shear forces, stretchS1P, ERKSensory neuron depolarization[68]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alibrandi, S.; Rinaldi, C.; Vinci, S.L.; Conti, A.; Donato, L.; Scimone, C.; Sidoti, A.; D’Angelo, R. Mechanotransduction in Development: A Focus on Angiogenesis. Biology 2025, 14, 346. https://doi.org/10.3390/biology14040346

AMA Style

Alibrandi S, Rinaldi C, Vinci SL, Conti A, Donato L, Scimone C, Sidoti A, D’Angelo R. Mechanotransduction in Development: A Focus on Angiogenesis. Biology. 2025; 14(4):346. https://doi.org/10.3390/biology14040346

Chicago/Turabian Style

Alibrandi, Simona, Carmela Rinaldi, Sergio Lucio Vinci, Alfredo Conti, Luigi Donato, Concetta Scimone, Antonina Sidoti, and Rosalia D’Angelo. 2025. "Mechanotransduction in Development: A Focus on Angiogenesis" Biology 14, no. 4: 346. https://doi.org/10.3390/biology14040346

APA Style

Alibrandi, S., Rinaldi, C., Vinci, S. L., Conti, A., Donato, L., Scimone, C., Sidoti, A., & D’Angelo, R. (2025). Mechanotransduction in Development: A Focus on Angiogenesis. Biology, 14(4), 346. https://doi.org/10.3390/biology14040346

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop