Previous Article in Journal
Unraveling the Complexities of Hypersensitivity Pneumonitis with Autoimmune Features: A Retrospective Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

A Unified Map of Airway Interactions: Secretome and Mechanotransduction Loops from Development to Disease

1
Open University System, Graduate School Department, Polytechnic University of the Philippines, A. Mabini Campus, Anonas Street, Sta. Mesa, Manila 1016, Philippines
2
Department of Chemical Engineering, Technological Institute of the Philippines, Quiapo, Manila 1001, Philippines
3
Department of Biomedical Engineering, Batangas State University—Alangilan Campus, Golden Country Homes, Brgy. Alangilan, Batangas City 4200, Philippines
*
Author to whom correspondence should be addressed.
Adv. Respir. Med. 2025, 93(6), 51; https://doi.org/10.3390/arm93060051
Submission received: 3 September 2025 / Revised: 14 October 2025 / Accepted: 28 October 2025 / Published: 12 November 2025

Highlights

What are the main findings?
  • First closed-loop model mapping bidirectional secretome–mechanotransduction feedback in airways, where mechanical cues trigger cytokine release and vice versa through YAP/TAZ signalling.
  • A novel cellular specialization framework defining epithelial cells as environmental activators, smooth muscle as mechanical actuators, and chondrocytes as calcium-dependent regulators in airway homeostasis.
What is the implication of the main finding?
  • Enables targeted therapy of airway diseases by interrupting pathological feedback loops (e.g., YAP/TAZ-mediated feed-forward stiffness traps in asthma/COPD).
  • Provides a system-based framework for airway tissue engineering by incorporating mechanotransduction feedback loops essential for functional airway constructs.

Abstract

Human airways maintain homeostasis through intricate cellular interactomes combining secretome-mediated signalling and mechanotransduction feedback loops. This review presents the first unified map of bidirectional mechanobiology–secretome interactions between airway epithelial cells (AECs), smooth muscle cells (ASMCs), and chondrocytes. We unify a novel three-component regulatory architecture: epithelium functioning as environmental activators, smooth muscle as mechanical actuators, and cartilage as calcium-dependent regulators. Critical mechanotransduction pathways, particularly YAP/TAZ signalling and TRPV4 channels, directly couple matrix stiffness to cytokine release, creating a closed-loop feedback system. During development, ASM-driven FGF-10 signalling and peristaltic contractions orchestrate cartilage formation and epithelial differentiation through mechanically guided morphogenesis. In disease states, these homeostatic circuits become pathologically dysregulated; asthma and COPD exhibit feed-forward stiffness traps where increased matrix rigidity triggers YAP/TAZ-mediated hypercontractility, perpetuating further remodelling. Aberrant mechanotransduction drives smooth muscle hyperplasia, cartilage degradation, and epithelial dysfunction through sustained inflammatory cascades. This system-level understanding of airway cellular networks provides mechanistic frameworks for targeted therapeutic interventions and tissue engineering strategies that incorporate essential mechanobiological signalling requirements.

1. Introduction: Gaps in Mapping Airway Interactomes

Despite their relative architectural simplicity, the human airways have a profoundly important role in the maintaining lung function. Upon inhalation, the airways are not only used as a pathway of air to the lungs but they also (1) improve the quality of the air by humidification and heating, (2) serve as a barrier to the external environment, (3) produce mucous to aid ejection of foreign particles and bacteria through coughing, and (4) provide structural support [1]. Each of the cell types present within the airways has a crucial role in one or more of these functions of the airways. Compromise of airway function results in a variety of pathological conditions including asthma, chronic obstructive pulmonary disease (COPD), Acute Respiratory Distress Syndrome (ARDS) and cystic fibrosis, which leads to increased mortality, morbidity, and reduces the quality of life of patients diagnosed with such diseases. These acute and chronic lung diseases currently have no single cure due to multiple contributions to the development of the disease; thus, the need to improve system-level understanding of the interactomes may elucidate how these diseases develop and could lead to potentially developing a multi-faceted treatment or cure.
Clinically, lung dysfunction manifests through two primary physiological patterns: obstructive and restrictive disorders [2]. Obstructive airway diseases, exemplified by asthma and chronic obstructive pulmonary disease (COPD), are characterised by airflow limitation on spirometry, with asthma showing bronchodilator reversibility while COPD demonstrates persistent fixed obstruction [2,3]. These conditions represent heterogeneous phenotypes that are distinguished by features such as bronchodilator response, eosinophil counts, and inflammatory patterns, which guide both management strategies and prognosis [3]. Restrictive lung diseases, in contrast, are characterised by reduced lung volumes and compliance, limiting the lung’s ability to expand during inspiration.
Efforts to resolve airway damage by trauma and disease has inspired researchers to engineer the organ itself, and over the past decade, various methodologies have been employed in vivo. Unfortunately, however, long-term success has thus far been elusive, with tissue engineered airway constructs mostly failing in animal models [4]. These failures have likely been caused by mechanical failure, inflammation, malacia, or stenosis [5,6,7,8,9]. Long-term viability of tissue-engineered airway constructs has been reported in dogs [10] and humans [11], although controversies have arisen over the scientific integrity of the trials [12]. The direct mechanisms of why these failures occurred are still unclear. Many tissue-engineered tracheas have been reported to be similar to native airways in terms of mechanical strength, glycosaminoglycan and hydroxyproline content, matured epithelial lining, and other histological aspects [13,14,15]. However, it has become clear that a major dissimilarity between these engineered airways and native airways is the absence of airway smooth muscle (ASM) in the engineered constructs. Molecular and mechanical cross-talk between cells is often fundamental to tissue growth and homeostasis [16,17,18]; therefore, the lack of ASM cells within the engineered airways could have profound effects on cellular functions that compromise airway integrity.
In this review, we aim to bring together published studies to unravel the complex interactions occurring between the cells comprising human airways and discuss their potential contribution to the maintenance of airway function.

Anatomic Scope of the Review

To establish clear anatomical and technical contexts for this review, we define key terms as they relate to cellular interactions and mechanotransduction in airway biology.
Airways in this review encompass the conducting structures from trachea to bronchi that serve as conduits for air to the lungs, emphasising their multifunctional role in humidification, heating, barrier protection, mucus production for particle ejection, and structural support. Our focus centres on the complex cellular architecture comprising airway epithelial cells (AECs), airway smooth muscle cells (ASMCs), chondrocytes, and mesenchymal stem cells (MSCs) from the central airways (trachea and bronchi) that collectively maintain airway homeostasis through coordinated cellular interactions.
For anatomic clarity, the cartilaginous support of the airways follows a precise anatomical gradient that is critical for understanding tissue engineering applications. The trachea is supported by 16–20 horseshoe-shaped (C-shaped) hyaline cartilage rings connected by annular ligaments anteriorly and laterally, with a posterior smooth muscle (trachealis) membrane [19]. This rigid hyaline cartilage serves as the major load-bearing component of the tracheal wall [20]. Moving distally, the main bronchi maintain substantial cartilaginous support, but the cartilage progressively decreases in both quantity and structural prominence through successive bronchial generations [20]. In the smaller bronchi, cartilage appears as irregular plates rather than complete rings, while bronchioles (airways < 1 mm diameter) completely lack cartilaginous support, relying instead on smooth muscle and elastic fibres for structural integrity [20]. This proximal-to-distal cartilage gradient directly influences the mechanical properties and engineering requirements for different airway segments.
Lungs are referenced as the ultimate destination for airway-conducted air, with particular emphasis on their inherently mechanical nature—constantly subjected to stretching and relaxation during breathing cycles. This review focuses on how lung mesenchymal stem cells contribute to airway homeostasis and how mechanical processes are critical for normal lung development and regeneration following surgical interventions.
Tissue engineering in this context refers to systematic efforts to resolve airway damage from trauma and disease through the creation of functional airway constructs that incorporate essential cellular components and mechanotransduction pathways. Our review addresses the current limitations where engineered constructs often fail due to mechanical issues, inflammation, malacia, or stenosis, with a critical gap being the absence of airway smooth muscle (ASM) in many engineered airways. We propose a systems-based framework for airway tissue engineering that incorporates bidirectional secretome–mechanotransduction feedback loops, utilising mesenchymal stem cells as global mediators capable of differentiation into specific airway cell types while contributing to immune regulation and regeneration.

2. Airway Cell Specialisation in a Feedback Loop Model

The human airway is composed of several different types of cells, including chondrocytes, airway smooth muscle cells (ASMC), airway epithelial cells (AEC), mesenchymal stromal cells (MSC), fibroblasts, neurons, adipocytes, and endothelial cells. This review acknowledges the heterogeneity of cell types within the population, specifically the AECs, which has subphenotypes, including ciliated cells, goblet and club cells, and basal cells. The AECs discussed therein is a collective term for all its subpopulations.
Single-cell RNA sequencing studies have revolutionised our understanding of airway cellular heterogeneity and intercellular communication networks. Recent comprehensive analyses have identified previously unknown epithelial cell subtypes, characterised their distinct secretory profiles, and mapped ligand–receptor interactions between airway cell populations [21,22]. These studies reveal that mechanical perturbations reshape epithelial transcriptional states, altering paracrine signalling to mesenchymal and immune cells [23]. In addition, critical recognition of cellular heterogeneity based on developmental origins is essential for understanding differential injury responses in airway mechanotransduction networks. Airway basal cells from distinct developmental lineages—such as ventral cartilage-associated versus dorsal smooth muscle-associated regions—maintain different transcriptional signatures and mechanosensitive pathway activation patterns that directly influence their capacity for self-renewal, differentiation, and secretome production following tissue damage [24]. This developmental origin-dependent heterogeneity extends to chondrocytes, smooth muscle cells, and epithelial subtypes, creating spatially distinct repair programmes where YAP/TAZ signalling, TRPV4 channel activation, and cytokine release patterns vary significantly based on cellular developmental history. For the purposes of this review, we will focus on interactions between general cell types of chondrocytes, ASMC, AEC, and MSC.
Chondrocytes are the resident cells of cartilage and produce abundant extracellular matrix (ECM) components made up of collagens, glycosaminoglycan, and proteoglycans to create a hyaline cartilage [25,26], which mechanically supports the overall structure of the airway. Lung MSCs are capable of differentiating into chondrocytes, ASMCs, or AECs to replace cells and maintain airway homeostasis [27]. ASMCs form together a muscular tissue with connective fibres attached to the cartilage, capable of contracting the airway luminal diameter, which could modulate air resistance as it travels in and out of the lungs [28]. AECs are located on the luminal side of the airways and provide a barrier to the external environment. These cells release mucous, which traps particles, pathogens, and other foreign components for ease of ejection through coughing or sneezing [29]. Central to airway defence is the mucociliary clearance system, a coordinated mechanism involving both structural and functional components of the airway epithelium [30,31]. This system relies on the synchronised action of ciliated epithelial cells, which generate directional fluid flow through coordinated ciliary beating, and secretory cells that produce the mucus layer containing antimicrobial peptides and surfactant proteins [31,32]. The mucociliary elevator represents a critical first-line defence mechanism, continuously transporting trapped particles, pathogens, and debris from the lower respiratory tract toward the pharynx for elimination [30].
As with all organs and tissues, interaction between each of these cells is likely to be fundamental for the maintenance of airway function and organ homeostasis. Understanding such interactions will be crucial if efforts to tissue engineering human airways are to be successful. Cell–cell interactions may occur via a combination of mechanisms, including secretion of a vast array of functional molecules which can act in a paracrine manner, and mechanobiological events where cell–cell interactions occur via transmission and detection of mechanical changes.

2.1. Dynamic Secretome: Mechanical Drivers and Reciprocal Rewiring

The integration of mechanical and biochemical signalling represents a fundamental principle underlying airway function and pathology. Mechanical forces, including cyclic stretch from breathing, matrix stiffness changes during remodelling, and compressive forces from bronchoconstriction, are not merely passive physical phenomena but active regulators of cellular behaviour [33]. These forces are transduced into biochemical signals through specialised mechanosensitive pathways, creating feedback loops where mechanical stimuli influence secretome composition, which in turn can modify tissue mechanical properties and cellular responses.
The dynamic secretome operates through sophisticated reciprocal rewiring mechanisms that enable bidirectional phenotype switching and cellular reprogramming in response to changing mechanical and chemical environments. This reciprocal rewiring represents a fundamental property of airway cell networks where cells can transition between different functional states through mechanotransduction-mediated feedback [34]. Epithelial cells demonstrate remarkable plasticity, undergoing partial epithelial–mesenchymal transition (EMT) in response to matrix stiffness and TGF-β signalling, while retaining the capacity to reverse these changes when mechanical and chemical cues shift toward homeostatic conditions [35]. Fibroblasts exhibit mechanomemory phenomena, where previous mechanical conditioning influences their transcriptional and contractile responses yet demonstrate reversibility when transferred from stiff to soft mechanical environments, indicating that activation states can be dynamically reprogrammed [36]. The reciprocal nature of this rewiring is exemplified by epithelial–mesenchymal communication networks where healthy epithelium suppresses fibroblast activation through BMP signalling, while activated fibroblasts can promote epithelial differentiation through IL-6 and FGF secretion, creating context-dependent cooperative or competitive loops [37,38]. Matrix viscoelasticity emerges as a critical regulator of cellular plasticity, with slower stress relaxation enhancing chromatin accessibility at pluripotency-associated elements and improving cellular reprogramming efficiency, suggesting that mechanical properties can induce profound epigenetic rewiring rather than merely transient phenotype shifts [39]. This reciprocal rewiring framework reveals that airway pathology involves not just dysregulated signalling, but fundamental alterations in cellular plasticity networks that become locked into pathological states through positive feedback loops involving matrix stiffening, pro-fibrotic secretion, and mechanotransduction pathway activation [40].
A plethora of bioactive components including growth factors and cytokines can be released by the airway cells as described in Table 1, with a summary of interactions in Figure 1. More detailed lists of the secretory profiles of chondrocytes [41,42,43,44,45] MSCs [42,46], ASMCs [47], and AECs [48] can be found elsewhere. Once released, many of these components are capable of autocrine and paracrine effects to induce either local cellular level changes or more wide-spread tissue-level changes in the airway to affect a vast array of signalling pathways that contribute to normal airway function and homeostasis.
Functional molecules released in the airways can be class clustered on their functionality. Mitogenic Growth Factors including Epidermal Growth Factor (EGF), Fibroblast Growth Factor (FGF) and Insulin Growth Factor (IGF) promote cellular proliferation [22,49,50]. Tissue remodelling Growth Factors that modify the expression of extracellular matrix components include Transforming Growth Factor β (TGF-β), Interleukin-1β (IL-1β), vascular epithelial Growth Factor (VEGF), and Connective tissue Growth Factor (CTGF). Each of these factors are involved in wound repair and/or matrix remodelling pathways for the maintenance of tissue homeostasis [51,52,53,54]. Cytokines such as Interleukins act as inflammatory mediators in response to an injurious or inflammatory stimuli or when chronic pathological conditions arise, and they can alter the phenotype and secretions of molecules of airway cells [21,55,56,57,58]. Most of these functional molecules are produced by the cells comprising the airways in either homeostatic and/or pathological processes (see Table 1) and can change the function and/or phenotype of other cells.
An analysis of Table 1 reveals that multiple cell types secrete identical signalling molecules, suggesting both functional redundancy and coordinated responses that ensure robust tissue-level communication [59]. For example, TGF-β is secreted by chondrocytes, ASMCs, and AECs, but in response to different stimuli (IL-1β for chondrocytes, matrix stiffness for ASMCs, and compression for AECs). This distributed secretion pattern ensures robust TGF-β signalling throughout the airway while allowing for stimulus-specific activation patterns that can be fine-tuned by local mechanical and chemical environments.
The mechanotransduction pathways underlying this coordinated secretion involve TRPV4-mediated calcium signalling and YAP/TAZ transcriptional regulation [60]. Recent evidence demonstrates that TRPV4 is necessary for matrix stiffness- and TGF-β1-induced responses across multiple cell types, suggesting a conserved mechanochemical coupling mechanism that enables coordinated secretome regulation (Sharma et al., 2019) [61]. This mechanistic conservation explains how different cell types can produce coordinated responses to mechanical stimuli while maintaining cell-type-specific secretion profiles.
Similarly, VEGF secretion by MSCs (hypoxia-stimulated), chondrocytes (hypoxia-stimulated), ASMCs (injury-stimulated), and AECs (hypoxia-stimulated) creates overlapping angiogenic signals that respond to diverse pathological conditions. The mechanochemical regulation of VEGF secretion has been directly demonstrated in MSCs, where matrix rigidity and cyclic compression increase VEGF secretion via YAP-dependent mechanisms [62]. This redundancy provides resilience against single-cell-type dysfunction while enabling coordinated vascular responses that are essential for tissue repair and remodelling.
The diversity of stimuli listed in Table 1 demonstrates how different cell types serve as specialised sensors for distinct environmental changes, with mechanotransduction pathways providing the molecular machinery for stimulus detection and response coordination. AECs respond to external stimuli (cigarette smoke, compression) through mechanosensitive pathways that couple physical forces to transcriptional programmes (Kilic et al., 2019) [5]. ASMCs respond to mechanical changes (matrix stiffness, stretch) through stiffness-mediated mechanosensation mechanisms that have been characterised on linear stiffness gradient systems [63].
Chondrocytes respond to both mechanical (fluid shear stress) and inflammatory signals (IL-1β) through TRPV4 and Piezo channels that mediate mechanosensing of the biomechanical microenvironment [64]. This dual sensitivity enables chondrocytes to integrate mechanical and inflammatory inputs, creating coordinated responses that address both structural and immunological aspects of tissue remodelling. MSCs respond primarily to tissue damage signals (hypoxia, inflammation) with their secretome profiles being dynamically regulated by mechanical cues through YAP-dependent mechanisms [65].
This distributed sensing network ensures comprehensive environmental monitoring and appropriate tissue-level responses. The specificity of stimulus-response relationships also suggests potential therapeutic windows where targeting specific mechanotransduction pathways could modulate secretion patterns without disrupting normal homeostatic functions [33].
Table 1. Secretion of functional molecules of airway cells and their effects.
Table 1. Secretion of functional molecules of airway cells and their effects.
Secretory SignalStimulantEffect on Airway CellsReferences
Secreting Cell: Mesenchymal Stromal Cell
Activin A Differentiation regulation[66,67]
Angiopoietin-1 Vascular stabilisation[68]
Angiopoietin-2Shear forcesAngiogenic remodelling[69]
Bone morphogenic protein-2 and 4Cyclic tensile strain, PGE2 [70,71,72]
Connective tissue Growth Factor Fibrotic remodelling[46,73,74,75]
Fibroblast Growth Factor-2Hypoxia, TNF-αProliferation effects[76,77]
Hepatocyte Growth FactorHypoxia, TNF-αEpithelial repair[76,77]
Insulin Growth Factor-1Hypoxia, TNF-αProliferation effects[76,77,78,79]
Interleukin-1 Inflammatory signalling[79]
Interleukin-6, 19, 23Cyclic tensile strain, DexamethasoneInflammatory modulation[80]
Interleukin-7 Lymphocyte support[81]
Interleukin-8 Neutrophil chemotaxis[80]
Interleukin-10, 19, 20 Anti-inflammatory effects
Osteoprotegerin [67]
Platelet-derived Growth Factor Proliferation effects, Remodelling signal[82]
Transforming Growth Factor-βHypoxia, TNF-αAirway cell contraction and ECM remodelling across cell types[67,77,83]
Vascular endothelial Growth FactorHypoxia, TNF-αAngiogenic/remodelling cues affecting multiple airway cells[67,76,77,78,79,84]
Secreting cell: Chondrocyte
AdrenomedullinHypoxiaVasodilation, anti-apoptotic[85,86]
Angiopoietin-like 4Hypoxia [87]
Angiopoietin-like 7Mechanical stressAngiogenic signalling[46,73]
Connective tissue Growth FactorTGF-β, mechanical stressInduces EMT in airway epithelial cells; supports perichondrium formation via fibroblast generation[46,73,74,75,88]
Chitinase 3-like 2Inflammatory cytokinesERK activation[46,73,89]
Epidermal Growth FactorEGFTriggers chondrocyte PGE2 release, which increases AEC proliferation[90,91]
Fibroblast Growth Factor-2Interleukin-1βMaintains epithelial integrity; supports barrier/homeostasis[92]
Interleukin-6Fluid shear stressPro-inflammatory activation[93]
Nitric OxideInterleukin-1Vasodilation[94]
Osteomodulin Regulates mineralization[46,73,95]
Prostaglandin E2 Increases proliferation of AECs (via paracrine loop).[90]
Transforming Growth Factor-α [91]
Vascular endothelial Growth Factor Angiogenic/remodelling cues affecting multiple airway cells[96]
Secreting cell: Airway Smooth Muscle Cell
AdrenomedullinIL-1β, TNF-αVasodilation, angiogenesis[97,98]
AmphiregulinTNF-α, IL-4In AECs: ↑VEGF, ↑PGE2, ↑COX-2, ↑CXCL8; modulates ASM contraction/proliferation[99,100]
Connective tissue Growth FactorInjury, TGF-βOverexpression promotes AEC senescence (pathologic); may drive EMT-like changes[101,102,103]
Fibroblast Growth Factor-2TNF-α, IL-1β MMitogenic across airway cells (general)[98,104]
Fibroblast Growth Factor-9Hypoxia, mechanical stressMitogenic across airway cells (general)[105,106]
Fibroblast Growth Factor-10Epithelial injuryPromotes epithelial repair[103,107]
Interleukin-6TNF-α, epithelial co-cultureInflammatory response activation[107,108,109]
Interleukin-8Interleukin-1β, TNF-α,
epithelial injury
Neutrophil chemotaxis[99,107,110]
Nerve Growth FactorInterleukin-1β,
inflammatory mediators
Neuronal sensitization[98,111]
Nitric OxideInflammatory cytokinesSmooth muscle relaxation[106,108]
Prostaglandin E2Mechanical stress, cytokinesBronchodilation, anti-inflammatory[99,106]
Transforming Growth Factor-αEGF receptor activationEpithelial proliferation[91,100]
Transforming Growth Factor-β1Mechanical injury, hypoxiaDrives ECM remodelling; promotes pathological changes across cell types (context-dependent)[104,112]
Vascular endothelial Growth FactorAngiotensin-2, Endothelin-1, TGF-β1, Bradykinin,IL-4, IL-5, IL-13, PGE2 Angiogenic/remodelling cues affecting multiple airway cells[113,114]
TGF-β1Mechanical stress, injuryECM remodelling; activates epithelial responses (context-dependent)[102,115]
Stem cell factorNeutrophil elastase,
Increased matrix stiffness
Mast cell activation[104,116]
Secreting cell: Airway Epithelial Cell
Adrenomedullin [117,118]
AmphiregulinCigarette smoke [119]
Angiopoietin [117]
Chitinase 3-like 1Viral dsRNA, chitinInduces IL-8 secretion[46,89,120,121,122]
Endothelin-1CompressionASMC proliferation, contraction[117,123,124]
Epidermal Growth Factor Induces chondrocyte PGE2 release → increases AEC proliferation (paracrine loop)[91,117]
Insulin Growth Factor-1 Mitogenic across airway cells (general)[117,125]
Interleukin-1βThrombin, Trypsin, TNF-αStimulates chondrocyte FGF-2 secretion to maintain epithelial integrity[29]
Interleukin-4, 10, 13, 22 Induces mucus hyperproduction; promotes ciliated differentiation[126]
Interleukin-6Thrombin, Trypsin [29,127]
Interleukin-8Thrombin, Trypsin, TNF-α [29,127]
Nitric Oxide Relaxes ASM by decreasing Ca2+ oscillations[128]
Platelet-derived Growth Factor (PDGF)Inflammatory cytokinesSmooth muscle proliferation[129]
Interleukin-11 [29]
Prostaglandin E2Thrombin, TrypsinRelaxes ASM (paracrine effect)[127]
Prostaglandin D2Allergen exposure, inflammationBronchoconstriction[130]
Transforming Growth Factor-β1, β2Thrombin, Trypsin, Hypoxia, AmphiregulinPromotes ECM remodelling; cross-talk with ASM and cartilage[29,99,117,131]
Tumour necrosis factor α [29]
Vascular endothelial Growth FactorThrombin, TrypsinAngiogenic/remodelling cues affecting multiple airway cells[99,127,131]

2.2. Mechanotransduction and ECM Feedback: YAP/TAZ Networks Unveiled

The lungs are an inherently mechanical organ, subjected to constant cycles of stretching and relaxation during breathing. An adult on average takes 12–20 breaths per minute during tidal breathing [132]. The cells comprising the airways are therefore adapted to this mechanical environment and in many cases can respond biologically to it in a process known as mechanobiology. In fact, mechanical processes are critical to normal lung development and regrowth of the lung following surgical resections [17]. This is supported by the emerging role of YAP (Yes-associated protein) and TAZ (transcriptional coactivator with PDZ-binding motif) transcription factors as mediators for mechanical stimuli. YAP/TAZ signalling has emerged as a central mechanotransduction pathway in airway cells, with recent studies demonstrating context-dependent roles in epithelial repair versus pathological remodelling. Sustained YAP/TAZ activation promotes aberrant alveolar epithelial cell differentiation and drives persistent fibrotic remodelling, while controlled activation supports regenerative processes [46,133].
YAP/TAZ are main effectors of the Hippo pathway which ultimately affect cell growth, proliferation, and inhibition of apoptosis [134]. Stretching the matrix in which cells are embedded can activate YAP/TAZ, which in turn deactivates contact inhibition and thereby activates proliferation [133]. Furthermore, it has been shown that stretch-mediated activation of TGF-β regulates macrophage function [135], suggesting that breathing may play a crucial role in maintaining immune homeostasis in the lung.
Current single-cell transcriptomics and mechanobiology studies have revealed that airway cells possess a sophisticated mechanosensing apparatus, including Piezo1 channels and YAP/TAZ signalling pathways, that directly couples physical stimuli to cellular responses and paracrine signalling programmes [48,73,117,136]. Piezo1 has been identified as a critical mechanosensor in airway smooth muscle cells, where single-cell hypertrophy promotes contractile function through Piezo1-mediated YAP autoregulation [136]. The RhoA/ROCK pathway functions as a key mechanotransduction cascade in airway remodelling, with enhanced activation observed in asthmatic airways leading to increased smooth muscle contractility and matrix deposition [44].
Dynamic nucleocytoplasmic shuttling of YAP/TAZ in response to tissue stretch is crucial for proper airway branching morphogenesis and alveolar cell differentiation [137]. During lung morphogenesis, nuclear YAP drives actomyosin-mediated tension via RhoA–ROCK signalling, a process required for normal bronchial tree formation [137]. Dysregulation of this YAP/TAZ mechanotransduction axis can lead to branching defects and aberrant cell differentiation, underscoring its importance in lung organogenesis [137].
Airway cells also sense and respond to extracellular matrix (ECM) stiffness via integrin–cytoskeletal pathways. On stiff substrates, YAP/TAZ translocate to the nucleus and actively drive gene transcription, whereas on soft matrices YAP/TAZ remain sequestered in the cytoplasm [33]. This stiffness-dependent YAP/TAZ activation can occur independently of the canonical Hippo kinases, instead relying on cytoskeletal tension; inhibiting Rho/ROCK-mediated actin polymerization prevents YAP/TAZ nuclear accumulation [33]. These findings demonstrate that changes in ECM rigidity are directly translated into altered cell proliferation and differentiation programmes in the lung through mechanotransduction.
Mechanosensitive ion channels further expand the lung’s ability to transduce physical forces. The TRPV4 cation channel, for example, is expressed in airway smooth muscle and epithelial cells and opens in response to mechanical stimuli such as stretch or osmotic swelling [138]. Activation of TRPV4 causes a Ca2+ influx that can trigger downstream signalling; in smooth muscle, TRPV4-mediated Ca2+ entry contributes to bronchoconstriction and airway remodelling, while in epithelial cells it can influence ciliary beating and cytokine release [138]. Thus, channels like TRPV4 provide a rapid link between mechanical perturbations and cellular responses in the airways.
Mechanical forces can even modulate immune function in the lung. Increasing matrix stiffness has been shown to induce macrophage polarisation toward a pro-fibrotic phenotype via YAP-dependent transcription [33]. Likewise, stiffness-induced YAP activation in the lung microenvironment can suppress T-cell proliferation through metabolic reprogramming [33]. These examples illustrate that an imbalanced mechanical environment can directly skew immune responses, linking mechanobiology to inflammation and tissue remodelling in the lung.

2.3. Secretome Mechanotransduction Feedback Signalling: Closed Loop Model and Control Points

There is a crosstalk between mechanobiology and secretory pathways in the lung. Mechanical signals can influence the secretion of functional molecules in the airway, which can then contribute to a positive feedback manner to cellular and tissue-level mechanical action such as contraction and relaxation. For example, mechanical signals including increased stiffness, stretching, and contraction can cause the activation and release of TGFb from epithelial cells and ASM cells [139,140,141]. Once activated, TGFb has the potential to influence the contraction of airway cells [142,143] as well as regulate remodelling processes [144,145,146].
Altered mechanical movements can then drive cells to secrete growth factors and cytokines and so perpetuate the loop. Figure 2 shows the proposed feedback loop on secretome–mechanotransduction pathways, where cellular-to-organ-level responses are mapped with corresponding secretome and mechanobiological actions. Evidence of the feedback mechanism can be observed in the early works of Fedan’s team, where it has been shown ex vivo that the presence of cartilage and epithelium affected the contraction of the ASM [147,148]. Removal or denudation of cartilage and epithelium lead to the altered sensitivity of airway smooth muscle.
Airway epithelial cells sense compressive or shear stress and activate mechanotransduction cascades that change gene expression and drive release of paracrine mediators relevant to airway tone and remodelling. Several recent studies show mechanical compression/stretch elicits Ca2+-dependent signalling and secretion of ATP, prostaglandins, and extracellular-matrix proteins/vesicles that can influence neighbouring cells and tissue state. Compression of primary human bronchial epithelium alone produces inflammatory, repair, and fibrotic transcriptional programmes that mirror asthmatic signatures, consistent with force-driven epithelial activation by bronchospasm [149]. Mechanical compression of differentiated human bronchial epithelial cells increases tenascin-C expression and secretion in extracellular vesicles, implicating epithelial mechano-response in ECM remodelling [150]. TRPV2 in primary bronchial epithelial cells mediates mechanically induced ATP release, demonstrating a channel-dependent epithelial nucleotide signal triggered by mechanical stress [151]. Acetylcholine stimulated prostaglandin E2 release from tracheal epithelium and thereby induced smooth muscle relaxation in rat tracheal rings, showing an epithelial-derived biochemical pathway that changes ASM contractility [152]. Recent reviews highlight store-operated Ca2+ entry and other Ca2+ pathways as central hubs by which epithelial mechanostimuli regulate mediator production and secretion [153].
Airway smooth muscle functions as the downstream effector that detects mechanical and biochemical inputs and converts them into contractile or relaxant responses; recent work describes specific mechanosensors and Ca2+ regulators in ASM. The cited studies identify stretch-responsive pathways, Piezo-mediated responses, and stiffness-sensing that alter ASM biomechanics and contractility. STIM1 mediates stretch-induced signalling in human ASM, upregulating mechanosensitive channels (Piezo1/2), Orai1, and inflammasome components and modifying Ca2+ responses relevant to contractility and remodelling [154]. Chemical activation of Piezo1 (Yoda1) in cultured ASMCs produces transient Ca2+ signals and long-term reductions in cell stiffness and traction force consistent with pro-relaxation biomechanical changes [155]. Human ASM cells show stiffness-dependent changes in cell size, α-SMA expression, YAP translocation, and other mechanotransduction readouts, linking ECM mechanics to ASM contractile phenotype [63]. Comprehensive reviews place TRP and Piezo families at the centre of lung mechanosensing, influencing ASM behaviour and organ-level responses [156].
Recent reviews and syntheses frame epithelial–ASM mechanochemical interactions and ECM mechanotransduction as integrated regulatory pathways that maintain (or disrupt) airway homeostasis; these works link cellular mechanosensing to tissue remodelling and organ function. The literature emphasises multiscale mechanotransduction and ECM feedback as drivers of pulmonary pathophysiology and as potential therapeutic targets. Mechanotransduction coupled to extracellular matrix composition and mechanics is highlighted as a key driver of lung pathologies and of drug responsiveness, connecting cellular mechanosensors to organ-level homeostasis and disease progression [157]. Reviews of mechanosensitive channels in lung health discuss how epithelial and ASM channels (TRP, Piezo) mediate force to signal transduction that coordinates mucus hydration, inflammation, remodelling, and contractile behaviour, supporting a systems-level regulatory role [156]. Experimental demonstrations that epithelial compression induces asthma-like signatures and releases paracrine mediators (ATP, PGE2, ECM/EVs) provide mechanistic nodes that feed into ASM and ECM responses implicated in sustained changes to airway function [149,150,151,152].
The importance of such cellular interactions has been demonstrated in vivo. Mouse mutants genetically manipulated to inhibit cartilage or smooth muscle formation in the developing airways have shown the co-dependence of cartilage and smooth muscle upon growth, the important role of the cartilage on the epithelium differentiation, and that the absence of either tissue resulted in malformation of the airway [158]. This study highlights the necessity of understanding the interplay between cell types in the airway, and so we shall then look closely at the cellular level interactions that drives these processes altogether.

3. Interactions of Airway Cells

3.1. Physiology: Epithelium as Activator Smooth Muscle as Actuator Cartilage as Regulator

The cellular niche of the airways provides pathways for the homeostasis of the surrounding tissues. Structurally, the lumen of the airway is covered by AECs that are supported by the cartilage and modulated by ASMCs [19]. In the physiological condition, it can be hypothesised that the AECs receive stimuli from the environment and provide cues for airway dynamics; cell- and tissue-level event cascades initiated by the secretome can drive multiple responses in the airways (Figure 2). Growth factors secreted by AECs [48,119], ASMCs [101,102,105], and chondrocytes [46,73,89,120] can act as mitogens across all cell types, thereby driving cellular proliferation [22,159,160], migration [161], and cell survival through protease-activated receptor-2–, AKT-, ERK-, and p38-MAPK-dependent pathways [22,162,163]. Several of the factors released are induced either by other growth factors or via mechanotransduction, prompting a cascade of events to maintain cellular homeostasis (see Table 1 for a list of factors and known stimulants).
Apart from cellular proliferation, the integrity of the airways is also dependent on the secretion of molecules and proteins for maintenance of the extracellular environment.
Recent studies have identified specific molecular pathways mediating epithelial–smooth muscle communication. Airway epithelial cells promote smooth muscle cell proliferation by activating the Wnt/β-catenin pathway, while semaphorin3E/PlexinD1 signalling represents a novel regulatory axis in COPD pathogenesis [139,141]. These interactions are dynamically regulated by mechanical forces and inflammatory stimuli.
Acting as the mechanical framework of the airways, chondrocytes produce mechanically resilient ECM components like collagen type II and proteoglycans that are necessary for the load-bearing capacity of the airway [25,26]. These components can be upregulated through (1) the secretome, by factors secreted by AECs [29,48,123,125], and (2) mechanotransduction, by the contraction/relaxation mechanism of ASMCs that relates to TGFb activation [164]. The latter mechanism might be explained by the fact that chondrocytes, when subjected to cyclic compressive stress, upregulate their ECM production [165,166]. This suggests that the mechanotransducive events produced by the ASMCs can influence chondrocyte functionality. In the remodelling phase, AECs secrete several cytokines that may aid chondrocytes in their growth phase, inducing release of matrix remodelling enzymes for tissue expansion [51,52,53]. Additionally, a paracrine feedback response of EGF from AECs to chondrocytes can trigger release of PGE2 [90], which increases the rate of proliferation of AECs through induction of c-Jun and three-phosphoinositide dependent protein kinase-1 (PDK1) pathways [167].
Chondrocytes in turn regulate the physiological responses of AECs and ASMCs. The maintenance of epithelial integrity is dependent upon the Fibroblast Growth Factor 2 (FGF-2) [168], which chondrocytes secrete in response to Interleukin-1β (IL-1β) [92]. IL-1β is produced by AECs after an insult from an allergen or related stimuli [29]. This potential feedback loop provides an example of AECs responding to an environmental insult via secretion of mediators leading to activation of nearby chondrocytes to maintain AEC and airway homeostasis. In terms of ASMC regulation, ECM components produced by chondrocytes can serve as a reservoir of Ca2+ ions due to their highly negative net charge. This modulates ASM contraction [147,169], which affects its differentiation and growth. The feedback loop between the production of cartilage ECM and ASM contraction/relaxation affects the extent to which the mechanical effects (contraction and relaxation) of the airway is induced. Such effects have been investigated by Ramchandani et al. [170], where the proportion of cartilage to ASM influences the mechanical compliance of the airways. These interaction varies highly from the developmental to maturation state of the airways [170].
Advanced iPSC-derived multi-cellular co-culture systems have enabled detailed investigation of airway barrier integrity and intercellular signalling. These models incorporate epithelial, mesenchymal, endothelial, and immune cell interactions, providing physiologically relevant platforms to study disease mechanisms and therapeutic interventions [158]. Human and mouse pluripotent stem cell platforms now combine differentiated epithelial, mesenchymal, endothelial, and immune lineages to recapitulate airway cellular neighbourhoods and study interlineage signalling in vitro. These systems range from multi-lineage organoids to air–liquid interface (ALI) co-cultures that permit both paracrine and contact-dependent crosstalk and functional readouts of barrier, differentiation, and progenitor behaviour.
Multi-lineage ALI iAirway assembles iPSC-derived epithelium, mesenchyme (epithelial–mesenchymal organoid cores), endothelium, and macrophages in an ALI format to study barrier responses and pathogen/toxin effects while preserving cross-talk between compartments [158]. Mouse iPSC-derived lung-specific mesenchyme (iLM) can be combined with engineered epithelial progenitors to self-organise into 3D organoids with juxtaposed epithelium and mesenchyme; co-culture increases epithelial progenitor yield and modifies epithelial and mesenchymal differentiation programmes, showing functional reciprocity [171]. Human ESC/iPSC protocols generate airway organoids containing epithelial and mesenchymal populations and can be invested with mesodermal derivatives, enabling modelling of chondrogenesis, smooth muscle formation, and epithelial maturation within one system [172]. Co-culture experiments demonstrate that airway epithelium actively drives airway smooth muscle (ASM) phenotype switching (proliferative/pro-inflammatory) via secreted factors and microRNA-dependent pathways, emphasising the value of paired cultures to study mesenchymal responses [107].
iPSC co-cultures both exploit and reveal the same morphogen axes that pattern the embryonic airway; canonical Wnt, BMP, FGF, and Shh pathways act in reciprocal, context-dependent ways between the epithelium and mesenchyme. In vitro modulation of these signals in mixed-lineage cultures recapitulates lineage choices (chondrocyte vs. smooth muscle), regional identity, and differentiation timing seen in embryos. Bidirectional Wnt signalling between the endoderm (epithelium) and mesoderm (mesenchyme) is necessary to induce mesenchymal tracheal identity (Tbx4) and to generate periodic cartilage and smooth muscle structures in ESC/iPSC-derived cultures; human LPM requires WNT but needs SHH coactivation for correct tracheal mesoderm specification [173]. BMP4 and WNT cooperate during tracheal mesenchyme morphogenesis; mesenchymal BMP4 promotes chondrogenesis and restrains trachealis muscle, and loss of mesenchymal BMP perturbs Wnt target expression and lineage outcomes, findings that are recapitulated in co-culture and organoid assays [174]. Mesenchymal BMPR1A–BMP signalling promotes airway SMC differentiation via Smad-independent pathways (p38 MAPK) rather than solely by canonical Smad1/5, linking receptor-level BMP input in mesenchyme to SMC gene programmes in vitro and in vivo models that inform iPSC differentiation strategies [175]. Standard iPSC airway protocols use FGFs to drive epithelial and mesenchymal maturation and require retinoic acid/SHH (depending on species and protocol) to pattern ventral foregut and lateral-plate mesoderm derivatives, so co-cultures exploit timed FGF/SHH application to produce chondrocytes, smooth muscle, and mature epithelium [172,173].
Overall, a broad and complex interaction is seen between cell types. Fine-tuning of secretory and mechanotransducive effects initiated by resident cells in the airways are necessary for homeostasis conditions, and these conditions arise as concerns in the equilibrium achieved in the developing airway.

3.2. Development: ASM FGF10 and Peristalsis Pattern Cartilage and Epithelial Differentiation

The developmental aspect of airway morphogenesis represents a critical framework for understanding how mechanotransduction and secretome interactions establish the foundational architecture of respiratory tissues. During embryonic development, coordinated cellular interactions between airway smooth muscle cells (ASMCs), epithelial cells, and chondrocytes create the structural and functional blueprint that defines adult airway homeostasis [176,177]. This developmental paradigm is particularly relevant because pathological airway conditions often recapitulate these embryonic signalling pathways, suggesting that understanding developmental mechanobiology provides therapeutic insights for airway diseases. The temporal orchestration of mechanical forces, growth factor gradients, and cellular differentiation during development establishes the bidirectional feedback loops between secretome-mediated signalling and mechanotransduction that persist throughout adult life [158,171,177].
The overview of human airway development is outlined by Pansky; starting at week 4 of embryonic life, a laryngotracheal groove forms and deepens. This part forms the primitive airways, with two bronchi buds forming at week 5 [178]. Differentiation of ASMCs starts at the end of week 7, and, a week after, the visibility of the tracheal cartilage through mesenchymal rudiments can already be seen. The tracheal cartilaginous mass further develops in a cranial to caudal fashion, occurring concurrently as fibroelastic tissues between the rings and airway smooth muscle within the c-ring; a cartilage gap arises within two weeks. On the other hand, cartilage development is seen at week 10 in primary bronchi and week 12 in segmental bronchi [178]. Additionally, ASMCs aid the posterior wall formation of the larger bronchi where cartilage tissue is absent. In the trachea, ciliated epithelium appears at week 10, and mucosal glands are seen from week 12, similarly following a craniocaudal direction [176]. The bronchi, however, form a ciliated epithelium at week 12 and a week afterwards produce mucous glands [178]. At week 20, the microscopic features of the trachea are visible, followed by the bronchus; both the final forms are reached post-natal stage [176]. A general scheme of developmental signalling is shown in Figure 3.
The importance of mesenchymal cells on the developing organ highlights the potentially important role of airway smooth muscle for maturation of the airway, as it is differentiated earlier than the rest of the cells. ASMCs drive the layout structure of the airways via FGF-10 [177] signalling, which is required for lung morphogenesis [179]. Airway ASM peristalsis in the prenatal environment [180], which increases frequency towards birth [181], cues intermittent c-ring structure formation in the trachea as mechanical contraction varies periodically across the tissue. Cartilage maturation can also be observed in co-culture of chondrocytes with ASMCs, increasing production of collagen II and IX by chondrocytes and increasing their pro-chondrogenic activity [182]. Both cell types regulate each other on proliferation, differentiation, and airway biomechanics [158], which in turn affect the epithelial coverage and differentiation of the airways [183].
As the ASM forms, it pushes the luminal fluid to the terminal ends of the airways by peristaltic phasic contraction [184]. Rhythmic ASMC contractility is observed due to intercellular calcium wave propagation [185]. These calcium waves can then affect calcium-activated chloride channels that release chlorine ions to the adjacent AECs, stimulating the secretion of prenatal lung liquid. This is known as the fluid pump hypothesis [186], where the interaction between ASMCs and AECs via mechanotransduction and secretome pathways are highlighted as necessary to support lung development. AECs can also release nitric oxide (NO), which can act as an ASM relaxant by decreasing the Ca2+ oscillation on cells [187]. NO is found to be released in the bronchial and proximal bronchiolar epithelia in the foetal state, suggesting its contribution to airway morphogenesis [188]. Phenotypically, ASMCs that are in a non-contractile state are in a proliferative state, with the associated reduced expression of contractile components such as smooth muscle myosin heavy chain (sm-MHC), calponin, sm-alpha-actin, and desmin [189]. A similar effect on ASMC relaxation can also be induced by PGE2 [190], which can be secreted from AECs [191]. It is therefore apparent that the airway epithelium can regulate contraction of ASM [191,192,193] in the early lung development. Since the epithelium is the exposed layer of the airways, stimuli coming from external sources can cause the release of agonists that regulate the contraction and relaxation of ASM. AECs can be seen to provide biochemical stimuli, and ASMCs can be seen to provide mechanical stimuli.
Cartilage, on the other hand, can influence the phenotype and/or function of AECs. The absence of cartilage in the trachea has been proven to decrease basal cell density, precocious development of club cells, and the KRT14+ cell population (a cell that has high reparative effects on the airways), all likely due to altered FGF signalling [158]. Chondrocytes express and release of Connective Tissue Growth Factor (CTGF) [46,73,74,75,88], which can induce epithelial–mesenchymal transition (EMT) [194], generating fibroblasts [195] between the layers of where the AEC and chondrocyte reside. Such fibroblasts can proliferate under the presence of FGF [196] (which chondrocytes also produce), and so in time develop a perichondrium layer. The presence of a perichondrium in the intersection of epithelial and cartilage tissue layers could, therefore, result from the paracrine effects of the chondrocytes on the basal layer of the respiratory epithelium.
Critically, these developmental signalling pathways depicted in Figure 3 are reactivated during airway regeneration following injury, demonstrating that embryonic patterning programmes serve as templates for adult tissue repair [197,198]. Post-injury regeneration recapitulates key elements of developmental morphogenesis, where Wnt signalling activation promotes basal cell proliferation and differentiation, while BMP pathway modulation controls the balance between proliferation and differentiation phases essential for proper epithelial restoration [199,200]. FGF-10 signalling, crucial for embryonic epithelial branching, becomes reactivated after airway injury to stimulate basal stem cell responses and coordinate epithelial repair [201,202]. ASMCs, upon onset of injury, express FGF-10 [177], which promotes epithelial repair [103]. Similarly, Sox9-mediated pathways are reinitiated during injury repair to restore structural integrity, while maintaining progenitor cell states essential for airway regeneration [203,204]. Results from McVicar et al. also reinforce the likelihood of the cartilage as a developmental niche for airway basal cells [158], supporting the epithelial regeneration pathway. The temporal dynamics of these reactivated developmental cascades determine successful regeneration outcomes; early Wnt activation drives proliferative expansion, followed by precise signalling restoration to promote proper differentiation [197,205]. The crosstalk between cells helps maintain the integrity of the airways, following the same developmental pathways for whole-organ homeostasis. Understanding this developmental/regenerative pathway overlap is essential for designing therapeutic interventions that harness endogenous repair mechanisms while preventing pathological pathway dysregulation that can lead to aberrant remodelling. However, in pathological conditions, signalling and biomechanics are compromised, resulting in unsynchronised overgrowth or degradation of tissues within the airways, coupled with overlapping signals that perpetuate the condition.
Further elucidation of the developmental pathways via multicellular iPSC systems operationalizes fundamental embryology—reciprocal epithelial–mesenchymal signalling, spatially patterned progenitor competence, and mechanical/contractile feedback; these have been reproduced and are experimentally tractable in vitro. Recapitulation both validates embryologic models and allows researchers to perturb single axes while preserving the multicellular context. Co-culture organoids and ALI platforms model the epithelial–mesenchymal trophic unit (EMTU), showing how epithelial signals instruct mesenchymal differentiation, and, conversely, mesenchyme alters epithelial progenitor kinetics and fate—paralleling concepts from developmental lung biology and asthma co-culture studies [171,206].
Single-cell and live-imaging studies show that the sub-epithelial mesenchyme gives rise to airway smooth muscle and that Wnt activation induces early cytoskeletal (F-actin) and adhesion programmes that change epithelial morphology; iPSC co-cultures reproduce these spatiotemporal cues, allowing for the dissection of biochemical versus mechanical contributions to morphogenesis [207]. By rebuilding developmentally patterned signalling (Wnt↔BMP↔FGF↔Shh) in controlled multicellular cultures, iPSC co-cultures link embryologic mechanisms to human-specific differentiation outcomes and disease modelling (e.g., congenital airway malformations, ASM remodelling), and provide platforms to test how altering one compartment shifts the developmental trajectory of its neighbours [173,174,175].

3.3. Disease: Developmental Programmes Misapplied in Asthma and COPD and the Feed-Forward Stiffness Trap

The mechanobiology and signalling pathways in the airways are significantly altered in pathological conditions. Indeed, accumulating evidence indicates that aberrant mechanotransduction is a unifying feature of chronic airway diseases. In asthma and COPD, aberrant mechanobiology drives pathological remodelling through multiple interconnected pathways. Dynamic mechanical stimulation studies using advanced biomaterial systems demonstrate that cyclic strain induces mucus hypersecretion in human bronchial organoids, while altered matrix mechanics promotes fibroblast activation and collagen deposition [208,209]. Another example is the persistently elevated YAP/TAZ signalling observed in the fibrotic airway remodelling of asthma and COPD, analogous to its pro-fibrotic role in idiopathic pulmonary fibrosis [210]. Such dysregulated mechanical signalling can drive excessive cell proliferation and matrix deposition, suggesting that pathways maintaining normal airway structure become pathologically overactive in disease.
Current information on murine and porcine cystic fibrosis models show cartilage and ASM abnormalities prior to subsequent epithelial defects [211,212,213]. Similarly, in the developing airways, epithelial defects are also seen when there are aberrations in cartilage and ASM formation [158]. Mechanistic parallels between lung development and adult disease are now being recognised. The same Hippo–YAP/TAZ pathway that is crucial for normal airway morphogenesis can be inappropriately reactivated in asthma, promoting abnormal cell growth and airway remodelling. In other words, developmental mechanotransducive signals, when dysregulated, may contribute to the cascade of changes seen in the asthmatic airway. The hypothesis on recapitulation of developmental signalling in the adult state could also be reflected in the pathogenesis in asthma—inferior mechanical properties of the cartilage [214], increased ASM mass [215,216] coupled with hypercontractility [217,218], and an altered AEC phenotype whose integrity is lost [219]. Figure 4 shows the anatomy of COPD and asthma in the lower airways.
As previously discussed, mechanical events such as the cycles of contraction and relaxation in the airways can affect the release of soluble factors, which in turn can have paracrine effects of mediator release and mechanobiology. However, excessive mechanical activity and elevated soluble factors release can arise from pathologic conditions, such as asthma, which shifts the equilibrium so that cellular responses are aberrant. In addition, acute mechanical injury to the epithelium can initiate inflammatory cascades that exacerbate airway disease. It was recently shown that bronchiolar club cells act as mechanical damage sensors via the TRPV4 channel; when epithelial junctions are compromised, TRPV4-mediated Ca2+ signalling in club cells triggers release of “danger” signals that drive type 2 inflammation and allergic sensitization [220]. This mechanosensory pathway directly links epithelial barrier stress to asthma pathogenesis, providing a molecular explanation for how repeated epithelial damage (even in the absence of infection) can lead to chronic inflammation. These events alter the microenvironment, specifically the ECM components, and can lead to the perpetuation of pathologic conditions in the airways. For example in asthma, airway remodelling is vividly seen; ASM mass is increased [216], becoming hypercontractile [217], and degradation of cartilage with increased perichondral fibrosis occurs [214].
Excessive mechanical stress on the asthmatic airway wall can itself exacerbate pathology. A recent study demonstrated that increasing the ECM stiffness in airways (by exogenous collagen cross-linking) is sufficient to provoke excessive airway narrowing, even in the absence of inflammation [33]. This finding suggests that a stiffer airway matrix—a hallmark of asthmatic remodelling—directly heightens bronchial hyperreactivity. In asthma, such feed-forward mechanobiological disruption can create a vicious cycle in which remodelling begets stiffness, and stiffness in turn triggers hyperconstriction and further remodelling. This is affected by altered crosstalk between ASMCs, AECs, and chondrocytes. Asthmatic ASMCs release increased amounts of PGE2 [21] that can cause delayed development of chondrocytes by the inhibition of BMP signalling [221]. Dedifferentiated chondrocytes express elevated levels of smooth muscle actin [222], synthesise collagen type I [223], and acquire a contractile phenotype [222,224]. Additionally, IL-1β, which is involved in the pathogenesis of asthma, can causes FGF-2 release from chondrocytes [92], which could promote fibroblast proliferation in the perichondrium and contribute to perichondral fibrosis in asthma [214]. Furthermore, since ASMC contraction can activate TGF-β [139], which is a protein crucial for ECM production and the pathological remodelling of airways, it is possible that enhanced airway contraction in asthma drives structural changes within the airways, as shown in our previous study [164] Aberrant mechanotransduction in ASMCs is a major driver of asthma’s pathological airway remodelling. Asthmatic ASMCs show abnormally high nuclear YAP levels, which promote excessive proliferation and a hypercontractile phenotype [225]. Moreover, stiffened peribronchial matrices in asthma can further amplify ASMC dysfunction; the matrix protein Fibulin-5, for instance, engages β1-integrins on ASMCs and activates YAP/FOXM1 signalling, enhancing smooth muscle migration and contractility [225]. This YAP-mediated feed-forward loop in ASM cells contributes to sustained airway narrowing and increased bronchial wall thickness in asthma.
The collateral damage brought by structural and morphological changes in ASMCs and chondrocytes relays to the airway epithelium. Amphiregulin, which is highly secreted from asthmatic ASMCs, causes elevated expression of VEGF, PGE2, Cyclooxygenase-2, and CXCL8 in AECs [99,226], which can modulate ASMC contraction and proliferation. In addition, the mechanotransducive effect of the ASMCs can also be perpetuated by the secretion of Endothelin-1 by AECs upon compression via ASM contraction to drive further ASMC proliferation and contraction [124]. In chronic pathological conditions, the airway epithelium is dysfunctional [227], and one explanation suggests that this is possibly due to the CTGF release of ASMCs from TGF-β1 stimulation [102] through AEC secretion [29]. It was mentioned in a previous discussion concerning AEC–chondrocyte interaction that CTGF may play a role in the epithelial–mesenchymal transition of AECs, which could also be the case in ASMC-AEC crosstalk. However, in pathologic conditions such as COPD and asthma, CTGF overexpression promotes AEC senescence [228]. These senescent AECs alter the phenotype of the respiratory epithelium in pathological conditions. Peribronchial fibrosis, submucosal gland hypertrophy, and mucous metaplasia are the changes observed in the epithelium [214,229]. There is also growing appreciation for epithelial mechanobiology in chronic airway disease. An asthmatic bronchial epithelium often exhibits loss of integrity and stress-induced changes; studies have found regions of epithelial denudation, reduced ciliated cell numbers, and elevated EGFR and TGF-β signalling even in mild or early asthma, changes observed even in the absence of heavy inflammation [230].
These findings imply that mechanical injury (from repeated bronchoconstriction, coughing, or particulate exposure) contributes to epithelial damage and dysregulation in asthma. Notably, YAP/TAZ, which in healthy airways help restrain goblet cell hyperplasia, become mislocalised or inactivated in injured epithelium—leading to excessive goblet cell differentiation and mucus overproduction [231]. Thus, mechanical stress and YAP/TAZ dysfunction in the airway epithelium together drive the mucous metaplasia and barrier impairment characteristic of chronic asthma. Additionally, since ASMCs are highly proliferative and contractile in these conditions, they can likely affect neighbouring cells due to changes in secretory and mechanobiological processes. Exacerbated by the denudation of cartilage in several pathological conditions, renewal of epithelial integrity is difficult without any external intervention. Smooth muscle and cartilage dysfunction in chronic airway diseases are intimately connected. In both asthma and COPD, bronchial cartilage shows evidence of degeneration (loss of cartilage matrix and viable chondrocytes) along with increased perichondrial fibrosis [214]. This degraded cartilage provides less mechanical support to the airway, making the airway wall more susceptible to narrowing or collapse when surrounding smooth muscle contracts. In emphysema-associated COPD, the problem is compounded by the destruction of alveolar attachments (the elastic fibres tethering small airways open), which removes critical radial tension on the airway walls [225]. The net result of these structural changes is an airway that is abnormally prone to deformation—stiff in some regions and collapsible in others—leading to airflow limitation that is refractory to normal reversal mechanisms.
Organoid and co-culture platforms have been applied to airway disease modelling (fibrosis, asthma, infection), revealing how epithelial injury or inflammatory signalling remodels mesenchyme and how mesenchymal signalling reshapes epithelial fate and ECM, with mechanotransduction and cytoskeletal forces modulating these outcomes [158,232]. In disease-focused co-cultures and EMTU models, epithelial-derived cytokines and growth factors drive ASM phenotype changes toward pro-proliferative and pro-inflammatory states (increased IL-6/IL-8, miR-210, AKT activation), establishing a paracrine axis by which damaged or asthmatic epithelium amplifies mesenchymal remodelling [107,206]. Live-imaging and single-cell studies further show that Wnt-driven mesenchymal patterning coordinates cortical tension and migratory behaviours that influence epithelial morphology, implying mechanotransductive feedback during both normal branching and pathological remodelling [175].
Conditioned media and co-culture experiments show epithelial release of mitogens/cytokines that activate ASMC proliferation, inflammatory gene programmes, and miRNA-mediated repression of tumour suppressors, shifting ASM toward a synthetic/proliferative phenotype relevant to asthma and remodelling [107,206]. Disrupting BMP signalling in mesenchyme alters the cartilage/SMC balance, impairs elastin deposition, and produces cystic or fibrotic airway phenotypes; BMP–WNT reciprocity governs mesenchymal differentiation and thus affects epithelial architecture in disease models [178,179]. Early [174,175] Wnt activation in mesenchyme induces local F-actin accumulation and patterned cortical forces that change epithelial morphology; these cytoskeletal/mechanical cues are accessible in organoid/co-culture systems and likely contribute to disease-associated remodelling when signalling is aberrant [174,175]. Multi-lineage iPSC co-cultures (epithelium + mesenchyme + endothelium + immune cells) recapitulate barrier loss and inflammatory remodelling in response to viral infection or toxins and provide a testbed to probe epithelial–mesenchymal–chondrocyte interactions under pathological perturbation [158,232]. Collectively, recent iPSC-based organoids and layered co-cultures permit the manipulation of Wnt, BMP, FGF, and Shh inputs and allow paired biochemical and biophysical readouts (cytokines, miRNAs, ECM, cortical tension) to dissect epithelial–SMC–chondrocyte crosstalk in development and disease [173,174,175,206].

3.4. Mesenchymal Stromal Cells as Global Mediators

There have been several attempts to regulate the pathologies in the airways, from pharmaceutical to cellular-based methods. Looking back at the developmental origins of airway growth, progenitor cells may be the key to recapitulate the regenerative capacity of the airways. Lung mesenchymal stromal cells have been recently receiving attention regarding the possibility for them to replace dysfunctional cells and ameliorate symptoms associated with the pathologies described above.
With the plethora of growth factors, cytokines, and other functional molecules that they produce, MSCs provide many of the necessary conditions for proliferation and phenotype maintenance of other airway cells and therefore interact with each of the airway cell types. Moreover, MSCs are also capable of differentiating into a specific cell type to regenerate the tissue during repair [83]. It has been well documented that MSCs are present in the lungs [233,234] and are capable of differentiating to all classes of airway cells [235,236]. These lung-resident MSCs also contribute significantly to the immune regulation of the airways and are seen vividly in lung allografts where they intervene T-cell expansion [237]. Airway regeneration is mediated by MSCs from cellular and tissue-level damages, where MSC mobilisation is induced by FGF-10 [238] that is secreted by ASMCs in response to injury [103].
Cellular interactions of MSCs and airway cells are best viewed in co-culture systems. Le Visage et al. performed an air–liquid interface transwell co-culture of MSCs and AECs and found that the co-culture retained the capacity of AECs to produce mucins in contrast to AECs alone [239]. This functional retention can be attributed to MSCs’ ability not only to transfer molecules via paracrine secretion but also organelles via tunnelling nanotubes [240]. MSCs’ capability to transfer mitochondria to AECs proved to be beneficial in repairing damage induced by cigarette smoke in rats [240]. Furthermore, MSCs can replace lost AECs in the airway epithelium by differentiating into AECs themselves [241].
Modulation of MSCs in the asthmatic activity of ASMCs is evident in the study of Urbanek’s group. In a murine model, delivery of MSCs in trachea reduced the hyperplastic phase of ASM and lowered the mucous metaplasia of AECs. Also, MSCs decreased the levels of cytokines IL-4, 5, and 13 (these cytokines are involved in the recruitment of immune cells), and upregulated its expression of IL-10 and indoleamine 2,3-dioxygenase [242] (which reduces the rate of proliferation of ASMCs) [243]. This function of MSCs suggests they can modulate the cell proliferation of ASMCs and counteract the pathological condition by regulating present ASMC populations. Moreover, MSCs are likely to differentiate in case of cellular loss for replacement, as in cases of muscular atrophy treated with MSCs. Also, the ASMC contraction/relaxation phase can also be regulated by MSCs [244] due to their capability to transfer mitochondria and Ca2+ through tunnelling nanotubes [83], similar to the earlier discussed processes of MSC-AEC interaction. The presence of MSCs in the airways may prevent stenosis by controlling the hypertrophic growth of cells and modulating the contraction induced by ASM.
Co-culture experiments have also highlighted positive reinforcement between MSCs and chondrocytes. Several studies proved that the presence of MSCs in chondrocyte culture increased the expression of glycosaminoglycans and total collagen and eliminated the hypertrophic characteristics of cartilage by reduction in collagen X and MMP 13 expression and the absence of calcification [245,246]. Hypertrophy reduction in both cell types is believed to be the effect of parathyroid hormone-related protein chondrocyte secretion in the presence of MSCs that inhibit alkaline phosphatase and diminish cell enlargement [221,247,248]. Chondrocytes also improve the chondrogenic differentiation of MSCs [249], and, in turn, MSCs promotes proliferation and help maintain the phenotype and microenvironment of chondrocytes by upregulating the ECM protein released in cartilage tissues [250,251]. The mutualism exhibited by cell–cell interaction and paracrine effects of MSCs and chondrocytes is crucial in maintaining the functionality of cartilage in the airways to preserve the structural integrity of the whole organ. Overall, MSCs possess a regenerative capability, as well as modulate important functions of each cell type in the airway by secreting necessary growth factors and cytokines, in addition to transferring organelles such as mitochondria via tunnelling nanotubes.
As the predicted gold standard of airway management is tissue engineering, understanding pathways of the homeostasis maintenance of the airways could potentially unlock successful graft generation. Each cell within the airway secretes its set of functional substances, affecting local and global scales in the airway. An organ-scale effect is induced via mechanotransduction, which influences the overall integrity of the airways—airway smooth muscle as the actuator, cartilage as the regulator, and epithelium as the activator. As the limelight expands on the interactome of the airways, the need arises to look deeper into these interactions to fully understand integrity maintenance and pathophysiological conditions of the airways. Such understanding can lead to well-defined ways to successfully engineer the tissue of the airways.

4. Implications for Tissue Engineering and Therapy Design

4.1. Composition and Failure Modes of Current Constructs

The systematic analysis of engineered airway constructs reveals recurring compositional deficiencies that directly correlate with clinical and preclinical failures. Typical construct compositions and their documented limitations are summarised in Table 2.
We hypothesise that the fundamental failure of engineered airway constructs lies not simply in the absence of individual cell types but in the disruption of essential mechanotransduction–secretome feedback loops that maintain airway homeostasis. Recent insights into secretory mechanotransduction in our paper reveal that airways function through a closed-loop system where mechanical forces directly regulate cellular secretory processes, creating a dynamic equilibrium essential for tissue integrity.

4.2. Paradigm Shift: From Cell-Centric to Systems-Level Engineering

The traditional focus is on individual cell types and material properties; it assumes that successful constructs require correct cell seeding densities and biocompatible scaffolds. Analysis of failure with these mindsets attribute it to cell loss, poor integration, or mechanical mismatch. However, a mechanotransduction-informed approach with a focus on integrated mechanotransduction–secretome feedback loops might lead to the construction of a more matured and advanced engineered tissue. This comes with this article’s core concept that functional constructs require coordinated cellular communication networks and that failures were attributed to disrupted homeostatic mechanisms and lost mechanotransduction pathways, disabling the full maturation pathway of the tissue-engineered airways. Table 3 shows the comparative engineering strategies for a novel approach for tissue engineered constructs using a mechanotransductive–secretome paradigm.

4.3. Translational Considerations: Animal Model Selection and Scaling Effects

Cellular mechanotransduction effects vary significantly across different airway sizes and species due to fundamental allometric scaling laws that govern anatomical, ventilatory, and physiological differences [257,258,259]. This review focuses on generalised airway mechanotransduction effects while explicitly identifying whether data sources originate from mice, human, or porcine studies to address these species-specific variations. Small mammalian models, while valuable for mechanistic insights into secretome–mechanotransduction interactions, cannot replicate human-relevant mechanotransduction environments due to differences in airway geometry, ventilatory patterns, and device performance scaling [260,261,262]. Porcine airways serve as critical translational bridges, offering human-relevant anatomy and clinical-scale mechanotransduction assessment capabilities that enable realistic evaluation of tissue-engineered constructs under physiological mechanical loading conditions [262,263]. Readers should remain cognizant of these species-specific mechanotransduction pathway differences when interpreting the mechanisms underlying airway engineering applications across different experimental models [264,265].

4.4. Conclusions

This comprehensive review presents the first unified map of bidirectional mechanobiology–secretome interactions within human airways, revealing a novel three-component regulatory architecture where epithelial cells function as environmental activators, smooth muscle as mechanical actuators, and cartilage as calcium-dependent regulators. The framework identifies critical mechanotransduction pathways—particularly YAP/TAZ signalling, TRPV4 channels, and TGF-β activation—that create closed-loop feedback systems linking matrix stiffness to cytokine release and pathological airway remodelling [61,266].
As the field advances toward comprehensive airway interaction mapping, we advocate for a fundamental paradigm shift that places mechanobiology at the centre of experimental design and data interpretation. Researchers must abandon the traditional reductionist approach of studying isolated cell types in static culture conditions and instead embrace mechanobiology-aware systems thinking that recognises the airway as an inherently mechanical organ where cellular communication is inseparable from physical forces [267]. We strongly recommend that future studies integrate spatial multi-omics platforms with real-time mechanotransduction monitoring, combining single-nucleus RNA sequencing and spatial transcriptomics (Visium) with simultaneous measurement of YAP/TAZ localization, TRPV4 activation states, and matrix remodelling dynamics across anatomical niches [268,269]. The research community should prioritise organoid-based interaction mapping using lamination-based spatially resolved transcriptomics (LOSRT) that preserves tissue architecture while enabling mechanical conditioning through breath-mimicking cyclic stretch and controlled matrix stiffness gradients [270]. Critically, we believe researchers must move beyond correlational network analysis toward causal mechanistic frameworks that employ targeted perturbation experiments—using pharmacological inhibitors (verteporfin for YAP/TAZ, TRPV4 modulators), mechanical interventions (substrate stiffness manipulation), and secretome modulation (MSC-derived exosomes)—to establish directional signalling relationships rather than mere associations. The future success of airway interaction research depends on standardising mechanotransduction-inclusive experimental protocols where every study incorporates physiological mechanical stimuli as a fundamental variable, recognising that the mechanical environment governs not only individual cell behaviour but the entire landscape of intercellular communication networks that maintain airway homeostasis and drive disease progression.
In application for airway engineering, the mechanotransduction-informed approach represents a fundamental paradigm shift from component-based to systems-based airway tissue engineering. By understanding and recreating the bidirectional mechanotransduction–secretome feedback loops essential for airway homeostasis, this approach addresses the root causes of engineering failures rather than their symptoms. This approach transforms airway tissue engineering from empirical optimisation to mechanistically informed design, offering significantly improved prospects for clinical success.

Author Contributions

Conceptualization, J.R. and C.T.; methodology, J.R.; validation, C.T.; formal analysis, C.T.; investigation, J.R.; resources, J.R. and C.T.; writing—original draft preparation, J.R.; writing—review and editing, C.T.; visualisation, J.R.; supervision, J.R.; project administration, C.T.; funding acquisition, J.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analysed in this study.

Acknowledgments

J.R. would like to acknowledge the help of Francesco Pappalardo in proofreading the paper and Francesca Tarsitano for providing the drawings in Figure 3 and Figure 4. J.R. and C.T. would also like to thank Jesus for providing wisdom and knowledge during the time of writing this paper.

Conflicts of Interest

The authors declare no conflicts of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Abbreviations

The following abbreviations are used in this manuscript:
MSCMesenchymal Stromal Cell
AECAirway Epithelial Cell
ASMCAirway Smooth Muscle Cell
ECMExtracellular Matrix
YAPYes-associated Protein
TAZTranscriptional Coactivator with PDZ-binding Motif
TRPV4Transient Receptor Potential Vanilloid 4
FGF-10Fibroblast Growth Factor 10
COPDChronic Obstructive Pulmonary Disease
TGF-βTransforming Growth Factor Beta
PGE2Prostaglandin E2
NONitric Oxide
IL-1βInterleukin 1 Beta
VEGFVascular Endothelial Growth Factor
CTGFConnective Tissue Growth Factor
EGFEpidermal Growth Factor
IGFInsulin Growth Factor
PDK1Three-Phosphoinositide Dependent Protein Kinase 1
ROCKRho-associated, coiled-coil containing protein kinase
EMTEpithelial–Mesenchymal Transition

References

  1. Pierce, R.J.; Worsnop, C.J. Upper Airway function and dysfunction in respiration. Clin. Exp. Pharmacol. Physiol. 1999, 26, 1–10. [Google Scholar] [CrossRef]
  2. Joo, H.; Park, S.-Y.; Park, S.; Kim, S.-H.; Cho, Y.; Yoo, K.; Jung, K.; Rhee, C. Phenotype of Asthma-COPD Overlap in COPD and Severe Asthma Cohorts. J. Korean Med. Sci. 2022, 37, e236. [Google Scholar] [CrossRef]
  3. Kume, H.; Watanabe, N.; Suzuki, Y. Airway Disorders as Predictive Factors of Exacerbations in Asthma and COPD. In Airway Management in Emergency Medicine; Aslanidis, T., Bersot, C.D.A., Eds.; IntechOpen: London, UK, 2023. [Google Scholar]
  4. ten Hallers, E.J.O.; Rakhorst, G.; Marres, H.A.M.; Jansen, J.A.; van Kooten, T.G.; Schutte, H.K.; van Loon, J.P.; van der Houwen, E.B.; Verkerke, G.J. Animal models for tracheal research. Biomaterials 2004, 25, 1533–1543. [Google Scholar] [CrossRef] [PubMed]
  5. Vacanti, C.A.; Paige, K.T.; Kim, W.S.; Sakata, J.; Upton, J.; Vacanti, J.P. Experimental tracheal replacement using tissue-engineered cartilage. J. Pediatr. Surg. 1994, 29, 201–205. [Google Scholar] [CrossRef] [PubMed]
  6. Wechselberger, G.; Russell, R.C.; Neumeister, M.W.; Schoeller, T.; Piza-Katzer, H.; Rainer, C. Successful transplantation of three tissue-engineered cell types using capsule induction technique and fibrin glue as a delivery vehicle. Plast. Reconstr. Surg. 2002, 110, 123–129. [Google Scholar] [CrossRef] [PubMed]
  7. Weidenbecher, M.; Tucker, H.M.; Gilpin, D.A.; Dennis, J.E. Tissue-engineered trachea for airway reconstruction. Laryngoscope 2009, 119, 2118–2123. [Google Scholar] [CrossRef]
  8. Kojima, K.; Bonassar, L.J.; Roy, A.K.; Vacanti, C.A.; Cortiella, J. Autologous tissue-engineered trachea with sheep nasal chondrocytes. J. Thorac. Cardiovasc. Surg. 2002, 123, 1177–1184. [Google Scholar] [CrossRef]
  9. Olze, H.; Kaschke, O.; Müller, W.D. Investigations to improve the design of an alloplastic epithelialized tracheal replacement. HNO 1997, 45, 453–459. [Google Scholar] [CrossRef]
  10. Kim, J.; Suh, S.W.; Shin, J.Y.; Kim, J.H.; Choi, Y.S.; Kim, H. Replacement of a tracheal defect with a tissue-engineered prosthesis: Early results from animal experiments. J. Thorac. Cardiovasc. Surg. 2004, 128, 124–129. [Google Scholar] [CrossRef]
  11. Gonfiotti, A.; Jaus, M.O.; Barale, D.; Baiguera, S.; Comin, C.; Lavorini, F.; Fontana, G.; Sibila, O.; Rombolà, G.; Jungebluth, P.; et al. The first tissue-engineered airway transplantation: 5-year follow-up results. Lancet 2014, 383, 238–244, Retracted in Lancet 2023, 402, 1510.. [Google Scholar] [CrossRef]
  12. Vogel, G. Report Finds Trachea Surgeon Committed Misconduct. Available online: http://www.sciencemag.org/news/2015/05/report-finds-trachea-surgeon-committed-misconduct (accessed on 27 April 2017).
  13. Kojima, K.; Bonassar, L.J.; Roy, A.K.; Mizuno, H.; Cortiella, J.; Vacanti, C.A. A composite tissue-engineered trachea using sheep nasal chondrocyte and epithelial cells. FASEB J. 2003, 17, 823–828. [Google Scholar] [CrossRef]
  14. Gao, M.; Zhang, H.; Dong, W.; Bai, J.; Gao, B.; Xia, D.; Feng, B.; Chen, M.; He, X.; Yin, M.; et al. Tissue-engineered trachea from a 3D-printed scaffold enhances whole-segment tracheal repair. Sci. Rep. 2017, 7, 5246. [Google Scholar] [CrossRef]
  15. Hatachi, G.; Machino, R.; Tsuchiya, T.; Taura, Y.; Elgalad, A.; Taniguchi, D.; Takagi, K.; Matsumoto, K.; Gunge, K.; Matsuo, N.; et al. Scaffold-free trachea regeneration by tissue engineering with bio-3D printing†. Interact. Cardiovasc. Thorac. Surg. 2018, 26, 745–752. [Google Scholar] [CrossRef]
  16. Lin, C.; Yao, E.; Zhang, K.; Jiang, X.; Croll, S.; Thompson-Peer, K.; Chuang, P.-T. YAP is essential for mechanical force production and epithelial cell proliferation during lung branching morphogenesis. eLife 2017, 6, e21130. [Google Scholar] [CrossRef]
  17. Sanchez-Esteban, J.; Tsai, S.-W.; Sang, J.; Qin, J.; Torday, J.S.; Rubin, L.P. Effects of Mechanical Forces on Lung-Specific Gene Expression. Am. J. Med. Sci. 1998, 316, 200–204. [Google Scholar] [CrossRef]
  18. Yu, X.; Feng, L.; Han, Z.; Wu, B.; Wang, S.; Xiao, Y.; Li, F.; Zhang, L.; Cao, B.; Di, X.; et al. Crosstalk of dynamic functional modules in lung development of rhesus macaques. Mol. Biosyst. 2016, 12, 1342–1349. [Google Scholar] [CrossRef] [PubMed]
  19. Mieczkowski, B.; Seavey, B.F. Anatomy, Head and Neck, Trachea. In StatPearls; StatPearls Publishing: Tampa, FL, USA, 2023. [Google Scholar]
  20. Chawla, A. Imaging of Large and Small Airways: Basic to Advanced. In Thoracic Imaging; Springer: Singapore, 2019; pp. 31–64. [Google Scholar]
  21. Sin, D.D. What Single Cell RNA Sequencing Has Taught Us about Chronic Obstructive Pulmonary Disease. Tuberc. Respir. Dis. 2024, 87, 252–260. [Google Scholar] [CrossRef] [PubMed]
  22. Cao, W.; Li, J.; Che, L.; Yang, R.; Wu, Z.; Hu, G.; Zou, W.; Zhao, Z.; Zhou, Y.; Jiang, X.; et al. Single-cell transcriptomics reveals e-cigarette vapor-induced airway epithelial remodeling and injury. Respir. Res. 2024, 25, 353. [Google Scholar] [CrossRef] [PubMed]
  23. Renaut, S.; Saavedra Armero, V.; Boudreau, D.K.; Gaudreault, N.; Desmeules, P.; Thériault, S.; Mathieu, P.; Joubert, P.; Bossé, Y. Single-cell and single-nucleus RNA-sequencing from paired normal-adenocarcinoma lung samples provide both common and discordant biological insights. PLoS Genet. 2024, 20, e1011301. [Google Scholar] [CrossRef]
  24. Zhou, Y.; Yang, Y.; Guo, L.; Qian, J.; Ge, J.; Sinner, D.; Ding, H.; Califano, A.; Cardoso, W.V. Airway basal cells show regionally distinct potential to undergo metaplastic differentiation. eLife 2022, 11, e80083. [Google Scholar] [CrossRef]
  25. Sophia Fox, A.J.; Bedi, A.; Rodeo, S.A. The basic science of articular cartilage: Structure, composition, and function. Sports Health 2009, 1, 461–468. [Google Scholar] [CrossRef]
  26. Archer, C.W.; Francis-West, P. The chondrocyte. Int. J. Biochem. Cell Biol. 2003, 35, 401–404. [Google Scholar] [CrossRef]
  27. Rojas, M.; Xu, J.; Woods, C.R.; Mora, A.L.; Spears, W.; Roman, J.; Brigham, K.L. Bone marrow–derived mesenchymal stem cells in repair of the injured lung. Am. J. Respir. Cell Mol. Biol. 2005, 33, 145–152. [Google Scholar] [CrossRef] [PubMed]
  28. Amrani, Y.; Panettieri, R.A. Airway smooth muscle: Contraction and beyond. Int. J. Biochem. Cell Biol. 2003, 35 (Suppl. S1), 272–276. [Google Scholar] [CrossRef] [PubMed]
  29. Mills, P.R.; Davies, R.J.; Devalia, J.L. Airway Epithelial Cells, Cytokines, and Pollutants. Am. J. Respir. Crit. Care Med. 1999, 160, S38–S43. [Google Scholar] [CrossRef] [PubMed]
  30. Jesenak, M.; Durdik, P.; Oppova, D.; Franova, S.; Diamant, Z.; Golebski, K.; Banovcin, P.; Vojtkova, J.; Novakova, E. Dysfunctional mucociliary clearance in asthma and airway remodeling—New insights into an old topic. Respir. Med. 2023, 218, 107372. [Google Scholar] [CrossRef] [PubMed]
  31. Roth, D.; Şahin, A.T.; Ling, F.; Tepho, N.; Senger, C.N.; Quiroz, E.J.; Calvert, B.A.; van der Does, A.M.; Güney, T.G.; Glasl, S.; et al. Structure and function relationships of mucociliary clearance in human and rat airways. Nat. Commun. 2025, 16, 2446. [Google Scholar] [CrossRef]
  32. Walentek, P. Signaling Control of Mucociliary Epithelia: Stem Cells, Cell Fates, and the Plasticity of Cell Identity in Development and Disease. Cells Tissues Organs 2022, 211, 736–753. [Google Scholar] [CrossRef]
  33. Guo, T.; He, C.; Venado, A.; Zhou, Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr. Physiol. 2022, 12, 3523–3558. [Google Scholar] [CrossRef]
  34. Horta, C.A.; Doan, K.; Yang, J. Mechanotransduction pathways in regulating epithelial-mesenchymal plasticity. Curr. Opin. Cell Biol. 2023, 85, 102245. [Google Scholar] [CrossRef]
  35. Feng, K.N.; Meng, P.; Zou, X.L.; Zhang, M.; Li, H.K.; Yang, H.L.; Li, H.T.; Zhang, T.T. IL-37 protects against airway remodeling by reversing bronchial epithelial–mesenchymal transition via IL-24 signaling pathway in chronic asthma. Respir. Res. 2022, 23, 244. [Google Scholar] [CrossRef]
  36. Novak, C.M.; Wheat, J.S.; Ghadiali, S.N.; Ballinger, M.N. Mechanomemory of pulmonary fibroblasts demonstrates reversibility of transcriptomics and contraction phenotypes. Biomaterials 2025, 314, 122830. [Google Scholar] [CrossRef] [PubMed]
  37. Tan, Q.; Ma, X.Y.; Liu, W.; Meridew, J.A.; Jones, D.L.; Haak, A.J.; Sicard, D.; Ligresti, G.; Tschumperlin, D.J. Nascent Lung Organoids Reveal Epithelium- and Bone Morphogenetic Protein–mediated Suppression of Fibroblast Activation. Am. J. Respir. Cell Mol. Biol. 2019, 61, 607–619. [Google Scholar] [CrossRef] [PubMed]
  38. Yao, Y.; Miethe, S.; Kattler, K.; Colakoglu, B.; Walter, J.; Schneider-Daum, N.; Herr, C.; Garn, H.; Ritzmann, F.; Bals, R.; et al. Mutual Regulation of Transcriptomes between Murine Pneumocytes and Fibroblasts Mediates Alveolar Regeneration in Air-Liquid Interface Cultures. Am. J. Respir. Cell Mol. Biol. 2024, 70, 203–214. [Google Scholar] [CrossRef] [PubMed]
  39. Wu, Y.; Song, Y.; Soto, J.; Hoffman, T.; Lin, X.; Zhang, A.; Chen, S.; Massad, R.N.; Han, X.; Qi, D.; et al. Viscoelastic extracellular matrix enhances epigenetic remodeling and cellular plasticity. Nat. Commun. 2025, 16, 4054. [Google Scholar] [CrossRef]
  40. Chang, Y.; Lee, J.W.N.; Holle, A.W. The mechanobiology of fibroblast activation in disease. APL Bioeng. 2025, 9, 021505. [Google Scholar] [CrossRef]
  41. Polacek, M.; Bruun, J.A.; Johansen, O.; Martinez, I. Differences in the secretome of cartilage explants and cultured chondrocytes unveiled by SILAC technology. J. Orthop. Res. 2010, 28, 1040–1049. [Google Scholar] [CrossRef]
  42. Polacek, M.; Bruun, J.-A.; Elvenes, J.; Figenschau, Y.; Martinez, I. The secretory profiles of cultured human articular chondrocytes and mesenchymal stem cells: Implications for autologous cell transplantation strategies. Cell Transplant. 2011, 20, 1381–1393. [Google Scholar] [CrossRef]
  43. Rosenthal, A.K.; Gohr, C.M.; Ninomiya, J.; Wakim, B.T. Proteomic analysis of articular cartilage vesicles from normal and osteoarthritic cartilage. Arthritis Rheum. 2011, 63, 401–411. [Google Scholar] [CrossRef]
  44. Cui, Y.; Yu, C.; Lu, Q.; Huang, X.; Lin, W.; Huang, T.; Cao, L.; Yang, Q. The Function of RhoA/ROCK Pathway and MYOCD in Airway Remodeling in Asthma. Int. Arch. Allergy Immunol. 2025, 186, 103–119. [Google Scholar] [CrossRef]
  45. Turcatel, G.; Millette, K.; Thornton, M.; Leguizamon, S.; Grubbs, B.; Shi, W.; Warburton, D. Cartilage rings contribute to the proper embryonic tracheal epithelial differentiation, metabolism, and expression of inflammatory genes. Am. J. Physiol. Lung Cell. Mol. Physiol. 2017, 312, L196–L207. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, J.; Zhu, F.; Luo, R.; Cui, Y.; Zhang, Z.; Xu, M.; Zhao, Y.; He, Y.; Yang, W.; Li, N.; et al. YAP Alleviates Pulmonary Fibrosis Through Promoting Alveolar Regeneration via Modulating the Stemness of Alveolar Type 2 Cells. Stem Cells Dev. 2024, 33, 586–594. [Google Scholar] [CrossRef] [PubMed]
  47. Rossios, C.; Pavlidis, S.; Gibeon, D.; Mumby, S.; Durham, A.; Ojo, O.; Horowitz, D.; Loza, M.; Baribaud, F.; Rao, N.; et al. Impaired innate immune gene profiling in airway smooth muscle cells from chronic cough patients. Biosci. Rep. 2017, 37, BSR20171090. [Google Scholar] [CrossRef] [PubMed]
  48. Hackett, N.R.; Shaykhiev, R.; Walters, M.S.; Wang, R.; Zwick, R.K.; Ferris, B.; Witover, B.; Salit, J.; Crystal, R.G. The Human Airway Epithelial Basal Cell Transcriptome. PLoS ONE 2011, 6, e18378. [Google Scholar] [CrossRef]
  49. Barrow, R.E.; Wang, C.Z.; Evans, M.J.; Herndon, D.N. Growth factors accelerate epithelial repair in sheep trachea. Lung 1993, 171, 335–344. [Google Scholar] [CrossRef]
  50. Retsch-Bogart, G.Z.; Stiles, A.D.; Moats-Staats, B.M.; Van Scott, M.R.; Boucher, R.C.; D’Ercole, A.J. Canine tracheal epithelial cells express the type 1 insulin-like growth factor receptor and proliferate in response to insulin-like growth factor I. Am. J. Respir. Cell Mol. Biol. 1990, 3, 227–234. [Google Scholar] [CrossRef]
  51. Lotz, M. Cytokines in Cartilage Injury and Repair. Clin. Orthop. Relat. Res. 2001, 391, S108–S115. [Google Scholar] [CrossRef]
  52. Pufe, T.; Harde, V.; Petersen, W.; Goldring, M.B.; Tillmann, B.; Mentlein, R. Vascular endothelial growth factor (VEGF) induces matrix metalloproteinase expression in immortalized chondrocytes. J. Pathol. 2004, 202, 367–374. [Google Scholar] [CrossRef]
  53. Aizawa, T.; Kon, T.; Einhorn, T.; Gerstenfeld, L. Induction of apoptosis in chondrocytes by tumor necrosis factor-alpha. J. Orthop. Res. 2001, 19, 785–796. [Google Scholar] [CrossRef]
  54. Ivkovic, S.; Yoon, B.S.; Popoff, S.N.; Safadi, F.F.; Libuda, D.E.; Stephenson, R.C.; Daluiski, A.; Lyons, K.M. Connective tissue growth factor coordinates chondrogenesis and angiogenesis during skeletal development. Development 2003, 130, 2779–2791. [Google Scholar] [CrossRef]
  55. Wang, Y.; Lou, S. Direct protective effect of interleukin-10 on articular chondrocytes in vitro. Chin. Med. J. 2001, 114, 723–725. [Google Scholar]
  56. Jikko, A.; Wakisaka, T.; Iwamoto, M.; Hiranuma, H.; Kato, Y.; Maeda, T.; Fujishita, M.; Fuchihata, H. Effects of interleukin-6 on proliferation and proteoglycan metabolism in articular chondrocyte cultures. Cell Biol. Int. 1998, 22, 615–621. [Google Scholar] [CrossRef]
  57. Govindaraju, V.; Michoud, M.-C.; Ferraro, P.; Arkinson, J.; Safka, K.; Valderrama-Carvajal, H.; Martin, J.G. The effects of interleukin-8 on airway smooth muscle contraction in cystic fibrosis. Respir. Res. 2008, 9, 76. [Google Scholar] [CrossRef] [PubMed]
  58. Kuperman, D.A.; Huang, X.; Koth, L.L.; Chang, G.H.; Dolganov, G.M.; Zhu, Z.; Elias, J.A.; Sheppard, D.; Erle, D.J. Direct effects of interleukin-13 on epithelial cells cause airway hyperreactivity and mucus overproduction in asthma. Nat. Med. 2002, 8, 885. [Google Scholar] [CrossRef] [PubMed]
  59. Tschumperlin, D.J.; Drazen, J.M. Chronic effects of mechanical force on airways. Annu. Rev. Physiol. 2006, 68, 563–583. [Google Scholar] [CrossRef] [PubMed]
  60. Sharma, S.; Goswami, R.; Zhang, D.X.; Rahaman, S.O. TRPV4 regulates matrix stiffness and TGFβ1-induced epithelial-mesenchymal transition. J. Cell. Mol. Med. 2019, 23, 761–774. [Google Scholar] [CrossRef]
  61. Sharma, S.; Goswami, R.; Rahaman, S.O. The TRPV4-TAZ Mechanotransduction Signaling Axis in Matrix Stiffness- and TGFβ1-Induced Epithelial-Mesenchymal Transition. Cell. Mol. Bioeng. 2019, 12, 139–152. [Google Scholar] [CrossRef]
  62. Bandaru, P.; Cefaloni, G.; Vajhadin, F.; Lee, K.; Kim, H.-J.; Cho, H.-J.; Hartel, M.C.; Zhang, S.; Sun, W.; Goudie, M.J.; et al. Mechanical Cues Regulating Proangiogenic Potential of Human Mesenchymal Stem Cells through YAP-Mediated Mechanosensing. Small 2020, 16, 2001837. [Google Scholar] [CrossRef]
  63. Tan, Y.H.; Wang, K.C.W.; Chin, I.L.; Sanderson, R.W.; Li, J.; Kennedy, B.F.; Noble, P.B.; Choi, Y.S. Stiffness Mediated-Mechanosensation of Airway Smooth Muscle Cells on Linear Stiffness Gradient Hydrogels. Adv. Healthc. Mater. 2024, 13, 2304254. [Google Scholar] [CrossRef]
  64. Zhang, M.; Meng, N.; Wang, X.; Chen, W.; Zhang, Q. TRPV4 and PIEZO Channels Mediate the Mechanosensing of Chondrocytes to the Biomechanical Microenvironment. Membranes 2022, 12, 237. [Google Scholar] [CrossRef]
  65. Calloni, G.-W.; Stimamiglio, M.-A. Tuning mesenchymal stem cell secretome therapeutic potential through mechanotransduction. Biocell 2022, 46, 1375–1381. [Google Scholar] [CrossRef]
  66. Djouad, F.; Jackson, W.M.; Bobick, B.E.; Janjanin, S.; Song, Y.; Huang, G.T.; Tuan, R.S. Activin A expression regulates multipotency of mesenchymal progenitor cells. Stem Cell Res. Ther. 2010, 1, 11. [Google Scholar] [CrossRef]
  67. Burdon, T.J.; Paul, A.; Noiseux, N.; Prakash, S.; Shum-Tim, D. Bone Marrow Stem Cell Derived Paracrine Factors for Regenerative Medicine: Current Perspectives and Therapeutic Potential. Bone Marrow Res. 2011, 2011, 207326. [Google Scholar] [CrossRef]
  68. Fang, X.; Neyrinck, A.P.; Matthay, M.A.; Lee, J.W. Allogeneic human mesenchymal stem cells restore epithelial protein permeability in cultured human alveolar type II cells by secretion of angiopoietin-1. J. Biol. Chem. 2010, 285, 26211–26222. [Google Scholar] [CrossRef]
  69. Gardner, O.; Fahy, N.; Alini, M.; Stoddart, M. Differences in human mesenchymal stem cell secretomes during chondrogenic induction. Eur. Cells Mater. 2016, 30, 221–235. [Google Scholar] [CrossRef]
  70. Sumanasinghe, R.D.; Bernacki, S.H.; Loboa, E.G. Osteogenic differentiation of human mesenchymal stem cells in collagen matrices: Effect of uniaxial cyclic tensile strain on bone morphogenetic protein (BMP-2) mRNA expression. Tissue Eng. 2006, 12, 3459–3465. [Google Scholar] [CrossRef]
  71. Arikawa, T.; Omura, K.; Morita, I. Regulation of bone morphogenetic protein-2 expression by endogenous prostaglandin E2 in human mesenchymal stem cells. J. Cell. Physiol. 2004, 200, 400–406. [Google Scholar] [CrossRef] [PubMed]
  72. Wislet-Gendebien, S.; Bruyere, F.; Hans, G.; Leprince, P.; Moonen, G.; Rogister, B. Nestin-positive mesenchymal stem cells favour the astroglial lineage in neural progenitors and stem cells by releasing active BMP4. BMC Neurosci. 2004, 5, 33. [Google Scholar] [CrossRef] [PubMed]
  73. Xia, T.; Pan, Z.; Wan, H.; Li, Y.; Mao, G.; Zhao, J.; Zhang, F.; Pan, S. Mechanisms of Mechanical Stimulation in the Development of Respiratory System Diseases. Am. J. Physiol. Lung Cell Mol. Physiol. 2024, 327, L724–L739. [Google Scholar] [CrossRef] [PubMed]
  74. Omoto, S.; Nishida, K.; Yamaai, Y.; Shibahara, M.; Nishida, T.; Doi, T.; Asahara, H.; Nakanishi, T.; Inoue, H.; Takigawa, M. Expression and localization of connective tissue growth factor (CTGF/Hcs24/CCN2) in osteoarthritic cartilage. Osteoarthr. Cartil. 2004, 12, 771–778. [Google Scholar] [CrossRef]
  75. Fukunaga, T.; Yamashiro, T.; Oya, S.; Takeshita, N.; Takigawa, M.; Takano-Yamamoto, T. Connective tissue growth factor mRNA expression pattern in cartilages is associated with their type I collagen expression. Bone 2003, 33, 911–918. [Google Scholar] [CrossRef]
  76. Crisostomo, P.R.; Wang, Y.; Markel, T.A.; Wang, M.; Lahm, T.; Meldrum, D.R. Human mesenchymal stem cells stimulated by TNF-α, LPS, or hypoxia produce growth factors by an NFκB-but not JNK-dependent mechanism. Am. J. Physiol. Cell Physiol. 2008, 294, C675–C682. [Google Scholar] [CrossRef]
  77. Baraniak, P.R.; McDevitt, T.C. Stem cell paracrine actions and tissue regeneration. Regen. Med. 2010, 5, 121–143. [Google Scholar] [CrossRef]
  78. Chen, L.; Tredget, E.E.; Wu, P.Y.; Wu, Y. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS ONE 2008, 3, e1886. [Google Scholar] [CrossRef] [PubMed]
  79. Takahashi, M.; Li, T.S.; Suzuki, R.; Kobayashi, T.; Ito, H.; Ikeda, Y.; Matsuzaki, M.; Hamano, K. Cytokines produced by bone marrow cells can contribute to functional improvement of the infarcted heart by protecting cardiomyocytes from ischemic injury. Am. J. Physiol. Heart Circ. Physiol. 2006, 291, H886–H893. [Google Scholar] [CrossRef] [PubMed]
  80. Sumanasinghe, R.D.; Pfeiler, T.W.; Monteiro-Riviere, N.A.; Loboa, E.G. Expression of proinflammatory cytokines by human mesenchymal stem cells in response to cyclic tensile strain. J. Cell. Physiol. 2009, 219, 77–83. [Google Scholar] [CrossRef] [PubMed]
  81. Nemoto, Y.; Kanai, T.; Takahara, M.; Oshima, S.; Nakamura, T.; Okamoto, R.; Tsuchiya, K.; Watanabe, M. Bone marrow-mesenchymal stem cells are a major source of interleukin-7 and sustain colitis by forming the niche for colitogenic CD4 memory T cells. Gut 2013, 62, 1142–1152. [Google Scholar] [CrossRef]
  82. Windmolders, S.; De Boeck, A.; Koninckx, R.; Daniëls, A.; De Wever, O.; Bracke, M.; Hendrikx, M.; Hensen, K.; Rummens, J.-L. Mesenchymal stem cell secreted platelet derived growth factor exerts a pro-migratory effect on resident Cardiac Atrial appendage Stem Cells. J. Mol. Cell. Cardiol. 2014, 66, 177–188. [Google Scholar] [CrossRef]
  83. Spees, J.L.; Lee, R.H.; Gregory, C.A. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Res. Ther. 2016, 7, 125. [Google Scholar] [CrossRef]
  84. Li, F.; Armstrong, G.B.; Tombran-Tink, J.; Niyibizi, C. Pigment epithelium derived factor upregulates expression of vascular endothelial growth factor by human mesenchymal stem cells: Possible role in PEDF regulated matrix mineralization. Biochem. Biophys. Res. Commun. 2016, 478, 1106–1110. [Google Scholar] [CrossRef]
  85. Chosa, E.; Hamada, H.; Kitamura, K.; Kuwasako, K.; Yanagita, T.; Eto, T.; Tajima, N. Expression of adrenomedullin and its receptor by chondrocyte phenotype cells. Biochem. Biophys. Res. Commun. 2003, 303, 379–386. [Google Scholar] [CrossRef]
  86. Velard, F.; Chatron-Colliet, A.; Côme, D.; Ah-Kioon, M.-D.; Lin, H.; Hafsia, N.; Cohen-Solal, M.; Ea, H.-K.; Lioté, F. Adrenomedullin and truncated peptide adrenomedullin(22-52) affect chondrocyte response to apoptotis in vitro: Downregulation of FAS protects chondrocyte from cell death. Sci. Rep. 2020, 10, 16740. [Google Scholar] [CrossRef]
  87. Murata, M.; Yudo, K.; Nakamura, H.; Chiba, J.; Okamoto, K.; Suematsu, N.; Nishioka, K.; Beppu, M.; Inoue, K.; Kato, T.; et al. Hypoxia upregulates the expression of angiopoietin-like-4 in human articular chondrocytes: Role of angiopoietin-like-4 in the expression of matrix metalloproteinases and cartilage degradation. J. Orthop. Res. 2009, 27, 50–57. [Google Scholar] [CrossRef]
  88. Huang, B.-L.; Brugger, S.M.; Lyons, K.M. Stage-specific control of connective tissue growth factor (CTGF/CCN2) expression in chondrocytes by Sox9 and β-catenin. J. Biol. Chem. 2010, 285, 27702–27712. [Google Scholar] [CrossRef]
  89. Hu, B.; Trinh, K.; Figueira, W.F.; Price, P.A. Isolation and sequence of a novel human chondrocyte protein related to mammalian members of the chitinase protein family. J. Biol. Chem. 1996, 271, 19415–19420. [Google Scholar] [CrossRef] [PubMed]
  90. Huh, Y.-H.; Kim, S.-H.; Kim, S.-J.; Chun, J.-S. Differentiation status-dependent regulation of cyclooxygenase-2 expression and prostaglandin E2 production by epidermal growth factor via mitogen-activated protein kinase in articular chondrocytes. J. Biol. Chem. 2003, 278, 9691–9697. [Google Scholar] [CrossRef] [PubMed]
  91. Ruocco, S.; Lallemand, A.; Tournier, J.M.; Gaillard, D. Expression and localization of epidermal growth factor, transforming growth factor-α, and localization of their common receptor in fetal human lung development. Pediatr. Res. 1996, 39, 448–455. [Google Scholar] [CrossRef] [PubMed]
  92. Chien, S.-Y.; Huang, C.-Y.; Tsai, C.-H.; Wang, S.-W.; Lin, Y.-M.; Tang, C.-H. Interleukin-1β induces fibroblast growth factor 2 expression and subsequently promotes endothelial progenitor cell angiogenesis in chondrocytes. Clin. Sci. 2016, 130, 667–681. [Google Scholar] [CrossRef]
  93. Mohtai, M.; Gupta, M.K.; Donlon, B.; Ellison, B.; Cooke, J.; Gibbons, G.; Schurman, D.J.; Smith, R.L. Expression of interleukin-6 in osteoarthritic chondrocytes and effects of fluid-induced shear on this expression in normal human chondrocytes in vitro. J. Orthop. Res. 1996, 14, 67–73. [Google Scholar] [CrossRef]
  94. Stadler, J.; Stefanovic-Racic, M.; Billiar, T.R.; Curran, R.D.; Mcintyre, L.A.; Georgescu, H.I.; Simmons, R.L.; Evans, C.H. Articular chondrocytes synthesize nitric oxide in response to cytokines and lipopolysaccharide. J. Immunol. 1991, 147, 3915–3920. [Google Scholar] [CrossRef]
  95. Zappia, J.; Tong, Q.; Van der Cruyssen, R.; Cornelis, F.M.F.; Lambert, C.; Pinto Coelho, T.; Grisart, J.; Kague, E.; Lories, R.J.; Muller, M.; et al. Osteomodulin downregulation is associated with osteoarthritis development. Bone Res. 2023, 11, 49. [Google Scholar] [CrossRef] [PubMed]
  96. Lingaraj, K.; Poh, C.K.; Wang, W. Vascular endothelial growth factor (VEGF) is expressed during articular cartilage growth and re-expressed in osteoarthritis. Ann. Acad. Med. Singap. 2010, 39, 399. [Google Scholar] [CrossRef] [PubMed]
  97. Upton, P.D.; Wharton, J.; Davie, N.; Ghatei, M.A.; Smith, D.M.; Morrell, N.W. Differential adrenomedullin release and endothelin receptor expression in distinct subpopulations of human airway smooth-muscle cells. Am. J. Respir. Cell Mol. Biol. 2001, 25, 316–325. [Google Scholar] [CrossRef]
  98. Alagappan, V.K.T.; McKay, S.; Widyastuti, A.; Garrelds, I.M.; Bogers, A.J.J.C.; Hoogsteden, H.C.; Hirst, S.J.; Sharma, H.S. Proinflammatory cytokines upregulate mRNA expression and secretion of vascular endothelial growth factor in cultured human airway smooth muscle cells. Cell Biochem. Biophys. 2005, 43, 119–129. [Google Scholar] [CrossRef]
  99. Deacon, K.; Knox, A.J. Human airway smooth muscle cells secrete amphiregulin via bradykinin/COX-2/PGE(2), inducing COX-2, CXCL8, and VEGF expression in airway epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2015, 309, L237–L249. [Google Scholar] [CrossRef]
  100. Shim, J.Y.; Park, S.W.; Kim, D.S.; Shim, J.W.; Jung, H.L.; Park, M.S. The Effect of Interleukin-4 and Amphiregulin on the Proliferation of Human Airway Smooth Muscle Cells and Cytokine Release. J. Korean Med. Sci. 2008, 23, 857–863. [Google Scholar] [CrossRef]
  101. Johnson, P.R.A.; Burgess, J.K.; Ge, Q.; Poniris, M.; Boustany, S.; Twigg, S.M.; Black, J.L. Connective Tissue Growth Factor Induces Extracellular Matrix in Asthmatic Airway Smooth Muscle. Am. J. Respir. Crit. Care Med. 2006, 173, 32–41. [Google Scholar] [CrossRef]
  102. Xie, S.; Sukkar, M.B.; Issa, R.; Oltmanns, U.; Nicholson, A.G.; Chung, K.F. Regulation of TGF-β1-induced connective tissue growth factor expression in airway smooth muscle cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2005, 288, L68–L76. [Google Scholar] [CrossRef]
  103. Volckaert, T.; Dill, E.; Campbell, A.; Tiozzo, C.; Majka, S.; Bellusci, S.; De Langhe, S.P. Parabronchial smooth muscle constitutes an airway epithelial stem cell niche in the mouse lung after injury. J. Clin. Investig. 2011, 121, 4409–4419. [Google Scholar] [CrossRef]
  104. Oliveira, L.C.; Danilucci, T.M.; Chaves-Neto, A.H.; Campanelli, A.P.; Silva, T.C.; Oliveira, S.H. Tracheal Smooth Muscle Cells Stimulated by Stem Cell Factor-c-Kit Coordinate the Production of Transforming Growth Factor-beta1 and Fibroblast Growth Factor-2 Mediated by Chemokine (C-C Motif) Ligand 3. J. Interferon Cytokine Res. 2016, 36, 401–411. [Google Scholar] [CrossRef]
  105. Coffey, E.; Newman, D.R.; Sannes, P.L. Expression of Fibroblast Growth Factor 9 in Normal Human Lung and Idiopathic Pulmonary Fibrosis. J. Histochem. Cytochem. 2013, 61, 671–679. [Google Scholar] [CrossRef]
  106. Wen, F.-Q.; Liu, X.; Manda, W.; Terasaki, Y.; Kobayashi, T.; Abe, S.; Fang, Q.; Ertl, R.; Manouilova, L.; Rennard, S.I. TH2 Cytokine-enhanced and TGF-β-enhanced vascular endothelial growth factor production by cultured human airway smooth muscle cells is attenuated by IFN-γ and corticosteroids. J. Allergy Clin. Immunol. 2003, 111, 1307–1318. [Google Scholar] [CrossRef]
  107. O’Sullivan, M.J.; Jang, J.H.; Panariti, A.; Bedrat, A.; Ijpma, G.; Lemos, B.; Park, J.A.; Lauzon, A.M.; Martin, J.G. Airway Epithelial Cells Drive Airway Smooth Muscle Cell Phenotype Switching to the Proliferative and Pro-inflammatory Phenotype. Front. Physiol. 2021, 12, 687654. [Google Scholar] [CrossRef]
  108. Johnson, S.R.; Knox, A.J. Synthetic functions of airway smooth muscle in asthma. Trends Pharmacol. Sci. 1997, 18, 288–292. [Google Scholar] [CrossRef] [PubMed]
  109. McKay, S.; Hirst, S.J.; Haas, M.B.-d.; De Jongste, J.C.; Hoogsteden, H.C.; Saxena, P.R.; Sharma, H.S. Tumor necrosis factor-α enhances mRNA expression and secretion of interleukin-6 in cultured human airway smooth muscle cells. Am. J. Respir. Cell Mol. Biol. 2000, 23, 103–111. [Google Scholar] [CrossRef] [PubMed]
  110. John, M.; Au, B.-T.; Jose, P.J.; Lim, S.; Saunders, M.; Barnes, P.J.; Mitchell, J.A.; Belvisi, M.G.; Fan Chung, K. Expression and release of interleukin-8 by human airway smooth muscle cells: Inhibition by Th-2 cytokines and corticosteroids. Am. J. Respir. Cell Mol. Biol. 1998, 18, 84–90. [Google Scholar] [CrossRef] [PubMed]
  111. Freund, V.; Pons, F.; Joly, V.; Mathieu, E.; Martinet, N.; Frossard, N. Upregulation of nerve growth factor expression by human airway smooth muscle cells in inflammatory conditions. Eur. Respir. J. 2002, 20, 458–463. [Google Scholar] [CrossRef]
  112. McKay, S.; De Jongste, J.C.; Saxena, P.R.; Sharma, H.S. Angiotensin II induces hypertrophy of human airway smooth muscle cells: Expression of transcription factors and transforming growth factor-β1. Am. J. Respir. Cell Mol. Biol. 1998, 18, 823–833. [Google Scholar] [CrossRef]
  113. Knox, A.J.; Corbett, L.; Stocks, J.; Holland, E.; Zhu, Y.M.; Pang, L. Human airway smooth muscle cells secrete vascular endothelial growth factor: Up-regulation by bradykinin via a protein kinase C and prostanoid-dependent mechanism. FASEB J. 2001, 15, 2480–2488. [Google Scholar] [CrossRef]
  114. Alagappan, V.K.T.; Willems-Widyastuti, A.; Seynhaeve, A.L.B.; Garrelds, I.M.; ten Hagen, T.L.M.; Saxena, P.R.; Sharma, H.S. Vasoactive peptides upregulate mRNA expression and secretion of vascular endothelial growth factor in human airway smooth muscle cells. Cell Biochem. Biophys. 2007, 47, 109–118. [Google Scholar] [CrossRef]
  115. Fehrenbach, H.; Wagner, C.; Wegmann, M. Airway remodeling in asthma: What really matters. Cell Tissue Res. 2017, 367, 551–569. [Google Scholar] [CrossRef] [PubMed]
  116. Lee, K.-Y.; Ho, S.-C.; Lin, H.-C.; Lin, S.-M.; Liu, C.-Y.; Huang, C.-D.; Wang, C.-H.; Chung, K.F.; Kuo, H.-P. Neutrophil-derived elastase induces TGF-β1 secretion in human airway smooth muscle via NF-κB pathway. Am. J. Respir. Cell Mol. Biol. 2006, 35, 407–414. [Google Scholar] [CrossRef] [PubMed]
  117. Gerayeli, F.V.; Park, H.Y.; Milne, S.; Li, X.; Yang, C.X.; Tuong, J.; Eddy, R.L.; Vahedi, S.M.; Guinto, E.; Cheung, C.Y.; et al. Single-cell sequencing reveals cellular landscape alterations in the airway mucosa of patients with pulmonary long COVID. Eur. Respir. J. 2024, 64, 2301947. [Google Scholar] [CrossRef] [PubMed]
  118. Martinez, A.; Miller, M.; Unsworth, E.J.; Siegfried, J.M.; Cuttitta, F. Expression of adrenomedullin in normal human lung and in pulmonary tumors. Endocrinology 1995, 136, 4099–4105. [Google Scholar] [CrossRef]
  119. Maunders, H.; Patwardhan, S.; Phillips, J.; Clack, A.; Richter, A. Human bronchial epithelial cell transcriptome: Gene expression changes following acute exposure to whole cigarette smoke in vitro. Am. J. Physiol. Lung Cell Mol. Physiol. 2007, 292, L1248–L1256. [Google Scholar] [CrossRef]
  120. Recklies, A.D.; White, C.; Hua, L. The chitinase 3-like protein human cartilage glycoprotein 39 (HC-gp39) stimulates proliferation of human connective-tissue cells and activates both extracellular signal-regulated kinase-and protein kinase B-mediated signalling pathways. Biochem. J. 2002, 365, 119–126. [Google Scholar] [CrossRef]
  121. Lee, J.W.; Kim, M.N.; Kim, E.G.; Leem, J.S.; Baek, S.M.; Kim, M.J.; Kim, K.W.; Sohn, M.H. Chitinase 3-like 1 is involved in the induction of IL-8 expression by double-stranded RNA in airway epithelial cells. Biochem. Biophys. Res. Commun. 2022, 592, 106–112. [Google Scholar] [CrossRef]
  122. Hübner, K.; Karwelat, D.; Pietsch, E.; Beinborn, I.; Winterberg, S.; Bedenbender, K.; Benedikter, B.J.; Schmeck, B.; Vollmeister, E. NF-κB-mediated inhibition of microRNA-149-5p regulates Chitinase-3-like 1 expression in human airway epithelial cells. Cell. Signal. 2020, 67, 109498. [Google Scholar] [CrossRef]
  123. Fagan, K.A.; McMurtry, I.F.; Rodman, D.M. Role of endothelin-1 in lung disease. Respir. Res. 2001, 2, 90. [Google Scholar] [CrossRef]
  124. Lan, B.; Mitchel, J.A.; O’Sullivan, M.J.; Park, C.Y.; Kim, J.H.; Cole, W.C.; Butler, J.P.; Park, J.-A. Airway epithelial compression promotes airway smooth muscle proliferation and contraction. Am. J. Physiol. Lung Cell. Mol. Physiol. 2018, 315, L645–L652. [Google Scholar] [CrossRef]
  125. Chetty, A.; Andersson, S.; Lassus, P.; Nielsen, H.C. Insulin-like growth factor-1 (IGF-1) and IGF-1 receptor (IGF-1R) expression in human lung in RDS and BPD. Pediatr. Pulmonol. 2004, 37, 128–136. [Google Scholar] [CrossRef]
  126. Simões, F.B.; Kmit, A.; Amaral, M.D. Cross-talk of inflammatory mediators and airway epithelium reveals the cystic fibrosis transmembrane conductance regulator as a major target. ERJ Open Res. 2021, 7, 00247–02021. [Google Scholar] [CrossRef] [PubMed]
  127. Asokananthan, N.; Graham, P.T.; Fink, J.; Knight, D.A.; Bakker, A.J.; McWilliam, A.S.; Thompson, P.J.; Stewart, G.A. Activation of Protease-Activated Receptor (PAR)-1, PAR-2, and PAR-4 Stimulates IL-6, IL-8, and Prostaglandin E2 Release from Human Respiratory Epithelial Cells. J. Immunol. 2002, 168, 3577–3585. [Google Scholar] [CrossRef] [PubMed]
  128. Guo, F.H.; De Raeve, H.R.; Rice, T.W.; Stuehr, D.J.; Thunnissen, F.; Erzurum, S.C. Continuous nitric oxide synthesis by inducible nitric oxide synthase in normal human airway epithelium in vivo. Proc. Natl. Acad. Sci. USA 1995, 92, 7809–7813. [Google Scholar] [CrossRef]
  129. Levine, S.J. Bronchial epithelial cell-cytokine interactions in airway inflammation. J. Investig. Med. 1995, 43, 241–249. [Google Scholar]
  130. Suto, W.; Sakai, H.; Chiba, Y. Sustained exposure to prostaglandin D(2) augments the contraction induced by acetylcholine via a DP(1) receptor-mediated activation of p38 in bronchial smooth muscle of naive mice. J. Smooth Muscle Res. 2019, 55, 1–13. [Google Scholar] [CrossRef]
  131. Boussat, S.; Eddahibi, S.; Coste, A.; Fataccioli, V.; Gouge, M.; Housset, B.; Adnot, S.; Maitre, B. Expression and regulation of vascular endothelial growth factor in human pulmonary epithelial cells. Am. J. Physiol. Lung Cell. Mol. Physiol. 2000, 279, L371–L378. [Google Scholar] [CrossRef]
  132. Huether, S.E.; McCance, K.L. Pathophysiology: The biologic basis for disease in adults and children. Dimens. Crit. Care Nurs. 1994, 13, 315. [Google Scholar] [CrossRef]
  133. Gaona, I.P.; McCall, A.S.; Geis, N.M.; Colvard, A.C.; DiGiovanni, G.T.; Sherrill, T.P.; Singha, U.K.; Nichols, D.S.; Serezani, A.P.; David, H.E.; et al. Sustained Yap/Taz activation promotes aberrant alveolar epithelial cell differentiation and drives persistent fibrotic remodeling. bioRxiv 2025. [Google Scholar] [CrossRef]
  134. Mohri, Z.; Del Rio Hernandez, A.; Krams, R. The emerging role of YAP/TAZ in mechanotransduction. J. Thorac. Dis. 2017, 9, E507–E509. [Google Scholar] [CrossRef]
  135. John, A.E.; Wilson, M.R.; Habgood, A.; Porte, J.; Tatler, A.L.; Stavrou, A.; Miele, G.; Jolly, L.; Knox, A.J.; Takata, M.; et al. Loss of epithelial Gq and G11 signaling inhibits TGFβ production but promotes IL-33–mediated macrophage polarization and emphysema. Sci. Signal. 2016, 9, ra104. [Google Scholar] [CrossRef]
  136. Ni, K.; Che, B.; Gu, R.; Wang, C.; Pan, Y.; Li, J.; Liu, L.; Luo, M.; Deng, L. Single-Cell Hypertrophy Promotes Contractile Function of Cultured Human Airway Smooth Muscle Cells via Piezo1 and YAP Auto-Regulation. Cells 2024, 13, 1697. [Google Scholar] [CrossRef]
  137. Govorova, I.A.; Nikitochkina, S.Y.; Vorotelyak, E.A. Influence of intersignaling crosstalk on the intracellular localization of YAP/TAZ in lung cells. Cell Commun. Signal 2024, 22, 289. [Google Scholar] [CrossRef] [PubMed]
  138. Scheraga, R.G.; Southern, B.D.; Grove, L.M.; Olman, M.A. The Role of Transient Receptor Potential Vanilloid 4 in Pulmonary Inflammatory Diseases. Front. Immunol. 2017, 8, 503. [Google Scholar] [CrossRef] [PubMed]
  139. Liu, Y.; Li, J.; Chen, R.; Shi, F.; Xiong, Y. Airway epithelial cells promote in vitro airway smooth muscle cell proliferation by activating the Wnt/β-catenin pathway. Respir. Physiol. Neurobiol. 2025, 331, 104368. [Google Scholar] [CrossRef] [PubMed]
  140. Jenkins, R.G.; Su, X.; Su, G.; Scotton, C.J.; Camerer, E.; Laurent, G.J.; Davis, G.E.; Chambers, R.C.; Matthay, M.A.; Sheppard, D. Ligation of protease-activated receptor 1 enhances α v β 6 integrin–dependent TGF-β activation and promotes acute lung injury. J. Clin. Investig. 2006, 116, 1606–1614. [Google Scholar] [CrossRef]
  141. Alsubait, D.; Rajani, H.F.; Shan, L.; Koussih, L.; Halayko, A.J.; Lamkhioued, B.; Gounni, A.S. Expression of Semaphorin3E/PlexinD1 in human airway smooth muscle cells of patients with COPD. Am. J. Physiol. Lung Cell Mol. Physiol. 2024, 327, L831–L838. [Google Scholar] [CrossRef]
  142. Goldsmith, A.M.; Bentley, J.K.; Zhou, L.; Jia, Y.; Bitar, K.N.; Fingar, D.C.; Hershenson, M.B. Transforming growth factor-beta induces airway smooth muscle hypertrophy. Am. J. Respir. Cell Mol. Biol. 2006, 34, 247–254. [Google Scholar] [CrossRef]
  143. Zaleskas, J.M.; Kinner, B.; Freyman, T.M.; Yannas, I.V.; Gibson, L.J.; Spector, M. Growth factor regulation of smooth muscle actin expression and contraction of human articular chondrocytes and meniscal cells in a collagen-GAG matrix. Exp. Cell Res. 2001, 270, 21–31. [Google Scholar] [CrossRef]
  144. McMillan, S.J.; Xanthou, G.; Lloyd, C.M. Manipulation of Allergen-Induced Airway Remodeling by Treatment with Anti-TGF-β Antibody: Effect on the Smad Signaling Pathway. J. Immunol. 2005, 174, 5774–5780. [Google Scholar] [CrossRef]
  145. Postma, D.S.; Timens, W. Remodeling in asthma and chronic obstructive pulmonary disease. Proc. Am. Thorac. Soc. 2006, 3, 434–439. [Google Scholar] [CrossRef] [PubMed]
  146. Le, A.V.; Cho, J.Y.; Miller, M.; McElwain, S.; Golgotiu, K.; Broide, D.H. Inhibition of Allergen-Induced Airway Remodeling in Smad 3-Deficient Mice. J. Immunol. 2007, 178, 7310–7316. [Google Scholar] [CrossRef] [PubMed]
  147. Raeburn, D.; Rodger, I.W.; Hay, D.W.P.; Fedan, J.S. The dependence of airway smooth muscle on extracellular Ca2+ for contraction is influenced by the presence of cartilage. Life Sci. 1986, 38, 1499–1505. [Google Scholar] [CrossRef] [PubMed]
  148. Hay, D.W.; Farmer, S.G.; Raeburn, D.; Robinson, V.A.; Fleming, W.W.; Fedan, J.S. Airway epithelium modulates the reactivity of guinea-pig respiratory smooth muscle. Eur. J. Pharmacol. 1986, 129, 11–18. [Google Scholar] [CrossRef]
  149. Kılıç, A.; Ameli, A.; Park, J.-A.; Kho, A.T.; Tantisira, K.; Santolini, M.; Cheng, F.; Mitchel, J.A.; McGill, M.; O’Sullivan, M.J.; et al. Mechanical forces induce an asthma gene signature in healthy airway epithelial cells. Sci. Rep. 2020, 10, 966. [Google Scholar] [CrossRef]
  150. Mwase, C.; Phung, T.-K.N.; O’Sullivan, M.J.; Mitchel, J.A.; De Marzio, M.; Kılıç, A.; Weiss, S.T.; Fredberg, J.J.; Park, J.-A. Mechanical Compression of Human Airway Epithelial Cells Induces Release of Extracellular Vesicles Containing Tenascin C. Cells 2022, 11, 256. [Google Scholar] [CrossRef]
  151. Dunne, O.M.; Martin, S.L.; Sergeant, G.P.; McAuley, D.F.; O’Kane, C.M.; Button, B.; McGarvey, L.P.; Lundy, F.T. TRPV2 modulates mechanically Induced ATP Release from Human bronchial epithelial cells. Respir. Res. 2024, 25, 188. [Google Scholar] [CrossRef]
  152. Zhao, L.; Liang, Y.-T.; Tian, D.-B.; Zhang, R.-G.; Huang, J.; Zhu, Y.-X.; Zhou, W.-L.; Zhang, Y.-L. Regulation of smooth muscle contractility by the epithelium in rat tracheas: Role of prostaglandin E2 induced by the neurotransmitter acetylcholine. Ann. Transl. Med. 2021, 9, 313. [Google Scholar] [CrossRef]
  153. Jairaman, A.; Prakriya, M. Calcium Signaling in Airway Epithelial Cells: Current Understanding and Implications for Inflammatory Airway Disease. Arterioscler. Thromb. Vasc. Biol. 2024, 44, 772–783. [Google Scholar] [CrossRef]
  154. Yao, Y.; Zheng, M.; Borkar, N.A.; Thompson, M.A.; Zhang, E.Y.; Koloko Ngassie, M.L.; Wang, S.; Pabelick, C.M.; Vogel, E.R.; Prakash, Y.S. Role of STIM1 in stretch-induced signaling in human airway smooth muscle. Am. J. Physiol. Lung Cell. Mol. Physiol. 2024, 327, L150–L159. [Google Scholar] [CrossRef]
  155. Luo, M.; Ni, K.; Gu, R.; Qin, Y.; Guo, J.; Che, B.; Pan, Y.; Li, J.; Liu, L.; Deng, L. Chemical Activation of Piezo1 Alters Biomechanical Behaviors toward Relaxation of Cultured Airway Smooth Muscle Cells. Biol. Pharm. Bull. 2023, 46, 1–11. [Google Scholar] [CrossRef] [PubMed]
  156. Migulina, N.; Kelley, B.; Zhang, E.Y.; Pabelick, C.M.; Prakash, Y.S.; Vogel, E.R. Mechanosensitive Channels in Lung Health and Disease. Compr. Physiol. 2023, 13, 5157–5178. [Google Scholar] [CrossRef] [PubMed]
  157. Burgess, J.K.; Gosens, R. Mechanotransduction and the extracellular matrix: Key drivers of lung pathologies and drug responsiveness. Biochem. Pharmacol. 2024, 228, 116255. [Google Scholar] [CrossRef]
  158. McVicar, R.N.; Smith, E.; Melameka, M.; Bush, A.; Goetz, G.; Constantino, G.; Kumar, M.; Kwong, E.; Snyder, E.Y.; Leibel, S.L. iPSC-Derived Epithelial, Mesenchymal, Endothelial, and Immune Cell Co-Culture to Model Airway Barrier Integrity in Lung Health and Disease. J. Vis. Exp. 2024, 214, e67247. [Google Scholar] [CrossRef]
  159. Nishida, T.; Kubota, S.; Nakanishi, T.; Kuboki, T.; Yosimichi, G.; Kondo, S.; Takigawa, M. CTGF/Hcs24, a hypertrophic chondrocyte-specific gene product, stimulates proliferation and differentiation, but not hypertrophy of cultured articular chondrocytes. J. Cell Physiol. 2002, 192, 55–63. [Google Scholar] [CrossRef]
  160. Weksler, N.B.; Lunstrum, G.P.; Reid, E.S.; Horton, W.A. Differential effects of fibroblast growth factor (FGF) 9 and FGF2 on proliferation, differentiation and terminal differentiation of chondrocytic cells in vitro. Biochem. J. 1999, 342 Pt 3, 677–682. [Google Scholar] [CrossRef]
  161. Takeda, N.; Sumi, Y.; Préfontaine, D.; Abri, J.A.; Heialy, N.A.; Al-Ramli, W.; Michoud, M.C.; Martin, J.G.; Hamid, Q. Epithelium-derived chemokines induce airway smooth muscle cell migration. Clin. Exp. Allergy 2009, 39, 1018–1026. [Google Scholar] [CrossRef]
  162. Walker, F.; Kato, A.; Gonez, L.J.; Hibbs, M.L.; Pouliot, N.; Levitzki, A.; Burgess, A.W. Activation of the Ras/mitogen-activated protein kinase pathway by kinase-defective epidermal growth factor receptors results in cell survival but not proliferation. Mol. Cell. Biol. 1998, 18, 7192–7204. [Google Scholar] [CrossRef]
  163. Malavia, N.K.; Raub, C.B.; Mahon, S.B.; Brenner, M.; Jr, R.A.P.; George, S.C. Airway Epithelium Stimulates Smooth Muscle Proliferation. Am. J. Respir. Cell Mol. Biol. 2009, 41, 297–304. [Google Scholar] [CrossRef]
  164. Ramis, J.; Middlewick, R.; Pappalardo, F.; Cairns, J.T.; Stewart, I.D.; John, A.E.; Naveed, S.U.; Krishnan, R.; Miller, S.; Shaw, D.E.; et al. Lysyl oxidase like 2 is increased in asthma and contributes to asthmatic airway remodelling. Eur. Respir. J. 2022, 60, 2004361. [Google Scholar] [CrossRef]
  165. Sanchez-Adams, J.; Leddy, H.A.; McNulty, A.L.; O’Conor, C.J.; Guilak, F. The Mechanobiology of Articular Cartilage: Bearing the Burden of Osteoarthritis. Curr. Rheumatol. Rep. 2014, 16, 451. [Google Scholar] [CrossRef] [PubMed]
  166. Mauck, R.L.; Soltz, M.A.; Wang, C.C.B.; Wong, D.D.; Chao, P.-H.G.; Valhmu, W.B.; Hung, C.T.; Ateshian, G.A. Functional Tissue Engineering of Articular Cartilage Through Dynamic Loading of Chondrocyte-Seeded Agarose Gels. J. Biomech. Eng. 2000, 122, 252–260. [Google Scholar] [CrossRef] [PubMed]
  167. Fan, Y.; Wang, Y.; Wang, K. Prostaglandin E(2) stimulates normal bronchial epithelial cell growth through induction of c-Jun and PDK1, a kinase implicated in oncogenesis. Respir. Res. 2015, 16, 149. [Google Scholar] [CrossRef] [PubMed]
  168. Guzy, R.D.; Stoilov, I.; Elton, T.J.; Mecham, R.P.; Ornitz, D.M. Fibroblast Growth Factor 2 Is Required for Epithelial Recovery, but Not for Pulmonary Fibrosis, in Response to Bleomycin. Am. J. Respir. Cell Mol. Biol. 2015, 52, 116–128. [Google Scholar] [CrossRef]
  169. Gupta, P.; Markham, A.; Morgan, R.M. Ca2+ ion sequestration by guinea-pig tracheal cartilage: Its influence on trachealis reactivity to KCl. Br. J. Pharmacol. 1991, 104, 123–127. [Google Scholar] [CrossRef]
  170. Ramchandani, R.; Shen, X.; Elmsley, C.L.; Ambrosius, W.T.; Gunst, S.J.; Tepper, R.S. Differences in airway structure in immature and mature rabbits. J. Appl. Physiol. 2000, 89, 1310–1316. [Google Scholar] [CrossRef]
  171. Alber, A.B.; Marquez, H.A.; Ma, L.; Kwong, G.; Thapa, B.R.; Villacorta-Martin, C.; Lindstrom-Vautrin, J.; Bawa, P.; Wang, F.; Luo, Y.; et al. Directed differentiation of mouse pluripotent stem cells into functional lung-specific mesenchyme. Nat. Commun. 2023, 14, 3488. [Google Scholar] [CrossRef]
  172. Leibel, S.L.; McVicar, R.N.; Winquist, A.M.; Niles, W.D.; Snyder, E.Y. Generation of Complete Multi−Cell Type Lung Organoids From Human Embryonic and Patient-Specific Induced Pluripotent Stem Cells for Infectious Disease Modeling and Therapeutics Validation. Curr. Protoc. Stem Cell Biol. 2020, 54, e118. [Google Scholar] [CrossRef]
  173. Kishimoto, K.; Furukawa, K.T.; Luz-Madrigal, A.; Yamaoka, A.; Matsuoka, C.; Habu, M.; Alev, C.; Zorn, A.M.; Morimoto, M. Bidirectional Wnt signaling between endoderm and mesoderm confers tracheal identity in mouse and human cells. Nat. Commun. 2020, 11, 4159. [Google Scholar] [CrossRef]
  174. Bottasso-Arias, N.; Leesman, L.; Burra, K.; Snowball, J.; Shah, R.; Mohanakrishnan, M.; Xu, Y.; Sinner, D. BMP4 and Wnt signaling interact to promote mouse tracheal mesenchyme morphogenesis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2022, 322, L224–L242. [Google Scholar] [CrossRef]
  175. Luo, Y.; Cao, K.; Chiu, J.; Chen, H.; Wang, H.-J.; Thornton, M.E.; Grubbs, B.H.; Kolb, M.; Parmacek, M.S.; Mishina, Y.; et al. Defective mesenchymal Bmpr1a-mediated BMP signaling causes congenital pulmonary cysts. eLife 2024, 12, RP91876. [Google Scholar] [CrossRef]
  176. Pansky, B. Development Of The Lower Respiratory System: Larynx And Trachea. In Review of Medical Embryology; McGraw-Hill: Columbus, OH, USA, 1982. [Google Scholar]
  177. Mailleux, A.A.; Kelly, R.; Veltmaat, J.M.; De Langhe, S.P.; Zaffran, S.; Thiery, J.P.; Bellusci, S. Fgf10 expression identifies parabronchial smooth muscle cell progenitors and is required for their entry into the smooth muscle cell lineage. Development 2005, 132, 2157–2166. [Google Scholar] [CrossRef]
  178. Pansky, B. Development Of The Lower Respiratory System: The Bronchi And Surrounding Structures. In Review of Medical Embryology; McGraw-Hill: Columbus, OH, USA, 1982; Volume 59. [Google Scholar]
  179. Bellusci, S.; Grindley, J.; Emoto, H.; Itoh, N.; Hogan, B.L. Fibroblast growth factor 10 (FGF10) and branching morphogenesis in the embryonic mouse lung. Development 1997, 124, 4867–4878. [Google Scholar] [CrossRef] [PubMed]
  180. Jesudason, E.C.; Smith, N.P.; Connell, M.G.; Spiller, D.G.; White, M.R.; Fernig, D.G.; Losty, P.D. Peristalsis of airway smooth muscle is developmentally regulated and uncoupled from hypoplastic lung growth. Am. J. Physiol. Lung Cell Mol. Physiol. 2006, 291, L559–L565. [Google Scholar] [CrossRef] [PubMed]
  181. Parvez, O.; Voss, A.-M.; de Kok, M.; Roth-Kleiner, M.; Belik, J. Bronchial Muscle Peristaltic Activity in the Fetal Rat. Pediatr. Res. 2006, 59, 756. [Google Scholar] [CrossRef] [PubMed]
  182. Cairns, D.M.; Lee, P.G.; Uchimura, T.; Seufert, C.R.; Kwon, H.; Zeng, L. The role of muscle cells in regulating cartilage matrix production. J. Orthop. Res. 2010, 28, 529–536. [Google Scholar] [CrossRef]
  183. Shannon, J.M.; Nielsen, L.D.; Gebb, S.A.; Randell, S.H. Mesenchyme specifies epithelial differentiation in reciprocal recombinants of embryonic lung and trachea. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 1998, 212, 482–494. [Google Scholar] [CrossRef]
  184. Schittny, J.C.; Miserocchi, G.; Sparrow, M.P. Spontaneous Peristaltic Airway Contractions Propel Lung Liquid through the Bronchial Tree of Intact and Fetal Lung Explants. Am. J. Respir. Cell Mol. Biol. 2000, 23, 11–18. [Google Scholar] [CrossRef]
  185. Featherstone, N.C.; Jesudason, E.C.; Connell, M.G.; Fernig, D.G.; Wray, S.; Losty, P.D.; Burdyga, T.V. Spontaneous propagating calcium waves underpin airway peristalsis in embryonic rat lung. Am. J. Respir. Cell Mol. Biol. 2005, 33, 153–160. [Google Scholar] [CrossRef]
  186. Jesudason, E.C. Airway smooth muscle: An architect of the lung? Thorax 2009, 64, 541–545. [Google Scholar] [CrossRef]
  187. Perez-Zoghbi, J.F.; Bai, Y.; Sanderson, M.J. Nitric oxide induces airway smooth muscle cell relaxation by decreasing the frequency of agonist-induced Ca2+ oscillations. J. Gen. Physiol. 2010, 135, 247–259. [Google Scholar] [CrossRef]
  188. Sherman, T.S.; Chen, Z.; Yuhanna, I.S.; Lau, K.S.; Margraf, L.R.; Shaul, P.W. Nitric oxide synthase isoform expression in the developing lung epithelium. Am. J. Physiol. 1999, 276 Pt 1, L383–L390. [Google Scholar] [CrossRef]
  189. Halayko, A.J.; Salari, H.; MA, X.; Stephens, N.L. Markers of airway smooth muscle cell phenotype. Am. J. Physiol. Lung Cell. Mol. Physiol. 1996, 270, L1040–L1051. [Google Scholar] [CrossRef]
  190. Lan, R.S.; Knight, D.A.; Stewart, G.A.; Henry, P.J. Role of PGE2 in protease-activated receptor-1, −2 and −4 mediated relaxation in the mouse isolated trachea. Br. J. Pharmacol. 2001, 132, 93–100. [Google Scholar] [CrossRef]
  191. Barnett, K.; Jacoby, D.B.; Nadel, J.A.; Lazarus, S.C. The effects of epithelial cell supernatant on contractions of isolated canine tracheal smooth muscle. Am. Rev. Respir. Dis. 1988, 138, 780–783. [Google Scholar] [CrossRef] [PubMed]
  192. Panitch, H.B.; Wolfson, M.R.; Shaffer, T.H. Epithelial modulation of preterm airway smooth muscle contraction. J. Appl. Physiol. 1993, 74, 1437. [Google Scholar] [CrossRef] [PubMed]
  193. Barnes, P.J.; Cuss, F.M.; Palmer, J.B. The effect of airway epithelium on smooth muscle contractility in bovine trachea. Br. J. Pharmacol. 1985, 86, 685–691. [Google Scholar] [CrossRef] [PubMed]
  194. Sonnylal, S.; Xu, S.; Jones, H.; Tam, A.; Sreeram, V.R.; Ponticos, M.; Norman, J.; Agrawal, P.; Abraham, D.; de Crombrugghe, B. Connective tissue growth factor causes EMT-like cell fate changes in vivo and in vitro. J. Cell Sci. 2013, 126, 2164–2175. [Google Scholar] [CrossRef]
  195. Câmara, J.; Jarai, G. Epithelial-mesenchymal transition in primary human bronchial epithelial cells is Smad-dependent and enhanced by fibronectin and TNF-α. Fibrogenes. Tissue Repair. 2010, 3, 2. [Google Scholar] [CrossRef]
  196. Gospodarowicz, D.; Moran, J.S. Mitogenic effect of fibroblast growth factor on early passage cultures of human and murine fibroblasts. J. Cell Biol. 1975, 66, 451–457. [Google Scholar] [CrossRef]
  197. Aros, C.J.; Pantoja, C.J.; Gomperts, B.N. Wnt signaling in lung development, regeneration, and disease progression. Commun. Biol. 2021, 4, 601. [Google Scholar] [CrossRef] [PubMed]
  198. Eenjes, E.; Tibboel, D.; Wijnen, R.M.H.; Rottier, R.J. Lung epithelium development and airway regeneration. Front. Cell Dev. Biol. 2022, 10, 1022457. [Google Scholar] [CrossRef] [PubMed]
  199. Hu, Y.; Ciminieri, C.; Hu, Q.; Lehmann, M.; Königshoff, M.; Gosens, R. WNT Signalling in Lung Physiology and Pathology. Handb. Exp. Pharmacol. 2021, 269, 305–336. [Google Scholar] [CrossRef]
  200. Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef]
  201. Lin, L.; Yang, L.; Wang, N.; Chen, S.; Du, X.; Chen, R.; Zhang, H.; Kong, X. FGF10 protects against LPS-induced epithelial barrier injury and inflammation by inhibiting SIRT1-ferroptosis pathway in acute lung injury in mice. Int. Immunopharmacol. 2024, 127, 111426. [Google Scholar] [CrossRef]
  202. Peng, W.; Song, Y.; Zhu, G.; Zeng, Y.; Cai, H.; Lu, C.; Abuduxukuer, Z.; Song, X.; Gao, X.; Ye, L.; et al. FGF10 attenuates allergic airway inflammation in asthma by inhibiting PI3K/AKT/NF-κB pathway. Cell Signal 2024, 113, 110964. [Google Scholar] [CrossRef]
  203. Ma, Q.; Ma, Y.; Dai, X.; Ren, T.; Fu, Y.; Liu, W.; Han, Y.; Wu, Y.; Cheng, Y.; Zhang, T.; et al. Regeneration of functional alveoli by adult human SOX9(+) airway basal cell transplantation. Protein Cell 2018, 9, 267–282. [Google Scholar] [CrossRef]
  204. Sun, D.; Llora Batlle, O.; van den Ameele, J.; Thomas, J.C.; He, P.; Lim, K.; Tang, W.; Xu, C.; Meyer, K.B.; Teichmann, S.A.; et al. SOX9 maintains human foetal lung tip progenitor state by enhancing WNT and RTK signalling. Embo J. 2022, 41, e111338. [Google Scholar] [CrossRef]
  205. Yang, J.; Li, Y.; Huang, Y.; Chen, H.; Sui, P. Unlocking lung regeneration: Insights into progenitor cell dynamics and metabolic control. Cell Regen. 2024, 13, 31. [Google Scholar] [CrossRef]
  206. Osei, E.T.; Booth, S.; Hackett, T.-L. What Have In Vitro Co-Culture Models Taught Us about the Contribution of Epithelial-Mesenchymal Interactions to Airway Inflammation and Remodeling in Asthma? Cells 2020, 9, 1694. [Google Scholar] [CrossRef] [PubMed]
  207. Goodwin, K.; Jaslove, J.M.; Tao, H.; Zhu, M.; Hopyan, S.; Nelson, C.M. Patterning the embryonic pulmonary mesenchyme. iScience 2022, 25, 103838. [Google Scholar] [CrossRef]
  208. Uwagboe, I.E.; Mumby, S.; Dunlop, I.E.; Adcock, I.M. Does mechanobiology drive respiratory disease? Biomechanical induction of mucus hypersecretion in human bronchial organoids using a photocontrolled biomaterial gel. bioRxiv 2025. [Google Scholar] [CrossRef]
  209. Al Yazeedi, S.; Guo, T.J.F.; Sohd, J.; Abokor, F.A.; Baher, J.Z.; Yee, L.; Cheung, C.; Sin, D.D.; Osei, E.T. Dynamic mechanical stimulation of alveolar epithelial-fibroblast models using the Flexcell tension system to study of lung disease mechanisms. Front. Med. 2025, 12, 1552803. [Google Scholar] [CrossRef] [PubMed]
  210. Guo, Y.; Zhou, Y.; Wang, R.; Lin, Y.; Lan, H.; Li, Y.; Wang, D.-Y.; Dong, J.; Li, K.; Yan, Y.; et al. YAP as a potential therapeutic target for myofibroblast formation in asthma. Respir. Res. 2025, 26, 51. [Google Scholar] [CrossRef]
  211. Pan, J.; Luk, C.; Kent, G.; Cutz, E.; Yeger, H. Pulmonary Neuroendocrine Cells, Airway Innervation, and Smooth Muscle Are Altered in Cftr Null Mice. Am. J. Respir. Cell Mol. Biol. 2006, 35, 320–326. [Google Scholar] [CrossRef]
  212. Bonvin, E.; Le Rouzic, P.; Bernaudin, J.F.; Cottart, C.H.; Vandebrouck, C.; Crie, A.; Leal, T.; Clement, A.; Bonora, M. Congenital tracheal malformation in cystic fibrosis transmembrane conductance regulator-deficient mice. J. Physiol. 2008, 586, 3231–3243. [Google Scholar] [CrossRef]
  213. Meyerholz, D.K.; Stoltz, D.A.; Namati, E.; Ramachandran, S.; Pezzulo, A.A.; Smith, A.R.; Rector, M.V.; Suter, M.J.; Kao, S.; McLennan, G.; et al. Loss of cystic fibrosis transmembrane conductance regulator function produces abnormalities in tracheal development in neonatal pigs and young children. Am. J. Respir. Crit. Care Med. 2010, 182, 1251–1261. [Google Scholar] [CrossRef]
  214. Haraguchi, M.; Shimura, S.; Shirato, K. Morphometric Analysis of Bronchial Cartilage in Chronic Obstructive Pulmonary Disease and Bronchial Asthma. Am. J. Respir. Crit. Care Med. 1999, 159, 1005–1013. [Google Scholar] [CrossRef]
  215. Carroll, N.; Elliot, J.; Morton, A.; James, A. The Structure of Large and Small Airways in Nonfatal and Fatal Asthma. Am. Rev. Respir. Dis. 1993, 147, 405–410. [Google Scholar] [CrossRef]
  216. Regamey, N.; Ochs, M.; Hilliard, T.N.; Mühlfeld, C.; Cornish, N.; Fleming, L.; Saglani, S.; Alton, E.W.F.W.; Bush, A.; Jeffery, P.K.; et al. Increased Airway Smooth Muscle Mass in Children with Asthma, Cystic Fibrosis, and Non-Cystic Fibrosis Bronchiectasis. Am. J. Respir. Crit. Care Med. 2008, 177, 837–843. [Google Scholar] [CrossRef]
  217. Ma, X.; Cheng, Z.; Kong, H.; Wang, Y.; Unruh, H.; Stephens, N.L.; Laviolette, M. Changes in biophysical and biochemical properties of single bronchial smooth muscle cells from asthmatic subjects. Am. J. Physiol. Lung Cell Mol. Physiol. 2002, 283, L1181–L1189. [Google Scholar] [CrossRef]
  218. Matsumoto, H.; Moir, L.M.; Oliver, B.G.; Burgess, J.K.; Roth, M.; Black, J.L.; McParland, B.E. Comparison of gel contraction mediated by airway smooth muscle cells from patients with and without asthma. Thorax 2007, 62, 848–854. [Google Scholar] [CrossRef] [PubMed]
  219. Naylor, B. The Shedding of the Mucosa of the Bronchial Tree in Asthma. Thorax 1962, 17, 69–72. [Google Scholar] [CrossRef] [PubMed]
  220. Wiesner, D.L.; Merkhofer, R.M.; Ober, C.; Kujoth, G.C.; Niu, M.; Keller, N.P.; Gern, J.E.; Brockman-Schneider, R.A.; Evans, M.D.; Jackson, D.J.; et al. Club Cell TRPV4 Serves as a Damage Sensor Driving Lung Allergic Inflammation. Cell Host Microbe 2020, 27, 614–628.e616. [Google Scholar] [CrossRef] [PubMed]
  221. Clark, C.A.; Li, T.-F.; Kim, K.-O.; Drissi, H.; Zuscik, M.J.; Zhang, X.; O’Keefe, R.J. Prostaglandin E2 inhibits BMP signaling and delays chondrocyte maturation. J. Orthop. Res. 2009, 27, 785–792. [Google Scholar] [CrossRef]
  222. Kinner, B.; Spector, M. Smooth muscle actin expression by human articular chondrocytes and their contraction of a collagen—Glycosaminoglycan matrix in vitro. J. Orthop. Res. 2001, 19, 233–241. [Google Scholar] [CrossRef]
  223. Marlovits, S.; Hombauer, M.; Truppe, M.; Vecsei, V.; Schlegel, W. Changes in the ratio of type-I and type-II collagen expression during monolayer culture of human chondrocytes. Bone Jt. J. 2004, 86, 286–295. [Google Scholar] [CrossRef]
  224. Parreno, J.; Raju, S.; Wu, P.-H.; Kandel, R.A. MRTF-A signaling regulates the acquisition of the contractile phenotype in dedifferentiated chondrocytes. Matrix Biol. 2017, 62, 3–14. [Google Scholar] [CrossRef]
  225. Zhong, B.; Du, J.; Liu, F.; Sun, S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm 2025, 6, e70128. [Google Scholar] [CrossRef]
  226. Kim, K.W.; Jee, H.M.; Park, Y.H.; Choi, B.S.; Sohn, M.H.; Kim, K.E. Relationship between amphiregulin and airway inflammation in children with asthma and eosinophilic bronchitis. Chest 2009, 136, 805–810. [Google Scholar] [CrossRef]
  227. Benayoun, L.; Druilhe, A.; Dombret, M.-C.; Aubier, M.; Pretolani, M. Airway Structural Alterations Selectively Associated with Severe Asthma. Am. J. Respir. Crit. Care Med. 2003, 167, 1360–1368. [Google Scholar] [CrossRef]
  228. Jang, J.-H.; Chand, H.S.; Bruse, S.; Doyle-Eisele, M.; Royer, C.; McDonald, J.; Qualls, C.; Klingelhutz, A.J.; Lin, Y.; Mallampalli, R.; et al. Connective Tissue Growth Factor Promotes Pulmonary Epithelial Cell Senescence and Is Associated with COPD Severity. J. Chronic Obstr. Pulm. Dis. 2017, 14, 228–237. [Google Scholar] [CrossRef]
  229. Chung, K.F. The Role of Airway Smooth Muscle in the Pathogenesis of Airway Wall Remodeling in Chronic Obstructive Pulmonary Disease. Proc. Am. Thorac. Soc. 2005, 2, 347–354. [Google Scholar] [CrossRef] [PubMed]
  230. López-Posadas, R.; Bagley, D.C.; Pardo-Pastor, C.; Ortiz-Zapater, E. The epithelium takes the stage in asthma and inflammatory bowel diseases. Front. Cell Dev. Biol. 2024, 12, 1258859. [Google Scholar] [CrossRef] [PubMed]
  231. Hicks-Berthet, J.; Ning, B.; Federico, A.; Tilston-Lunel, A.; Matschulat, A.; Ai, X.; Lenburg, M.E.; Beane, J.; Monti, S.; Varelas, X. Yap/Taz inhibit goblet cell fate to maintain lung epithelial homeostasis. Cell Rep. 2021, 36, 109347. [Google Scholar] [CrossRef] [PubMed]
  232. Chamorro-Herrero, I.; Zambrano, A. Modeling of Respiratory Diseases Evolving with Fibrosis from Organoids Derived from Human Pluripotent Stem Cells. Int. J. Mol. Sci. 2023, 24, 4413. [Google Scholar] [CrossRef]
  233. Lama, V.N.; Smith, L.; Badri, L.; Flint, A.; Andrei, A.-C.; Murray, S.; Wang, Z.; Liao, H.; Toews, G.B.; Krebsbach, P.H.; et al. Evidence for tissue-resident mesenchymal stem cells in human adult lung from studies of transplanted allografts. J. Clin. Investig. 2007, 117, 989–996. [Google Scholar] [CrossRef]
  234. Akram, K.M.; Patel, N.; Spiteri, M.A.; Forsyth, N.R. Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. Int. J. Mol. Sci. 2016, 17, 128. [Google Scholar] [CrossRef]
  235. Summer, R.; Fitzsimmons, K.; Dwyer, D.; Murphy, J.; Fine, A. Isolation of an adult mouse lung mesenchymal progenitor cell population. Am. J. Respir. Cell Mol. Biol. 2007, 37, 152–159. [Google Scholar] [CrossRef]
  236. Martin, J.; Helm, K.; Ruegg, P.; Varella-Garcia, M.; Burnham, E.; Majka, S. Adult lung side population cells have mesenchymal stem cell potential. Cytotherapy 2008, 10, 140–151. [Google Scholar] [CrossRef]
  237. Jarvinen, L.; Badri, L.; Wettlaufer, S.; Ohtsuka, T.; Standiford, T.J.; Toews, G.B.; Pinsky, D.J.; Peters-Golden, M.; Lama, V.N. Lung Resident Mesenchymal Stem Cells Isolated From Human Lung Allografts Inhibit T Cell Proliferation via a Soluble Mediator. J. Immunol. 2008, 181, 4389–4396. [Google Scholar] [CrossRef] [PubMed]
  238. Tong, L.; Zhou, J.; Rong, L.; Seeley, E.J.; Pan, J.; Zhu, X.; Liu, J.; Wang, Q.; Tang, X.; Qu, J.; et al. Fibroblast Growth Factor-10 (FGF-10) Mobilizes Lung-resident Mesenchymal Stem Cells and Protects Against Acute Lung Injury. Sci. Rep. 2016, 6, 21642. [Google Scholar] [CrossRef] [PubMed]
  239. Le Visage, C.; Dunham, B.; Flint, P.; Leong, K.W. Coculture of mesenchymal stem cells and respiratory epithelial cells to engineer a human composite respiratory mucosa. Tissue Eng. 2004, 10, 1426–1435. [Google Scholar] [CrossRef]
  240. Li, X.; Zhang, Y.; Yeung, S.C.; Liang, Y.; Liang, X.; Ding, Y.; Ip, M.S.M.; Tse, H.-F.; Mak, J.C.W.; Lian, Q. Mitochondrial Transfer of Induced Pluripotent Stem Cell–Derived Mesenchymal Stem Cells to Airway Epithelial Cells Attenuates Cigarette Smoke–Induced Damage. Am. J. Respir. Cell Mol. Biol. 2014, 51, 455–465. [Google Scholar] [CrossRef]
  241. Serikov, V.B.; Popov, B.; Mikhailov, V.M.; Gupta, N.; Matthay, M.A. Evidence of Temporary Airway Epithelial Repopulation and Rare Clonal Formation by BM-derived Cells Following Naphthalene Injury in Mice. Anat. Rec. Adv. Integr. Anat. Evol. Biol. 2007, 290, 1033–1045. [Google Scholar] [CrossRef]
  242. Urbanek, K.; De Angelis, A.; Spaziano, G.; Piegari, E.; Matteis, M.; Cappetta, D.; Esposito, G.; Russo, R.; Tartaglione, G.; De Palma, R.; et al. Intratracheal Administration of Mesenchymal Stem Cells Modulates Tachykinin System, Suppresses Airway Remodeling and Reduces Airway Hyperresponsiveness in an Animal Model. PLoS ONE 2016, 11, e0158746. [Google Scholar] [CrossRef]
  243. Taillé, C.; Almolki, A.; Benhamed, M.; Zedda, C.; Mégret, J.; Berger, P.; Lesèche, G.; Fadel, E.; Yamaguchi, T.; Marthan, R. Heme oxygenase inhibits human airway smooth muscle proliferation via a bilirubin-dependent modulation of ERK1/2 phosphorylation. J. Biol. Chem. 2003, 278, 27160–27168. [Google Scholar] [CrossRef]
  244. Marinas-Pardo, L.; Mirones, I.; Amor-Carro, O.; Fraga-Iriso, R.; Lema-Costa, B.; Cubillo, I.; Rodriguez Milla, M.A.; Garcia-Castro, J.; Ramos-Barbon, D. Mesenchymal stem cells regulate airway contractile tissue remodeling in murine experimental asthma. Allergy 2014, 69, 730–740. [Google Scholar] [CrossRef]
  245. Cooke, M.; Allon, A.; Cheng, T.; Kuo, A.; Kim, H.; Vail, T.; Marcucio, R.; Schneider, R.; Lotz, J.; Alliston, T. Structured three-dimensional co-culture of mesenchymal stem cells with chondrocytes promotes chondrogenic differentiation without hypertrophy. Osteoarthr. Cartil. 2011, 19, 1210–1218. [Google Scholar] [CrossRef]
  246. Bian, L.; Zhai, D.Y.; Mauck, R.L.; Burdick, J.A. Coculture of Human Mesenchymal Stem Cells and Articular Chondrocytes Reduces Hypertrophy and Enhances Functional Properties of Engineered Cartilage. Tissue Eng. Part A 2011, 17, 1137–1145. [Google Scholar] [CrossRef] [PubMed]
  247. Kim, Y.-J.; Kim, H.-J.; Im, G.-I. PTHrP promotes chondrogenesis and suppresses hypertrophy from both bone marrow-derived and adipose tissue-derived MSCs. Biochem. Biophys. Res. Commun. 2008, 373, 104–108. [Google Scholar] [CrossRef] [PubMed]
  248. Fischer, J.; Dickhut, A.; Rickert, M.; Richter, W. Human articular chondrocytes secrete parathyroid hormone–related protein and inhibit hypertrophy of mesenchymal stem cells in coculture during chondrogenesis. Arthritis Rheum. 2010, 62, 2696–2706. [Google Scholar] [CrossRef] [PubMed]
  249. Meretoja, V.V.; Dahlin, R.L.; Kasper, F.K.; Mikos, A.G. Enhanced Chondrogenesis in Co-Cultures with Articular Chondrocytes and Mesenchymal Stem Cells. Biomaterials 2012, 33, 6362–6369. [Google Scholar] [CrossRef] [PubMed]
  250. Qing, C.; Wei-ding, C.; Wei-min, F. Co-culture of chondrocytes and bone marrow mesenchymal stem cells in vitro enhances the expression of cartilaginous extracellular matrix components. Braz. J. Med. Biol. Res. 2011, 44, 303–310. [Google Scholar] [CrossRef]
  251. Tsuchiya, K.; Chen, G.; Ushida, T.; Matsuno, T.; Tateishi, T. The effect of coculture of chondrocytes with mesenchymal stem cells on their cartilaginous phenotype in vitro. Mater. Sci. Eng. C 2004, 24, 391–396. [Google Scholar] [CrossRef]
  252. Marin, A.E. Repopulation of De-Epithelialized Tracheal Grafts; University of Toronto: Toronto, ON, Canada, 2019. [Google Scholar]
  253. Zhou, Q.; Saijo, Y. Chapter 6—Induced pluripotent stem cells for trachea engineering. In iPSCs in Tissue Engineering; Birbrair, A., Ed.; Academic Press: Cambridge, MA, USA, 2021; pp. 143–165. [Google Scholar]
  254. West, A.R.; Osagie, J.; Syeda, S.; Guimond, M.; Parrrenas, L.; Haroon, A.; Imaseun, P.; Turner-Brannen, E. Development of a 3D Bioprinted Airway Smooth Muscle Model for Manipulating Structure and Measuring Contraction. In C109. Even Better Than the Real Thing: Advanced Models of Lung Disease; American Thoracic Society: New York, NY, USA, 2023; Volume 207, p. A6174. [Google Scholar]
  255. Kwong, G.; Marquez, H.A.; Yang, C.; Wong, J.Y.; Kotton, D.N. Generation of a Purified iPSC-Derived Smooth Muscle-like Population for Cell Sheet Engineering. Stem Cell Rep. 2019, 13, 499–514. [Google Scholar] [CrossRef]
  256. Varma, R.; Marin-Araujo, A.E.; Rostami, S.; Waddell, T.K.; Karoubi, G.; Haykal, S. Short-Term Preclinical Application of Functional Human Induced Pluripotent Stem Cell-Derived Airway Epithelial Patches. Adv. Heal. Mater. 2021, 10, e2100957. [Google Scholar] [CrossRef]
  257. Noel, F.E.E.; Karamaoun, C.; Dempsey, J.A.; Mauroy, B. How mammals adapt their breath to body activity—And how this depends on body size. Peer Commun. Math. Comput. Biol. 2020, 1, 100005. [Google Scholar]
  258. Basil, M.C.; Morrisey, E.E. Lung regeneration: A tale of mice and men. Semin. Cell Dev. Biol. 2020, 100, 88–100. [Google Scholar] [CrossRef]
  259. Noël, F.; Karamaoun, C.; Demsey, J.; Mauroy, B. The origin of the allometric scaling of lung ventilation in mammals. Peer Commun. J. 2022, 2, e2. [Google Scholar] [CrossRef]
  260. Li, Y.; Xu, G.K. Editorial: Mechanobiology at multiple scales. Front. Bioeng. Biotechnol. 2023, 11, 1226198. [Google Scholar] [CrossRef]
  261. Nayak, P.S.; Wang, Y.; Najrana, T.; Priolo, L.M.; Rios, M.; Shaw, S.K.; Sanchez-Esteban, J. Mechanotransduction via TRPV4 regulates inflammation and differentiation in fetal mouse distal lung epithelial cells. Respir. Res. 2015, 16, 60. [Google Scholar] [CrossRef]
  262. Castro, M.G.B.; Varble, N.A.; Yung, R.C.; Wood, B.J.; Karanian, J.W.; Pritchard, W.F. In Vivo Characterization of the Swine Airway Morphometry and Motion Based on Computed Tomographic Imaging During Respiration. J. Biomech. Eng. 2020, 142, 121009. [Google Scholar] [CrossRef] [PubMed]
  263. Mariano, C.A.; Sattari, S.; Maghsoudi-Ganjeh, M.; Tartibi, M.; Lo, D.D.; Eskandari, M. Novel Mechanical Strain Characterization of Ventilated ex vivo Porcine and Murine Lung using Digital Image Correlation. Front. Physiol. 2020, 11, 600492. [Google Scholar] [CrossRef] [PubMed]
  264. Parzianello Egúsquiza, M.G.; Otsuki, D.A.; Costa Auler Junior, J.O. Ex Vivo Porcine Experimental Model for Studying and Teaching Lung Mechanics. J. Vis. Exp. 2024, 206, e64850. [Google Scholar] [CrossRef] [PubMed]
  265. Chen, P.; Van Hassel, J.; Pinezich, M.R.; Diane, M.; Hudock, M.R.; Kaslow, S.R.; Gavaudan, O.P.; Fung, K.; Kain, M.L.; Lopez, H., 2nd; et al. Recovery of extracorporeal lungs using cross-circulation with injured recipient swine. J. Thorac. Cardiovasc. Surg. 2024, 167, e106–e130. [Google Scholar] [CrossRef]
  266. Mia, M.M.; Selvan, A.; Nilanthi, U.; Singh, M.K. YAP/TAZ activation in fibroblasts coordinates fibrotic remodeling, fibroinflammation, and epithelial dysfunction in pulmonary fibrosis. bioRxiv 2025. [Google Scholar] [CrossRef]
  267. Novak, C.; Ballinger, M.N.; Ghadiali, S. Mechanobiology of Pulmonary Diseases: A Review of Engineering Tools to Understand Lung Mechanotransduction. J. Biomech. Eng. 2021, 143, 110801. [Google Scholar] [CrossRef]
  268. Madissoon, E.; Oliver, A.; Kleshchevnikov, V.; Wilbrey-Clark, A.; Polnski, K.; Mamanova, L.; Bolt, L.; Pett, P.; Dabrowska, M.; Tuck, L.; et al. The multi-omics spatial lung atlas reveales new cell states and their functions in airway mesenchyme. ERJ Open Res. 2022, 8, 260. [Google Scholar]
  269. Megas, S.; Wilbrey-Clark, A.; Maartens, A.; Teichmann, S.A.; Meyer, K.B. Spatial Transcriptomics of the Respiratory System. Annu. Rev. Physiol. 2025, 87, 447–470. [Google Scholar] [CrossRef]
  270. Ma, S.; Wang, W.; Zhou, J.; Liao, S.; Hai, C.; Hou, Y.; Zhou, Z.; Wang, Z.; Su, Y.; Zhu, Y.; et al. Lamination-based organoid spatially resolved transcriptomics technique for primary lung and liver organoid characterization. Proc. Natl. Acad. Sci. USA 2024, 121, e2408939121. [Google Scholar] [CrossRef]
Figure 1. Summary of interaction of airway cells in normal physiology via paracrine signalling, cell–cell interaction, and mechanotransduction cascades. Legend: green upward arrow—upregulation; horizontal flat arrowhead—inhibition; check symbol—enabling function.
Figure 1. Summary of interaction of airway cells in normal physiology via paracrine signalling, cell–cell interaction, and mechanotransduction cascades. Legend: green upward arrow—upregulation; horizontal flat arrowhead—inhibition; check symbol—enabling function.
Arm 93 00051 g001
Figure 2. General mechanotransduction dynamics in the airways. Epithelium-induced ASM contraction coming from external stimuli such as particulates from inspired air or epithelial damage cascades throughout the airways by release of functional molecules that act as a stimulant to the ASMCs. Cyclic contraction or relaxation of ASM due to epithelial-derived agonists affects the whole organ in which a compressive force is induced, initiating ECM production in chondrocytes that both serves as a Ca2+ ion reservoir for modulation of ASM activity and as a scaffold on basal epithelial cells on the interface of cartilage and epithelium, where epithelial cells differentiate further into a pseudostratified epithelium with diverse phenotypes. Cyclic compressive force also increases ciliary beat frequency of AECs and ATP release, enhancing mucous clearance and inducing epithelial repair.
Figure 2. General mechanotransduction dynamics in the airways. Epithelium-induced ASM contraction coming from external stimuli such as particulates from inspired air or epithelial damage cascades throughout the airways by release of functional molecules that act as a stimulant to the ASMCs. Cyclic contraction or relaxation of ASM due to epithelial-derived agonists affects the whole organ in which a compressive force is induced, initiating ECM production in chondrocytes that both serves as a Ca2+ ion reservoir for modulation of ASM activity and as a scaffold on basal epithelial cells on the interface of cartilage and epithelium, where epithelial cells differentiate further into a pseudostratified epithelium with diverse phenotypes. Cyclic compressive force also increases ciliary beat frequency of AECs and ATP release, enhancing mucous clearance and inducing epithelial repair.
Arm 93 00051 g002
Figure 3. Signalling pathways in the developing airways. Key functional molecules are Sox9 (cartilage formation), FGF-10 (epithelial branching), and FGF-9 (ASMC differentiation).
Figure 3. Signalling pathways in the developing airways. Key functional molecules are Sox9 (cartilage formation), FGF-10 (epithelial branching), and FGF-9 (ASMC differentiation).
Arm 93 00051 g003
Figure 4. Anatomy of pathologies in bronchus. Inflammation, mucous overexcretion, and hypercontractility of the ASM are few of the pathologies seen in the trachea and bronchi.
Figure 4. Anatomy of pathologies in bronchus. Inflammation, mucous overexcretion, and hypercontractility of the ASM are few of the pathologies seen in the trachea and bronchi.
Arm 93 00051 g004
Table 2. Common airway construct compositions and associated failure modes.
Table 2. Common airway construct compositions and associated failure modes.
Scaffold TypeTypical
Cellularization
Key LimitationsReferences
Synthetic polymer scaffolds (PCL, PLA)Epithelial cells, MSCs,
chondrocytes
Variable epithelial
coverage; mechanical
mismatch causing stenosis
[252,253]
Decellularized
tracheal grafts
Epithelial cells + MSCs/
chondrocytes
Higher stenosis rates with single-cell seeding; contamination risk[252,253]
3D-bioprinted
hydrogels
Airway epithelial progenitors, MSC-derived SMCsMechanical collapse under contractile load; immature SMC phenotype[254,255]
Composite
biomaterials
(silk-collagen)
hiPSC-derived
epithelial patches
Short-term integration only; long-term durability unproven[256]
Table 3. Comparative analysis of engineering strategies.
Table 3. Comparative analysis of engineering strategies.
Cell Selection and Differentiation
AspectTraditional StrategyMechanotransduction-Informed Strategy
Cell SourcesPrimary cells, basic iPSC differentiationiPSCs engineered for enhanced mechanotransduction (TRPV4, YAP/TAZ)
Differentiation GoalsAchieve cell type identityAchieve functional mechanotransduction networks
Quality MetricsCell viability, marker expressionMechanotransduction pathway
activity, cellular specialisation roles
Scaffold Design Philosophy
AspectTraditional StrategyMechanotransduction-Informed Strategy
Material
Selection
Biocompatible polymers,
decellularized matrices
Biomimetic stiffness gradients
(2–50 kPa)
Mechanical
Properties
Uniform stiffness matching native tissueSpatially varied stiffness to guide YAP/TAZ activation
Design
Rationale
Provide structural
support
Create mechanotransduction-
responsive environments
Failure
Prevention
Mechanical reinforcementDynamic adaptation through
feedback loops
Culture and Conditioning Approaches
AspectTraditional StrategyMechanotransduction-Informed Strategy
Bioreactor
Design
Static or simple dynamic cultureMulti-compartment
mechanotransduction platforms
Mechanical StimulationGeneric cyclic loadingPathway-specific conditioning (YAP/TAZ, TRPV4, TGF-β)
Monitoring
Parameters
Cell growth, basic functionReal-time mechanotransduction pathway activity
Maturation GoalsTissue-like structureFunctional homeostatic mechanisms
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

Tugade, C.; Ramis, J. A Unified Map of Airway Interactions: Secretome and Mechanotransduction Loops from Development to Disease. Adv. Respir. Med. 2025, 93, 51. https://doi.org/10.3390/arm93060051

AMA Style

Tugade C, Ramis J. A Unified Map of Airway Interactions: Secretome and Mechanotransduction Loops from Development to Disease. Advances in Respiratory Medicine. 2025; 93(6):51. https://doi.org/10.3390/arm93060051

Chicago/Turabian Style

Tugade, Crizaldy, and Jopeth Ramis. 2025. "A Unified Map of Airway Interactions: Secretome and Mechanotransduction Loops from Development to Disease" Advances in Respiratory Medicine 93, no. 6: 51. https://doi.org/10.3390/arm93060051

APA Style

Tugade, C., & Ramis, J. (2025). A Unified Map of Airway Interactions: Secretome and Mechanotransduction Loops from Development to Disease. Advances in Respiratory Medicine, 93(6), 51. https://doi.org/10.3390/arm93060051

Article Metrics

Back to TopTop